WO2022120179A1 - Multi-tumor gene signatures and uses thereof - Google Patents

Multi-tumor gene signatures and uses thereof Download PDF

Info

Publication number
WO2022120179A1
WO2022120179A1 PCT/US2021/061823 US2021061823W WO2022120179A1 WO 2022120179 A1 WO2022120179 A1 WO 2022120179A1 US 2021061823 W US2021061823 W US 2021061823W WO 2022120179 A1 WO2022120179 A1 WO 2022120179A1
Authority
WO
WIPO (PCT)
Prior art keywords
pan
tumor
real number
aspects
pharmaceutical composition
Prior art date
Application number
PCT/US2021/061823
Other languages
French (fr)
Inventor
Peter M. SZABO
Scott Adams ELY
Keyur Hemantkumar Desai
Robin EDWARDS
Lan Zhang
Saumya PANT
Zhenhao Qi
Neeraj ADYA
Original Assignee
Bristol-Myers Squibb Company
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 Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Publication of WO2022120179A1 publication Critical patent/WO2022120179A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • the present disclosure provides a method for treating a subject afflicted with a tumor using an immunotherapy.
  • anti-cancer agents can vary in their effectiveness based on the unique patient characteristics. Accordingly, there is a need for targeted therapeutic strategies that identify patients who are more likely to respond to a particular anti-cancer agent and, thus, improve the clinical outcome for patients diagnosed with cancer.
  • the moderate pan-tumor gene signature score is characterized by a score of from
  • the subject identified as having a high gene signature score is to be administered an 1-0 therapy.
  • the subject identified as having a low gene signature score is to be administered an anticancer agent other than an 1-0 therapy.
  • the subject identified as having a moderate gene signature score is to be administered an 1-0 therapy in combination with an additional anticancer agent.
  • the gene panel further comprises one or more housekeeping genes.
  • the gene panel further comprises one or more housekeeping genes selected from the group consisting of ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof.
  • the gene panel further comprises one or more control genes.
  • the gene panel further comprises one or more control genes selected from the group consisting of ANTI, ANT2, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4, PCL- 5, PCL-6, PCL-7, PCL-8, PCL-9, POS1, POS2, POS3, POS4, and any combination thereof.
  • the 1-0 therapy comprises an immune checkpoint inhibitor.
  • the 1-0 therapy comprises an anti-PDl/PD-L2 antagonist.
  • the 1-0 therapy comprises an antibody that specifically binds a protein selected from PD-1, PD-L1, LAG-3, TIGIT, TIM3, NKG2a, CSF1R, 0X40, ICOS, MICA, MICB,CD137, KIR, TGFp, IL-10, IL-8, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, or any combination thereof.
  • the 1-0 comprises an anti-PD-1 antibody.
  • the 1-0 therapy comprises an anti -PD -LI antibody.
  • the tumor sample is not subjected to CD8 immunohi stochemi stry .
  • Some aspects of the present disclosure are directed to a method of identifying a patient in need of an 1-0 therapy, (a) obtaining a tumor sample from the patient; (b) analyzing the expression level of one or more genes in a gene panel selected from CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSF18, and any combination thereof; and (c) determining a pan-tumor gene signature score according to any method disclosed herein.
  • the method further comprises isolating mRNA from the tumor sample prior to analyzing the expression level of the one or more genes.
  • Some aspects of the present disclosure are directed to a method of treating a subject in need thereof, comprising: (a) obtaining a tumor sample from the subject; (b) isolating mRNA from the tumor sample; (c) analyzing the expression level of one or more genes in a gene panel selected from CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSF18, and any combination thereof; (d) determining a pan-tumor gene signature score according to any method disclosed herein; and (e) administering to the subject an 1-0 therapy
  • the expression level of the one or more genes in the gene panel is analyzed by measuring an mRNA level of the one or more genes in the gene panel in the tumor sample. In some aspects, the expression level is measured using a nuclease protection assay. In some aspects, the expression level is measured using next-generation sequencing. In some aspects, the expression level is measured using reverse transcriptase polymerase chain reaction (RT-PCR).
  • RT-PCR reverse transcriptase polymerase chain reaction
  • FIGs. 1A-1C are schematic representations, illustrating the methods used to develop a multi-tumor inflammatory signature based on GEP.
  • FIG. ID is a table listing the weighing coefficient for each gene in the CD8 signature gene panel, disclosed herein.
  • FIG. IE is a representative image of immunohistical (IHC) staining of a tumor sample to detect CD8 localization.
  • FIG. IF shows the comparison of the CD8 signature score and the CD8 IHC score (as determined by examining IHC staining of tumor samples).
  • MEL melanoma
  • SCCHN squamous cell carcinoma of the head and neck; + and - indicate the range of CD8 positivity, based on scoring of each sample in the dataset by digital pathology.
  • FIG. 2 is a scatter plot, showing CD8 expression by IHC (1) and CD8 signature score (2) across tumor types.
  • CD8 score represents the CD8 IHC or CD8 signature scores.
  • Each box represents the 25%-75% interquartile range (IQR) and the line is the median value. Whiskers represent values 1.5 x the upper and lower limits of the IQR.
  • IQR interquartile range
  • CRC colorectal cancer
  • GC gastric cancer
  • HCC hepatocellular carcinoma
  • MEL melanoma
  • NSCLC non-small cell lung cancer
  • OVC ovarian cancer
  • PANC pancreatic cancer
  • PC prostate cancer
  • RCC renal cell carcinoma
  • SCCHN squamous cell carcinoma of the head and neck
  • SCLC small cell lung cancer
  • UC urothelial carcinoma.
  • FIGs. 3A-3L are scatter plots, illustrating the correlation of CD8 IHC and CD8 signature scores across tumor types.
  • MEL melanoma
  • SCCHN squamous cell carcinoma of the head and neck
  • UC urothelial carcinoma
  • CRC colorectal cancer
  • GC gastric cancer
  • HCC hepatocellular carcinoma
  • NSCLC non-small cell lung cancer
  • OVC ovarian cancer
  • PANC pancreatic cancer
  • PC prostate cancer
  • FIG. 3J renal cell carcinoma
  • SCLC small cell lung cancer
  • FIGs. 4A-4L are graphical representations, showing the prevalence of inflammation by CD8 IHC and CD8 signature scores across tumor types.
  • CD8 score represents the CD8 IHC or CD8 signature scores.
  • UC urothelial carcinoma
  • CRC colorectal cancer
  • GC gastric cancer
  • HCC hepatocellular carcinoma
  • MEL melanoma
  • NSCLC non-small cell lung cancer
  • OVC ovarian cancer
  • PANC pancreatic cancer
  • FIG. 4H PC
  • PC prostate cancer
  • RCC renal cell carcinoma
  • SCCHN squamous cell carcinoma of the head and neck
  • SCLC small cell lung cancer
  • FIG. 5 is a scatter plot, illustrating the precision of the CD8 signature, and showing that the precision improves with increasing score across multiple tumor types. Precision of CD8 signature scores was assessed using 8 samples with 15 replicates per tumor type.
  • CRC colorectal cancer
  • GC gastric cancer
  • HCC hepatocellular carcinoma
  • MEL melanoma
  • NSCLC non-small cell lung cancer
  • OVC ovarian cancer
  • PANC pancreatic cancer
  • PC prostate cancer
  • RCC renal cell carcinoma
  • SCCHN squamous cell carcinoma of the head and neck
  • SCLC small cell lung cancer
  • UC urothelial carcinoma.
  • FIG. 6Q is a chart summarizing the correlation with RNA-seq (Pearson's r) for each of the genes of FIGs. 6A-6P as compared to each of the GEP panels (IO 360, Oncomine IRRA, TIP, and IOv2), as indicated.
  • FIGs. 7A-7C are scatter plots showing the correlation of CD8 signature scores derived using the IO 360 (FIG. 7A), the Oncomine IRRA (FIG.
  • FIGs. 8A-8C are scatter plots showing the correlation between gene expression signature score and CD8 immunohistochemistry (H4C) score for the CD8 signature (TIP; FIG. 8A), TCA signature (FIG. 8B), and TIS signature (IO 360; FIG. 8C).
  • FIGs. 8D-8F are scatter plots showing the correlation between gene expression signature score and PDL-1 expression on tumor cells (%) for the CD8 signature (TIP; FIG. 8D), TCA signature (FIG. 8E), and TIS signature (IO 360; FIG. 8F). Pearson's r values are indicated on each graph (FIGs. 8A-8F).
  • FIGs. 8A-8F Pearson's r values are indicated on each graph (FIGs. 8A-8F).
  • 8G-8I are box plots showing the distribution of gene signature scores relative to PD-L1 status (negative or positive) for the CD8 signature (TIP; FIG. 8G), TCA signature (FIG. 8H), and TIS signature (IO 360; FIG. 81).
  • FIGs. 9A-9X are scatter plots showing correlations between CD8 IHC (FIGs. 9A- 9L) or CD8 signature scores (FIGs. 9M-9X) and PD-L1 expression on tumor cells by IHC. Data are shown for melanoma (FIGs. 9A and 9M), SSCHN (FIGs. 9B and 9N), NSCLC (FIGs. 9C and 90), pancreatic cancer (FIGs. 9D and 9P), gastric cancer (FIGs. 9E and 9Q), RCC (FIGs. 9F and 9R), colorectal cancer (FIGs. 9G and 9S), urothelial cancer (FIGs.
  • FIG. 10 is a scatter plot showing the correlation between CD8 signature scores and CD8 IHC in UC samples.
  • FIGs. 11A-11B are Kaplan-Meier curves showing the association of CD8 signature scores with PFS (FIG. 11 A) and OS (FIG. 11B) with nivolumab in patients with UC, grouped by CD8 signature score tertile, defined as low ( ⁇ 7.9), medium (7.9-12.6), or high (> 12.6).
  • OS overall survival
  • PFS progression-free survival
  • UC urothelial carcinoma.
  • Certain aspects of the present disclosure are directed to methods of identifying a human subject suitable for an immune-oncology (1-0) therapy, e.g., an anti-PD-l/PD-Ll antagonist therapy.
  • the present disclosure is directed to a method of identifying a human subject suitable for an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist therapy, comprising measuring expression of CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9 (Galectin-9) , NKG7, TM 1X1173 (STING1), and TNFSF18 (GITRL).
  • a gene panel comprising the identified genes of the present disclosure and the gene signature are useful to identify a subject suitable for and/or responsive to an 1-0 therapy, especially in predicting an inflammatory phenotype in the tumor microenvironment (TME) across multiple tumor types. Therefore, in some aspects, the gene panel and its use can replace the inconvenient and burdensome CD8+ immunohistochemistry.
  • administering refers to the physical introduction of a composition comprising a therapeutic agent to a subject, using any of the various methods and delivery systems known to those skilled in the art.
  • Preferred routes of administration for the immunotherapy include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion.
  • parenteral administration means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal injection and infusion, as well as in vivo electroporation.
  • Other non-parenteral routes include an oral, topical, epidermal or mucosal route of administration, for example, intranasally, vaginally, rectally, sublingually or topically.
  • Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
  • An "adverse event” as used herein is any unfavorable and generally unintended or undesirable sign (including an abnormal laboratory finding), symptom, or disease associated with the use of a medical treatment.
  • an adverse event can be associated with activation of the immune system or expansion of immune system cells (e.g., T cells) in response to a treatment.
  • a medical treatment can have one or more associated AEs and each AE can have the same or different level of severity.
  • Reference to methods capable of "altering adverse events” means a treatment regime that decreases the incidence and/or severity of one or more AEs associated with the use of a different treatment regime.
  • an "antibody” shall include, without limitation, a glycoprotein immunoglobulin which binds specifically to an antigen and comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds, or an antigen-binding portion thereof.
  • Each H chain comprises a heavy chain variable region (abbreviated herein as Vzz) and a heavy chain constant region.
  • the heavy chain constant region comprises three constant domains, Cm, Cm and Cm.
  • Each light chain comprises a light chain variable region (abbreviated herein as Vz) and a light chain constant region.
  • the light chain constant region is comprises one constant domain, CL.
  • Vzz and Vz regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • Each Vzz and Vz comprises three CDRs and four FRs, arranged from amino-terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies can mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.
  • anti-PD-1 antibody includes a full antibody having two heavy chains and two light chains that specifically binds to PD-1 and antigen-binding portions of the full antibody.
  • Non limiting examples of the antigen-binding portions are shown elsewhere herein.
  • An immunoglobulin can derive from any of the commonly known isotypes, including but not limited to IgA, secretory IgA, IgG and IgM.
  • IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4.
  • Immunotype refers to the antibody class or subclass (e.g., IgM or IgGl) that is encoded by the heavy chain constant region genes.
  • antibody includes, by way of example, both naturally occurring and non-naturally occurring antibodies; monoclonal and polyclonal antibodies; chimeric and humanized antibodies; human or nonhuman antibodies; wholly synthetic antibodies; and single chain antibodies.
  • a nonhuman antibody can be humanized by recombinant methods to reduce its immunogenicity in man.
  • an antibody also includes an antigen-binding fragment or an antigenbinding portion of any of the aforementioned immunoglobulins, and includes a monovalent and a divalent fragment or portion, and a single chain antibody.
  • an "isolated antibody” refers to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that binds specifically to PD-1 is substantially free of antibodies that bind specifically to antigens other than PD-1).
  • An isolated antibody that binds specifically to PD-1 may, however, have cross-reactivity to other antigens, such as PD-1 molecules from different species.
  • an isolated antibody can be substantially free of other cellular material and/or chemicals.
  • mAb refers to a non-naturally occurring preparation of antibody molecules of single molecular composition, i.e., antibody molecules whose primary sequences are essentially identical, and which exhibits a single binding specificity and affinity for a particular epitope.
  • a monoclonal antibody is an example of an isolated antibody.
  • Monoclonal antibodies can be produced by hybridoma, recombinant, transgenic or other techniques known to those skilled in the art.
  • a “human antibody” refers to an antibody having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences.
  • the human antibodies of the disclosure can include amino acid residues not encoded by human germline immunoglobulin sequences e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo).
  • the term “human antibody,” as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences.
  • a “humanized antibody” refers to an antibody in which some, most or all of the amino acids outside the CDRs of a non-human antibody are replaced with corresponding amino acids derived from human immunoglobulins. In one aspect of a humanized form of an antibody, some, most or all of the amino acids outside the CDRs have been replaced with amino acids from human immunoglobulins, whereas some, most or all amino acids within one or more CDRs are unchanged. Small additions, deletions, insertions, substitutions or modifications of amino acids are permissible as long as they do not abrogate the ability of the antibody to bind to a particular antigen.
  • a "humanized antibody” retains an antigenic specificity similar to that of the original antibody.
  • a "chimeric antibody” refers to an antibody in which the variable regions are derived from one species and the constant regions are derived from another species, such as an antibody in which the variable regions are derived from a mouse antibody and the constant regions are derived from a human antibody.
  • an "anti-antigen antibody” refers to an antibody that binds specifically to the antigen.
  • an anti-PD-1 antibody binds specifically to PD-1
  • an anti-PD-Ll antibody binds specifically to PD-L1
  • an anti-CTLA-4 antibody binds specifically to CTLA-4.
  • an "antigen-binding portion" of an antibody refers to one or more fragments of an antibody that retain the ability to bind specifically to the antigen bound by the whole antibody. It has been shown that the antigenbinding function of an antibody can be performed by fragments of a full-length antibody.
  • binding fragments encompassed within the term "antigen-binding portion" of an antibody include (i) a Fab fragment (fragment from papain cleavage) or a similar monovalent fragment consisting of the VL, VH, LC and CHI domains; (ii) a F(ab')2 fragment (fragment from pepsin cleavage) or a similar bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341 :544-546), which consists of a VH domain; (vi) an isolated complementarity determining region (CDR)
  • the two domains of the Fv fragment, VL and VH are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883).
  • single chain Fv single chain Fv
  • Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody.
  • Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.
  • Antibodies useful in the methods and compositions described herein include, but are not limited to, antibodies and antigen-binding portions thereof that specifically bind a protein selected from the group consisting of Inducible T cell Co-Stimulator (ICOS), CD137 (4-1BB), CD 134 (0X40), NKG2A, CD27, CD96, Glucocorticoid-Induced TNFR-Related protein (GITR), and Herpes Virus Entry Mediator (HVEM), Programmed Death- 1 (PD-1), Programmed Death Ligand- 1 (PD-L1), Cytotoxic T-Lymphocyte Antigen-4 (CTLA-4), B and T Lymphocyte Attenuator (BTLA), T cell Immunoglobulin and Mucin domain-3 (TIM-3), Lymphocyte Activation Gene-3 (LAG-3), adenosine A2a receptor (A2aR), Killer cell Lectin-like Receptor G1 (KLRG-1), Natural Killer Cell Receptor 2B4 (
  • a "cancer” refers a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth divide and grow results in the formation of malignant tumors that invade neighboring tissues and can also metastasize to distant parts of the body through the lymphatic system or bloodstream.
  • immunotherapy refers to the treatment of a subject afflicted with, or at risk of contracting or suffering a recurrence of, a disease by a method comprising inducing, enhancing, suppressing or otherwise modifying an immune response.
  • Treatment or “therapy” of a subject refers to any type of intervention or process performed on, or the administration of an active agent to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting, slowing down or preventing the onset, progression, development, severity or recurrence of a symptom, complication or condition, or biochemical indicia associated with a disease.
  • PD-1 Programmed Death-1
  • PD-1 refers to an immunoinhibitory receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo, and binds to two ligands, PD-L1 and PD-L2.
  • the term "PD-1” as used herein includes human PD-1 (hPD-1), variants, isoforms, and species homologs of hPD-1, and analogs having at least one common epitope with hPD-1. The complete hPD-1 sequence can be found under GenBank Accession No. U64863.
  • P-L1 Programmed Death Ligand-1
  • PD-L1 is one of two cell surface glycoprotein ligands for PD-1 (the other being PD-L2) that downregulate T cell activation and cytokine secretion upon binding to PD-1.
  • the term "PD-L1" as used herein includes human PD-L1 (hPD- Ll), variants, isoforms, and species homologs of hPD-Ll, and analogs having at least one common epitope with hPD-Ll.
  • the complete hPD-Ll sequence can be found under GenBank Accession No. Q9NZQ7.
  • the human PD-L1 protein is encoded by the human CD274 gene (NCBI Gene lD: 29126).
  • a PD-1 or PD-L1 "inhibitor” refers to any molecule capable of blocking, reducing, or otherwise limiting the interaction between PD-1 and PD-L1 and/or the activity of PD-1 and/or PD-L1.
  • the inhibitor is an antibody or an antigenbinding fragment of an antibody.
  • the inhibitor comprises a small molecule.
  • T-Cell surface glycoprotein CD8 alpha chain or "CD8A” as used herein refers to an integral membrane glycoprotein that is involved in the immune response and that serves multiple functions in responses against both external and internal offenses.
  • CD8a functions primarily as a co-receptor for MHC class I molecule / peptide complex.
  • CD8A interacts simultaneously with the T-cell receptor (TCR) and the MHC class I proteins presented by antigen presenting cells (APCs). In turn, CD8a recruits the Src kinase LCK to the vicinity of the TCR- CD3 complex.
  • LCK then initiates different intracellular signaling pathways by phosphorylating various substrates ultimately leading to lymphokine production, motility, adhesion and activation of cytotoxic T-lymphocytes (CTLs).
  • CTLs cytotoxic T-lymphocytes
  • This mechanism enables CTLs to recognize and eliminate infected cells and tumor cells.
  • NK-cells the presence of CD8A homodimers at the cell surface provides a survival mechanism allowing conjugation and lysis of multiple target cells. CD8A homodimer molecules also promote the survival and differentiation of activated lymphocytes into memory CD8 T-cells.
  • the complete CD8a amino acid sequence can be found under UniProtKB identification number P01732.
  • the human CD8a protein is encoded by the human CD8a gene (NCBI Gene ID: 925).
  • Lymphocyte Activation Gene-3 refers to a type I transmembrane protein that is expressed on the cell surface of activated CD4+ and CD8+ T cells and subsets of NK and dendritic cells.
  • LAG-3 protein is closely related to CD4, which is a co-receptor for T helper cell activation. Both molecules have four extracellular Ig-like domains and require binding to their ligand, major histocompatibility complex (MHC) class II, for their functional activity.
  • MHC major histocompatibility complex
  • LAG-3 can also be found as a soluble protein, which does not bind to MHC class II. LAG-3 also plays an important role in promoting regulatory T cell (Treg) activity and in negatively regulating T cell activation and proliferation. Both natural and induced Treg express increased LAG-3, which is required for their maximal suppressive function.
  • the complete human LAG-3 amino acid sequence can be found under UniProtKB identification number Pl 8627.
  • the human LAG-3 protein is encoded by the human LAG3 gene (NCBI Gene ID: 3902).
  • Signal transducer and activator of transcription 1-alpha/beta refers to a signal transducer and transcription activator that mediates cellular responses to interferons (IFNs), cytokine KITLG/SCF, and other cytokines and other growth factors.
  • IFNs interferons
  • cytokine KITLG/SCF cytokine KITLG/SCF
  • other cytokines and other growth factors cytokines and other growth factors.
  • signaling via protein kinases leads to activation of Jak kinases (TYK2 and JAK1) and to tyrosine phosphorylation of STAT1 and STAT2.
  • ISGF3 The phosphorylated STATs dimerize and associate with ISGF3G/IRF-9 to form a complex termed ISGF3 transcription factor, which enters the nucleus.
  • ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of IFN- stimulated genes (ISG), which drive the cell in an antiviral state.
  • IFN- stimulated response element IFN- stimulated genes
  • STAT1 is tyrosine- and serine-phosphorylated.
  • IFN- gamma-activated factor migrates into the nucleus and binds to the IFN gamma activated sequence (GAS) to drive the expression of the target genes, inducing a cellular antiviral state.
  • GAF IFN- gamma activated sequence
  • STAT1 becomes activated in response to KITLG/SCF and KIT signaling. STAT1 may also mediate cellular responses to activated FGFR1, FGFR2, FGFR3, and FGFR4.
  • the complete human STAT1 amino acid sequence can be found under UniProtKB identification number P42224.
  • the human STAT1 protein is encoded by the human STAT1 gene (NCBI Gene ID: 6772).
  • CTLA-4 Cytotoxic T-Lymphocyte Antigen-4
  • CTLA-4 refers to an immunoinhibitory receptor belonging to the CD28 family.
  • CTLA-4 is expressed exclusively on T cells in vivo, and binds to two ligands, CD80 and CD86 (also called B7-1 and B7-2, respectively).
  • CD80 and CD86 also called B7-1 and B7-2, respectively.
  • CTLA-4" as used herein includes human CTLA-4 (hCTLA-4), variants, isoforms, and species homologs of hCTLA-4, and analogs having at least one common epitope with hCTLA-4.
  • the complete hCTLA-4 sequence can be found under GenBank Accession No. AAB59385.
  • a “subject” includes any human or nonhuman animal.
  • nonhuman animal includes, but is not limited to, vertebrates such as nonhuman primates, sheep, dogs, and rodents such as mice, rats and guinea pigs.
  • the subject is a human.
  • the terms, "subject” and “patient” are used interchangeably herein.
  • a “therapeutically effective amount” or “therapeutically effective dosage” of a drug or therapeutic agent is any amount of the drug that, when used alone or in combination with another therapeutic agent, protects a subject against the onset of a disease or promotes disease regression evidenced by a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction.
  • the ability of a therapeutic agent to promote disease regression can be evaluated using a variety of methods known to the skilled practitioner, such as in human subjects during clinical trials, in animal model systems predictive of efficacy in humans, or by assaying the activity of the agent in in vitro assays.
  • an “anti-cancer agent” promotes cancer regression in a subject.
  • a therapeutically effective amount of the drug promotes cancer regression to the point of eliminating the cancer.
  • “Promoting cancer regression” means that administering an effective amount of the drug, alone or in combination with an anti -neoplastic agent, results in a reduction in tumor growth or size, necrosis of the tumor, a decrease in severity of at least one disease symptom, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction.
  • the terms “effective” and “effectiveness” with regard to a treatment includes both pharmacological effectiveness and physiological safety.
  • Pharmacological effectiveness refers to the ability of the drug to promote cancer regression in the patient.
  • Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (adverse effects) resulting from administration of the drug.
  • an "immuno-oncology" therapy or an “I-O” therapy refers to a therapy that comprises utilizing an immune response to target and treat a tumor in a subject.
  • an 1-0 therapy is a type of anti-cancer therapy.
  • 1-0 therapy comprises administering an antibody or an antigen-binding fragment thereof to a subject.
  • an 1-0 therapy comprises administering to a subject an immune cell, e.g., a T cell, e.g., a modified T cell, e.g., a T cell modified to express a chimeric antigen receptor or an particular T cell receptor.
  • the 1-0 therapy comprises administering a therapeutic vaccine to a subject. In some aspects, the 1-0 therapy comprises administering a cytokine or a chemokine to a subject. In some aspects, the 1-0 therapy comprises administering an interleukin to a subject. In some aspects, the 1-0 therapy comprises administering an interferon to a subject. In some aspects, the 1-0 therapy comprises administering a colony stimulating factor to a subject.
  • a therapeutically effective amount of an anti-cancer agent preferably inhibits cell growth or tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects.
  • tumor regression can be observed and continue for a period of at least about 20 days, more preferably at least about 40 days, or even more preferably at least about 60 days. Notwithstanding these ultimate measurements of therapeutic effectiveness, evaluation of immunotherapeutic drugs must also make allowance for immune-related response patterns.
  • an "immune response” is as understood in the art, and generally refers to a biological response within a vertebrate against foreign agents or abnormal, e.g., cancerous cells, which response protects the organism against these agents and diseases caused by them.
  • An immune response is mediated by the action of one or more cells of the immune system (for example, a T lymphocyte, B lymphocyte, natural killer (NK) cell, macrophage, eosinophil, mast cell, dendritic cell or neutrophil) and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines, and complement) that results in selective targeting, binding to, damage to, destruction of, and/or elimination from the vertebrate's body of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues.
  • a T lymphocyte, B lymphocyte, natural killer (NK) cell for example, a T lymphocyte, B lymphocyte, natural killer (NK) cell, macrophage, eosinophil, mast cell, dendritic cell or neutrophil
  • soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines, and complement) that results
  • An immune reaction includes, e.g., activation or inhibition of a T cell, e.g., an effector T cell, a Th cell, a CD4 + cell, a CD8 + T cell, or a Treg cell, or activation or inhibition of any other cell of the immune system, e.g., NK cell.
  • an "immune-related response pattern” refers to a clinical response pattern often observed in cancer patients treated with immunotherapeutic agents that produce antitumor effects by inducing cancer-specific immune responses or by modifying native immune processes.
  • This response pattern is characterized by a beneficial therapeutic effect that follows an initial increase in tumor burden or the appearance of new lesions, which in the evaluation of traditional chemotherapeutic agents would be classified as disease progression and would be synonymous with drug failure. Accordingly, proper evaluation of immunotherapeutic agents can require longterm monitoring of the effects of these agents on the target disease.
  • treat refers to any type of intervention or process performed on, or administering an active agent to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting, or slowing down or preventing the progression, development, severity or recurrence of a symptom, complication, condition or biochemical indicia associated with a disease or enhancing overall survival.
  • Treatment can be of a subject having a disease or a subject who does not have a disease (e.g., for prophylaxis).
  • an effective dose or “effective dosage” is defined as an amount sufficient to achieve or at least partially achieve a desired effect.
  • a “therapeutically effective amount” or “therapeutically effective dosage” of a drug or therapeutic agent is any amount of the drug that, when used alone or in combination with another therapeutic agent, promotes disease regression evidenced by a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, an increase in overall survival (the length of time from either the date of diagnosis or the start of treatment for a disease, such as cancer, that patients diagnosed with the disease are still alive), or a prevention of impairment or disability due to the disease affliction.
  • a therapeutically effective amount or dosage of a drug includes a "prophylactically effective amount” or a “prophylactically effective dosage”, which is any amount of the drug that, when administered alone or in combination with another therapeutic agent to a subject at risk of developing a disease or of suffering a recurrence of disease, inhibits the development or recurrence of the disease.
  • a therapeutic agent to promote disease regression or inhibit the development or recurrence of the disease can be evaluated using a variety of methods known to the skilled practitioner, such as in human subjects during clinical trials, in animal model systems predictive of efficacy in humans, or by assaying the activity of the agent in in vitro assays.
  • an anti-cancer agent is a drug that promotes cancer regression in a subject.
  • a therapeutically effective amount of the drug promotes cancer regression to the point of eliminating the cancer.
  • "Promoting cancer regression” means that administering an effective amount of the drug, alone or in combination with an antineoplastic agent, results in a reduction in tumor growth or size, necrosis of the tumor, a decrease in severity of at least one disease symptom, an increase in frequency and duration of disease symptom-free periods, an increase in overall survival, a prevention of impairment or disability due to the disease affliction, or otherwise amelioration of disease symptoms in the patient.
  • the terms "effective” and “effectiveness” with regard to a treatment includes both pharmacological effectiveness and physiological safety.
  • Pharmacological effectiveness refers to the ability of the drug to promote cancer regression in the patient.
  • Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (adverse effects) resulting from administration of the drug.
  • a therapeutically effective amount or dosage of the drug inhibits cell growth or tumor growth by at least about 20%, by at least about 40%, by at least about 60%, or by at least about 80% relative to untreated subjects.
  • a therapeutically effective amount or dosage of the drug completely inhibits cell growth or tumor growth, i.e., inhibits cell growth or tumor growth by 100%.
  • the ability of a compound to inhibit tumor growth can be evaluated using an assay described herein. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit cell growth, such inhibition can be measured in vitro by assays known to the skilled practitioner.
  • tumor regression can be observed and continue for a period of at least about 20 days, at least about 40 days, or at least about 60 days.
  • a "cancer gene panel,” “hereditary cancer panel,” “comprehensive cancer panel,” or “multigene cancer panel,” as used herein, refers to a method of sequencing a subset of targeted cancer genes, including coding regions, introns, promoters, and/or mRNA transcripts.
  • the CGP comprises sequencing at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, or at least about 50 targeted cancer genes.
  • microsatellite instability refers to a change that occurs in the DNA of certain cells (such as tumor cells) in which the number of repeats of microsatellites (short, repeated sequences of DNA) is different than the number of repeats that was in the DNA when it was inherited.
  • MSI can be high microsatellite instability (MSI-H) or low microsatellite instability (MSI-L).
  • MSI-H microsatellite instability
  • MSI-L low microsatellite instability
  • Microsatellites are short tandem DNA repeat sequences of 1-6 bases. These are prone to DNA replication errors, which are repaired by mismatch repair (MMR). Hence microsatellites are good indicators of genome instability, especially deficient mismatch repair (dMMR).
  • MSI is usually diagnosed by screening 5 microsatellite markers (BAT-25, BAT-26, NR21, NR24, and NR27).
  • MSI-H represents the presence of at least 2 unstable markers among 5 microsatellite markers analyzed (or >30% of the markers if a larger panel is used).
  • MSI-L means instability of 1 MSI marker (or 10%-30% of markers in larger panels).
  • MSS means the absence of an unstable microsatellite marker.
  • the term "biological sample” as used herein refers to biological material isolated from a subject.
  • the biological sample can contain any biological material suitable for determining target gene expression, for example, by sequencing nucleic acids in the tumor (or circulating tumor cells) and identifying a genomic alteration in the sequenced nucleic acids.
  • the biological sample can be any suitable biological tissue or fluid such as, for example, tumor tissue, blood, blood plasma, and serum.
  • the sample is a tumor sample.
  • the tumor sample can be obtained from a tumor tissue biopsy, e.g., a formalin-fixed, paraffin- embedded (FFPE) tumor tissue or a fresh-frozen tumor tissue or the like.
  • the biological sample is a liquid biopsy that, in some aspects, comprises one or more of blood, serum, plasma, circulating tumor cells, exoRNA, ctDNA, and cfDNA.
  • a tumor sample refers to a biological sample that comprises tumor tissue.
  • a tumor sample is a tumor biopsy.
  • a tumor sample comprises tumor cells and one or more non-tumor cell present in the tumor microenvironment (TME).
  • TME tumor microenvironment
  • the TME is made up of at least two regions.
  • the tumor "parenchyma” is a region of the TME that includes predominantly tumor cells, e.g., the part (or parts) of the TME that includes the bulk of the tumor cells.
  • the tumor parenchyma does not necessarily consist of only tumor cells, rather other cells such as stromal cells and/or lymphocytes can also be present in the parenchyma.
  • the "stromal" region of the TME includes the adjacent non-tumor cells.
  • the tumor sample comprises all or part of the tumor parenchyma and one or more cells of the stroma.
  • the tumor sample is obtained from the parenchyma.
  • the tumor sample is obtained from the stroma.
  • the tumor sample is obtained from the parenchyma and the stroma.
  • the terms "about” or “comprising essentially of' refer to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, “about” or “comprising essentially of' can mean within 1 or more than 1 standard deviation per the practice in the art. Alternatively, “about” or “comprising essentially of' can mean a range of up to 10%. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an order of magnitude or up to 5-fold of a value. When particular values or compositions are provided in the application and claims, unless otherwise stated, the meaning of "about” or “comprising essentially of' should be assumed to be within an acceptable error range for that particular value or composition.
  • any concentration range, percentage range, ratio range or real number range is to be understood to include the value of any real number within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an real number), unless otherwise indicated.
  • Inflammation in the TME can be an indicator of potential responsiveness to an 1-0 therapy.
  • contemporary methods for measuring inflammation in a tumor require the laborious process of immunohistochemistry to detect and analyze CD8 expression in a tumor biopsy. It was surprisingly found that the expression pattern of a relatively small number of genes (in some aspects, at least about 16 genes) correlates with inflammation in a tumor microenvironment.
  • the methods described herein can replace the need for timeconsuming IHC.
  • the measuring is conducted in vitro.
  • the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18.
  • pan tumor gene signature score [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p)
  • pan-tumor gene panel for use in a method of identifying a subject suitable for an 1-0 therapy comprising calculating a pan- tumor gene signature score for a tumor sample obtained from the subject, wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the pantumor gene panel in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying Formula 1.
  • the subject is identified as being suitable for an 1-0 therapy if the subject has a high pan-tumor gene signature score.
  • pan-tumor gene panel for use in a method of stratifying a subject in need of an 1-0 therapy, comprising determining a pantumor gene signature score for a tumor sample obtained from the subject,
  • pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA- DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying Formula 1 to the measured expression values.
  • the subject is stratified as either having a high pan-tumor gene signature score, a moderate pan-tumor gene signature score, or a low pan-tumor gene signature score.
  • (a) is any real number between 0.01 and 0.05. In some aspects, (b) is any real number between 0.10 and 0.15. In some aspects, (c) is any real number between 0.01 and 0.05. In some aspects, (d) is any real number between -0.10 and -0.15. In some aspects, (e) is any real number between 0.03 and 0.09. In some aspects, (f) is any real number between 0.35 and 0.45. In some aspects, (g) is any real number between 0.07 and 0.13. In some aspects, (h) is any real number between 0.02 and 0.08. In some aspects, (i) is any real number between - 0.01 and -0.05. In some aspects, (j) is any real number between -0.0001 and -0.0010.
  • (k) is any real number between 0.01 and 0.06. In some aspects, (1) is any real number between 0.02 and 0.08. In some aspects, (m) is any real number between -0.008 and -0.040. In some aspects, (n) is any real number between 0.01 and 0.050. In some aspects, (o) is any real number between -0.03 and -0.080. In some aspects, (p) is any real number between -0.005 and - 0.010.
  • (a) is any real number between 0.01 and 0.05; (b) is any real number between 0.10 and 0.15; (c) is any real number between 0.01 and 0.05; (d) is any real number between -0.10 and -0.15; (e) is any real number between 0.03 and 0.09; (f) is any real number between 0.35 and 0.45; (g) is any real number between 0.07 and 0.13; (h) is any real number between 0.02 and 0.08; (i) is any real number between -0.01 and -0.05; (j) is any real number between -0.0001 and -0.0010; (k) is any real number between 0.01 and 0.06; (1) is any real number between 0.02 and 0.08; (m) is any real number between -0.008 and -0.040; (n) is any real number between 0.01 and 0.050; (o) is any real number between -0.03 and -0.080; and (p) is any real number between -0.005 and -0.010.
  • (a) is about 0.01, about 0.015, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.026, about 0.027, about 0.028, about 0.029, about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, about 0.04, about 0.041, about 0.042, about 0.043, about 0.044, about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, or about 0.05.
  • (a) is about 0.02 to about 0.05, about 0.02 to about 0.04, about 0.03 to about 0.05, or about 0.03 to about 0.04. In some aspects, (a) is about 0.03. In some aspects, (a) is about 0.031. In some aspects, (a) is about 0.032. In some aspects, (a) is about 0.033. In some aspects, (a) is about 0.034. In some aspects, (a) is about 0.035. In some aspects, (a) is about 0.036. In some aspects, (a) is about 0.037. In some aspects, (a) is about 0.038. In some aspects, (a) is about 0.039. In some aspects, (a) is about 0.04. In some aspects, (a) is about 0.041. In some aspects, (a) is about 0.042. In some aspects, (a) is about 0.043. In some aspects, (a) is about 0.044. In some aspects,
  • (a) is about 0.045. In some aspects, (a) is about 0.046. In some aspects, (a) is about 0.047. In some aspects, (a) is about 0.048. In some aspects, (a) is about 0.049. In some aspects, (a) is about 0.05. In certain aspects, (a) is about 0.03447.
  • (b) is about 0.1, about 0.105, about 0.11, about 0.115, about 0.12, about 0.125, about 0.13, about 0.131, about 0.132, about 0.133, about 0.134, about 0.135, about 0.136, about 0.137, about 0.138, about 0.139, about 0.14, about 0.141, about 0.142, about 0.143, about 0.144, about 0.145, about 0.146, about 0.147, about 0.148, about 0.149, or about 0.15.
  • (b) is about 0.11 to about 0.15, about 0.11 to about 0.14, about 0.12 to about 0.15, about 0.12 to about 0.14, or about 0.13 to about 0.14. In some aspects, (b) is about 0.131.
  • (b) is about 0.132. In some aspects, (b) is about 0.133. In some aspects, (b) is about 0.134. In some aspects, (b) is about 0.135. In some aspects, (b) is about 0.136. In some aspects,
  • (b) is about 0.137. In some aspects, (b) is about 0.138. In some aspects, (b) is about 0.139. In some aspects, (b) is about 0.14. In certain aspects, (b) is about 0.13825.
  • (c) is about 0.01, about 0.015, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.026, about 0.027, about 0.028, about 0.029, about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, about 0.04, about 0.045, or about 0.05.
  • (c) is about 0.01 to about 0.05, about 0.01 to about 0.04, about 0.01 to about 0.03, about 0.02 to about 0.05, about 0.02 to about 0.04, or about 0.02 to about 0.03.
  • (c) is about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.026, about 0.027, about 0.028, about 0.029, about 0.03. In certain aspects (c) is about 0.02756.
  • (d) is about -0.05, about -0.1, about -0.105, about -0.11, about - 0.115, about -0.12, about -0.125, about -0.126, about -0.127, about -0.128, about -0.129, about - 0.13, about -0.131, about -0.132, about -0.133, about -0.134, about -0.135, about -0.14, about - 0.145, or about -0.15.
  • (d) is about -0.10 to about -0.15, about -0.10 to about - 0.14, about -0.10 to about -0.13, about -0.11 to about -0.15, about -0.11 to about -0.14, about - 0.11 to about -0.13, about -0.12 to about -0.15, about -0.12 to about -0.14, about -0.12 to about - 0.13, about -0.13 to about -0.15, about -0.13 to about -0.14, about -0.13 to about -0.139, about - 0.13 to about -0.138, about -0.13 to about -0.137, about -0.13 to about -0.136, about -0.13 to about -0.135, about -0.13 to about -0.134, about -0.13 to about -0.133, about -0.13 to about - 0.132, or about -0.13 to about -0.131. In certain aspects, (d) is about -0.13089.
  • (e) is about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.061, about 0.062, about 0.063, about 0.064, about 0.065, about 0.066, about 0.067, about 0.068, about 0.069, about 0.07, about 0.075, about 0.08, about 0.085, or about 0.09.
  • (e) is about 0.04 to about 0.09, about 0.04 to about 0.08, about 0.05 to about 0.09, about 0.05 to about 0.08, about 0.06 to about 0.09, about 0.06 to about 0.08, about 0.06 to about 0.07, about 0.06 to about 0.069, about 0.06 to about 0.068, about 0.06 to about 0.067, about 0.06 to about 0.066, about 0.06 to about 0.065, about 0.06 to about 0.064, about 0.061 to about 0.069, about 0.061 to about 0.068, about 0.061 to about 0.067, about 0.061 to about 0.066, about 0.061 to about 0.065, about 0.061 to about 0.064, about 0.062 to about 0.069, about 0.062 to about 0.068, about 0.062 to about 0.067, about 0.062 to about 0.066, about 0.062 to about 0.065, about 0.062 to about 0.064, about 0.063 to about 0.069, about 0.063 to about 0.069,
  • (f) is about 0.35, about 0.36, about 0.37, about 0.38, about 0.385, about 0.386, about 0.387, about 0.388, about 0.389, about 0.39, about 0.391, about 0.392, about 0.393, about 0.394, about 0.395, about 0.396, about 0.397, about 0.398, about 0.399, about 0.40, about 0.41, about 0.42, about 0.43, about 0.44, or about 0.45.
  • (f) is about 0.37 to about 0.42, about 0.37 to about 0.41, about 0.37 to about 0.40, about 0.38 to about 0.42, about 0.38 to about 0.41, about 0.38 to about 0.40, about 0.39 to about 0.42, about 0.39 to about 0.41, about 0.39 to about 0.40, about 0.39 to about 0.399, about 0.39 to about 0.398, about 0.39 to about 0.397, about 0.39 to about 0.396, about 0.39 to about 0.395, about 0.39 to about 0.394, about 0.39 to about 0.393, about 0.39 to about 0.392, or about 0.391 to about 0.392. In certain aspects, (f) is about 0.39185.
  • (g) is about 0.07, about 0.075, about 0.08, about 0.085, about 0.09, about 0.095, about 0.10, about 0.105, about 0.106, about 0.107, about 0.108, about 0.109, about 0.11, about 0.112, about 0.113, about 0.114, about 0.115, about 0.12, about 0.125, or about 0.13.
  • (g) is about 0.08 to about 0.13, about 0.08 to about 0.12, about 0.08 to about 0.11, about 0.09 to about 0.13, about 0.09 to about 0.12, about 0.09 to about 0.11, about 0.09 to about 0.13, about 0.10 to about 0.13, about 0.10 to about 0.12, about 0.10 to about 0.11, about 0.105 to about 0.115, about 0.105 to about 0.115. In some aspects, (g) is about 0.105, about 0.106, about 0.107, about 0.108, about 0.109, about 0.11, about 0.112, about 0.113, about 0.114, or about 0.115. In certain aspects, (g) is about 0.10976.
  • (h) is about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.08.
  • (h) is about 0.02 to about 0.07, about 0.02 to about 0.06, about 0.02 to about 0.05, about 0.03 to about 0.07, about 0.03 to about 0.06, about 0.03 to about 0.05, about 0.04 to about 0.07, about 0.04 to about 0.06, about 0.04 to about 0.055, about 0.04 to about 0.05, about 0.045 to about 0.055, about 0.045 to about 0.054, about 0.045 to about 0.053, about 0.045 to about 0.052, about 0.045 to about 0.051, about 0.045 to about 0.050, about 0.045 to about 0.049, about 0.046 to about 0.054, about 0.047 to about 0.053, about 0.048 to about 0.052, about 0.049 to about 0.051, or about 0.049 to about 0.050.
  • (h) is about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, or about 0.055. In certain aspects, (h) is about 0.04913.
  • (i) is about -0.01, about -0.015, about -0.02, about -0.021, about - 0.022, about -0.023, about -0.024, about -0.025, about -0.026, about -0.027, about -0.028, about - 0.029, about -0.03, about -0.035, about -0.04, about -0.045, or about -0.05.
  • (i) is about -0.02 to about -0.05, about -0.02 to about -0.04, about -0.02 to about -0.03, about -0.02 to about -0.029, about -0.02 to about -0.028, about -0.02 to about -0.027, about -0.02 to about - 0.026, about -0.021 to about -0.029, about -0.021 to about -0.028, about -0.021 to about -0.027, about -0.021 to about -0.026, about -0.022 to about -0.029, about -0.022 to about -0.028, about - 0.022 to about -0.027, about -0.022 to about -0.026, about -0.023 to about -0.029, about -0.023 to about -0.028, about -0.023 to about -0.027, about -0.023 to about -0.026, about -0.024 to about - 0.029, about -0.024 to about -0.0
  • (i) is about -0.02, about -0.021, about -0.022, about - 0.023, about -0.024, about -0.025, about -0.026, about -0.027, about -0.028, or about -0.029. In certain aspects, (i) is about -0.02537.
  • (j) is about -0.0005, about -0.00055, about -0.0006, about - 0.00065, about -0.0007, about -0.00075, about -0.000756, about -0.000757, about -0.000758, about -0.000759, about -0.0008, about -0.00081, about -0.00082, about -0.00083, about -0.00084, about -0.00085, about -0.0009, about -0.00095, or about -0.0010. In some aspects, (j) is about -0.0005, about -0.00055, about -0.0006, about - 0.00065, about -0.0007, about -0.00075, about -0.000756, about -0.000757, about -0.000758, about -0.000759, about -0.0008, about -0.00081, about -0.00082, about -0.00083, about -0.00084, about -0.00085, about -0.0009, about -0.00095, or about -0.0010. In some aspects, (j) is about -
  • (k) is about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, about 0.04, about 0.045, about 0.05, about 0.055, or about 0.06.
  • (k) is about 0.01 to about 0.06, about 0.01 to about 0.05, about 0.01 to about 0.04, about 0.02 to about 0.06, about 0.02 to about 0.05, about 0.02 to about 0.04, about 0.03 to about 0.06, about 0.03 to about 0.05, about 0.03 to about 0.04, about 0.031 to about 0.039, about 0.032 to about 0.038, about 0.033 to about 0.037, about 0.034 to about 0.037, about 0.035 to about 0.037, or about 0.036 to about 0.037.
  • (k) is about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, or about 0.04. In certain aspects, (k) is about 0.03654.
  • (1) is about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.8.
  • (1) is about 0.02 to about 0.07, about 0.02 to about 0.06, about 0.02 to about 0.05, about 0.03 to about 0.07, about 0.03 to about 0.06, about 0.03 to about 0.05, about 0.04 to about 0.07, about 0.04 to about 0.06, about 0.04 to about 0.05, about 0.045 to about 0.055, about 0.046 to about 0.054, about 0.047 to about 0.053, about 0.047 to about 0.052, about 0.047 to about 0.051, about 0.047 to about 0.050, about 0.047 to about 0.049, or about 0.047 to about 0.048.
  • (1) is about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, or about 0.055. In certain aspects, (1) is about 0.04797.
  • (m) is about -0.008, about -0.010, about -0.015, about -0.016, about -0.017, about -0.018, about -0.019, about -0.020, about -0.021, about -0.022, about -0.023, about -0.024, about -0.025, about -0.030, about -0.035, or about -0.0400.
  • (m) is about -0.05 to about -0.03, about -0.05 to about -0.025, about -0.01 to about -0.03, about -0.01 to about -0.025, about -0.01 to about -0.02, about -0.01 to about -0.019, about -0.01 to about -0.018, about -0.01 to about -0.017, about -0.015 to about -0.02, about -0.015 to about -0.019, about - 0.015 to about -0.018, about -0.015 to about -0.017, about -0.016 to about -0.02, about -0.016 to about -0.019, about -0.016 to about -0.018, about -0.016 to about -0.017, about -0.017 to about -0.02, about -0.017 to about -0.019, or about -0.017 to about -0.018.
  • (m) is about -0.015, about -0.016, about -0.017, about -0.018, about -0.019, about -0.020, about -0.021, about -0.022, about -0.023, about -0.024, or about -0.025. In certain aspects, (m) is about -0.01751.
  • (n) is about 0.01, about 0.015, about 0.016, about 0.017, about 0.018, about 0.019, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.050.
  • (n) is about 0.01 to about 0.05, about 0.01 to about 0.04, about 0.01 to about 0.03, about 0.015 to about 0.05, about 0.015 to about 0.04, about 0.015 to about 0.03, about 0.02 to about 0.05, about 0.02 to about 0.04, about 0.02 to about 0.03, about 0.02 to about 0.025, about 0.021 to about 0.024, or about 0.022 to about 0.023.
  • (n) is about 0.015, about 0.016, about 0.017, about 0.018, about 0.019, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, or about 0.025.
  • (n) is about 0.02254.
  • (o) is about -0.03, about -0.035, about -0.04, about -0.045, about -0.05, about -0.051, about -0.052, about -0.053, about -0.054, about -0.055, about -0.056, about - 0.057, about -0.058, about -0.059, about -0.06, about -0.065, about -0.07, about -0.075, or about - 0.08.
  • (o) is about -0.03 to about -0.08, about -0.03 to about -0.07, about -0.03 to about -0.06, about -0.04 to about -0.08, about -0.04 to about -0.07, about -0.04 to about -0.06, about -0.05 to about -0.08, about -0.05 to about -0.07, about -0.05 to about -0.06, about -0.051 to about -0.059, about -0.052 to about -0.058, about -0.053 to about -0.059, about - 0.053 to about -0.058, about -0.053 to about -0.057, about -0.053 to about -0.056, about -0.053 to about -0.055, or about -0.053 to about -0.054.
  • (o) is about -0.045, about -0.05, about -0.051, about -0.052, about -0.053, about -0.054, about -0.055, about -0.056, about -0.057, about -0.058, about -0.059, or about -0.06. In certain aspects, (o) is about -0.05317.
  • (p) is about -0.005, about -0.0055, about -0.006, about -0.0065, about -0.0066, about -0.0067, about -0.0068, about -0.0069, about -0.007, about -0.0071, about - 0.0072, about -0.0073, about -0.0074, about -0.0075, about -0.008, about -0.0085, about -0.009, about -0.0095, or about -0.010.
  • (p) is about 0.005 to about 0.010, about 0.005 to about 0.009, about 0.005 to about 0.008, about 0.006 to about 0.010, about 0.006 to about 0.009, about 0.006 to about 0.008, about 0.007 to about 0.010, about 0.007 to about 0.009, about 0.007 to about 0.008, about 0.0065 to about 0.0075, about 0.0066 to about 0.0075, about 0.0067 to about 0.0075, about 0.0068 to about 0.0074, about 0.0069 to about 0.0073, about 0.0070 to about 0.0072, about 0.0071 to about 0.0072, or about 0.0070 to about 0.0071.
  • (p) is about -0.0065, about -0.0066, about -0.0067, about -0.0068, about -0.0069, about -0.007, about - 0.0071, about -0.0072, about -0.0073, about -0.0074, or about -0.0075. In certain aspects, (p) is about -0.00710.
  • (a) is about 0.03447; (b) is about 0.13825; (c) is about 0.02756; (d) is about -0.13089; (e) is about 0.06368; (f) is about 0.39185; (g) is about 0.10976; (h) is about 0.04913; (i) is about -0.02537; (j) is about -0.00081; (k) is about 0.03654; (1) is about 0.04797; (m) is about -0.01751; (n) is about 0.02254; (o) is about -0.05317; and (p) is about -0.00710.
  • the methods disclosed herein comprise administering an 1-0 therapy to a subject identified as having a high pan-tumor gene signature score.
  • a high pan-tumor gene signature score is characterized by a score of greater than about 12, greater than about 12.1, greater than about 12.2, greater than about 12.3, greater than about 12.4, greater than about 12.5, greater than about 12.6, greater than about 12.7, greater than about 12.8, greater than about 12.9, greater than about 13, greater than about 14, greater than about 15, greater than about 16, greater than about 17, greater than about 18, greater than about 19, greater than about 20.
  • a high pan-tumor gene signature score is characterized by a score of greater than about 12.1.
  • a high pan-tumor gene signature score is characterized by a score of greater than about 12.2. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.3. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.4. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.5. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.6. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.7.
  • a high pan-tumor gene signature score is characterized by a score of greater than about 12.8. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.9. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 13. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 14. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 15. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 16. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 17.
  • a high pan-tumor gene signature score is characterized by a score of greater than about 18. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 19. In some aspects, a high pantumor gene signature score is characterized by a score of greater than about 20.
  • the methods disclosed herein comprise administering an 1-0 therapy in combination with an additional anticancer agent to a subject identified as having a moderate pan-tumor gene signature score.
  • a moderate pan-tumor gene signature score is characterized by a score from about 7.0 to about 13.5, about 7.1 to about 13.4, about 7.2 to about 13.3, about 7.3 to about 13.2, about 7.4 to about 13.1, about 7.5 to about 13.0, about 7.6 to about 12.9, about 7.8 to about 12.8, about 7.9 to about 12.7, or about 7.9 to about 12.6.
  • a moderate pan-tumor gene signature score is characterized by a score from about 7.9 to about 12.6.
  • a moderate pan-tumor gene signature score is characterized by a score of about 8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.5. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 8.6.
  • a moderate pan-tumor gene signature score is characterized by a score of about 8.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.3.
  • a moderate pan-tumor gene signature score is characterized by a score of about 9.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.9. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 10.
  • a moderate pan-tumor gene signature score is characterized by a score of about 10.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.7.
  • a moderate pan-tumor gene signature score is characterized by a score of about 10.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.1. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 11.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.4.
  • a moderate pan-tumor gene signature score is characterized by a score of about 11.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.1.
  • a moderate pan-tumor gene signature score is characterized by a score of about 12.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.4. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 12.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.6.
  • the methods disclosed herein comprise administering an anticancer therapy to a subject identified as having a low pan-tumor gene signature score, wherein the anticancer therapy does not comprise an 1-0 therapy.
  • a low pantumor gene signature score is characterized by a score of less than about 12, less than about 12.1, less than about 11, less than about 10, less than about 9, less than about 8, less than about 7.9, less than about 7.8, less than about 7.7, less than about 7.6, less than about 7.5, less than about
  • a low pan-tumor gene signature score is characterized by a score of less than about 8. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.9. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.8. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.6. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 17.5.
  • a low pantumor gene signature score is characterized by a score of less than about 7.4. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.3. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.2. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.1. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.9.
  • a low pan-tumor gene signature score is characterized by a score of less than about 6.8. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.7. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.6. In some aspects, a low pantumor gene signature score is characterized by a score of less than about 6.5. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.4. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.3.
  • a low pan-tumor gene signature score is characterized by a score of less than about 6.2. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.1. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.
  • the gene panel further comprises one or more additional genes.
  • the gene panel comprises at least 2 to at least about 100 genes.
  • the gene panel comprises at least 2 to at least about 95, at least 2 to at least about 90, at least 2 to at least about 85, at least 2 to at least about 80, at least 2 to at least about 75, at least 2 to at least about 70, at least 2 to at least about 65, at least 2 to at least about 60, at least 2 to at least about 55, at least 2 to at least about 50, at least 2 to at least about 45, at least 2 to at least about 40, at least 2 to at least about 35, at least 2 to at least about 30, at least 2 to at least about 25, at least 2 to at least about 20, at least 2 to at least about 15, at least 2 to at least about 10, at least 2 to at least about 9, at least 2 to at least about 8, at least 2 to at least about 7, at least 2 to at least about 6, at least 2 to at least about 5, or at least 2 to at least about 4 genes.
  • the gene panel comprises at least 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, at least about 10, at least about 11, at least about 12, at least about 13, at least about 14, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, at least about 50, at least about 55, at least about 60, at least about 65, at least about 70, at least about 75, at least about 80, at least about 85, at least about 90, at least about 95, or at least about 100 genes.
  • the gene panel comprises at least 2 genes. In certain aspects, the gene panel comprises at least 3 genes. In certain aspects, the gene panel comprises at least 4 genes. In certain aspects, the gene panel comprises at least 5 genes. In certain aspects, the gene panel comprises at least 6 genes. In certain aspects, the gene panel comprises at least 7 genes. In certain aspects, the gene panel comprises at least 8 genes. In certain aspects, the gene panel comprises at least 9 genes. In certain aspects, the gene panel comprises at least 10 genes. In certain aspects, the gene panel comprises at least 11 genes. In certain aspects, the gene panel comprises at least 12 genes. In certain aspects, the gene panel comprises at least 13 genes. In certain aspects, the gene panel comprises at least 14 genes. In certain aspects, the gene panel comprises at least 15 genes.
  • the gene panel comprises at least 16 genes. In certain aspects, the gene panel comprises at least 17 genes. In certain aspects, the gene panel comprises at least 18 genes. In certain aspects, the gene panel comprises at least 19 genes. In certain aspects, the gene panel comprises at least 20 genes.
  • TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173
  • the gene panel comprises CCL4, CCL5, CD27, CD276, CD3D,
  • the gene panel comprises at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, at least about 10, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, at least about 50, at least about 55, at least about 60, at least about 65, at least about 70, or at least about 75 additional genes.
  • the gene panel comprises less than 95 genes, wherein the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18.
  • the gene panel comprises less than 90 genes, less than about 85 genes, less than about 80 genes, less than about 75 genes, less than about 70 genes, less than about 65 genes, less than about 60 genes, less than about 55 genes, less than about 50 genes, less than about 45 genes, less than about 40 genes, less than about 35 genes, less than about 30 genes, less than about 25 genes, or less than about 20, wherein the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18.
  • the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18.
  • the gene panel comprises 16 to 90 genes, 16 to about 85 genes, 16 to about 70 genes, 16 to about 65 genes, 16 to about 60 genes, 16 to about 55 genes, 16 to about 50 genes, 16 to about 45 genes, 16 to about 40 genes, 16 to about 35 genes, 16 to about 30 genes, 16 to about 25 genes, 16 to about 20 genes, wherein the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (SHNG1), and TNFSF18.
  • the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (SHNG1), and TNFSF18.
  • the one or more additional genes are selected from CCL2, CCL3, CCR2, CCR5, CD274, CD28, CDS, CD73, CD80, CD86, CMKLR1, CSF1R, CTLA-4, CXCL11, CXCR2, CXCR6, EP 4, GITR, GZMA, GZMK, HLA-DMA, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-E, IGOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, 0X40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof.
  • the panel of genes comprises at least one housekeeping gene.
  • a housekeeping gene is a gene that is expressed at a nearly constant level across various cell types. As such, the expression of a house keeping gene can be relative to the number of cells in a sample, acting as a means of normalizing the expression of other variable genes.
  • the panel of genes comprises at least 2 housekeeping genes, at least 3 housekeeping genes, at least housekeeping genes, at least 4 housekeeping genes, at least 5 housekeeping genes, at least 6 housekeeping genes, at least 7 housekeeping genes, at least 8 housekeeping genes, at least 9 housekeeping genes, at least 10 housekeeping genes, at least 11 housekeeping genes, at least 12 housekeeping genes, at least 13 housekeeping genes, at least 14 housekeeping genes, at least 15 housekeeping genes, at least 16 housekeeping genes, at least 17 housekeeping genes, at least 18 housekeeping genes, at least 19 housekeeping genes, at least 20 housekeeping genes, at least 21 housekeeping genes, at least 22 housekeeping genes, at least 23 housekeeping genes, at least 24 housekeeping genes, at least 25 housekeeping genes, at least 26 housekeeping genes, at least housekeeping genes, at least 27 housekeeping genes, at least 28 housekeeping genes, at least 29 housekeeping genes, at least 30 housekeeping genes, at least 35 housekeeping genes, at least 40 housekeeping genes, at least 45 housekeeping genes, at least 50 housekeeping genes, at least 55, at least
  • the panel of genes comprises at least 2 housekeeping genes. In some aspects, the panel of genes comprises at least 3 housekeeping genes. In some aspects, the panel of genes comprises at least 4 housekeeping genes. In some aspects, the panel of genes comprises at least 5 housekeeping genes. In some aspects, the panel of genes comprises at least 6 housekeeping genes. In some aspects, the panel of genes comprises at least 7 housekeeping genes. In some aspects, the panel of genes comprises at least 8 housekeeping genes. In some aspects, the panel of genes comprises at least 9 housekeeping genes. In some aspects, the panel of genes comprises at least 10 housekeeping genes. In some aspects, the panel of genes comprises at least 11 housekeeping genes. In some aspects, the panel of genes comprises at least 12 housekeeping genes. In some aspects, the panel of genes comprises at least 13 housekeeping genes. In some aspects, the panel of genes comprises at least 14 housekeeping genes. In some aspects, the panel of genes comprises at least 15 housekeeping genes.
  • housekeeping genes known in the art can be used in the panel of genes disclosed herein.
  • the housekeeping genes are selected from the group consisting of ACTB, ATP5F1, DDX5, EEF 1G, GAPDH, NCL, OAZ1, PPIA, RPI.38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof
  • the panel of genes comprises at least one control gene.
  • the panel of genes comprises at least 2 control genes, at least 3 control genes, at least control genes, at least 4 control genes, at least 5 control genes, at least 6 control genes, at least 7 control genes, at least 8 control genes, at least 9 control genes, at least 10 control genes, at least 11 control genes, at least 12 control genes, at least 13 control genes, at least 14 control genes, at least 15 control genes, at least 16 control genes, at least 17 control genes, at least 18 control genes, at least 19 control genes, at least 20 control genes, at least 21 control genes, at least 22 control genes, at least 23 control genes, at least 24 control genes, at least 25 control genes, at least 26 control genes, at least control genes, at least 27 control genes, at least 28 control genes, at least 29 control genes, at least 30 control genes, at least 35 control genes, at least 40 control genes, at least 45 control genes, at least 50 control genes, at least 55 control genes, at least 60 control genes, at least 65 control genes, at least 50 control genes, at least 55 control
  • the panel of genes comprises at least 2 control genes. In some aspects, the panel of genes comprises at least 3 control genes. In some aspects, the panel of genes comprises at least 4 control genes. In some aspects, the panel of genes comprises at least 5 control genes. In some aspects, the panel of genes comprises at least 6 control genes. In some aspects, the panel of genes comprises at least 7 control genes. In some aspects, the panel of genes comprises at least 8 control genes. In some aspects, the panel of genes comprises at least 9 control genes. In some aspects, the panel of genes comprises at least 10 control genes. In some aspects, the panel of genes comprises at least 11 control genes. In some aspects, the panel of genes comprises at least 12 control genes. In some aspects, the panel of genes comprises at least 13 control genes.
  • the panel of genes comprises at least 14 control genes. In some aspects, the panel of genes comprises at least 15 control genes. In some aspects, the panel of genes comprises at least 16 control genes. In some aspects, the panel of genes comprises at least 17 control genes. In some aspects, the panel of genes comprises at least 18 control genes. In some aspects, the panel of genes comprises at least 19 control genes. In some aspects, the panel of genes comprises at least 20 control genes. In some aspects, the control genes are selected from the group consisting of ANTI, ANTS, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4, PCL-5, PCL-6, PCL-7, PCL-8, PCL-9, POS1, POS2, POS3, POS4, and any combination thereof.
  • the one or more additional genes are selected from CCL2, CCL3, CCR2, CCR5, CD274, CD28, CDS, CD73, CD80, CD86, CMKLR1, CSF1R, CTLA-4, CXCL11, CXCR2, CXCR6, EP 4, GITR, GZMA, GZMK, HLA-DMA, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAGS, Nectin-2, NKG2D, 0X40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIMS, ANTI, ANT2, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4
  • the one or more genes of the pan-tumor inflammation panel comprises at least one gene involved in inflammation, e.g., at least one inflammatory gene. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene that is a T cell marker. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene that is a macrophage marker. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene that is an HLA gene. In some aspects, the HLA gene is an MHC class I gene. In some aspects, the HLA gene is an MHC class II gene.
  • the one or more genes of the pan-tumor inflammation panel comprises at least one gene is an interferon gene. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene encodes a protein that is part of the interferon gamma pathway. In some aspects, the one or more genes of the pantumor inflammation panel comprises at least one gene encodes a protein that is a checkpoint inhibitor.
  • Gene expression profiling is a measurement of the combined expression level of the one or more genes of the pan-tumor inflammation gene panel disclosed herein.
  • the measurement is made using a sample obtained from a subject.
  • the sample is a tumor sample. Any biological sample comprising one or more tumor cell can be used in the methods disclosed herein.
  • the sample is selected from a tumor biopsy, a blood sample, a serum sample, or any combination thereof.
  • the sample is a tumor biopsy collected from the subject prior to administration of a therapy described herein, e.g., an 1-0 therapy, e.g., an anti-PD-l/PD-Ll agonist.
  • the sample obtained from the subject is a formalin-fixed tumor biopsy.
  • the sample obtained from the subject is a paraffin-embedded tumor biopsy.
  • the sample obtained from the subject is a fresh-frozen tumor biopsy.
  • any method known in the art for measuring the expression of a particular gene or a panel of genes can be used in the methods of the present disclosure.
  • the expression of one or more of the genes in the pan-tumor inflammation gene panel is determined by detecting the presence of mRNA transcribed from the gene, the presence of a protein encoded by the gene, or both.
  • the expression of a gene is determined by measuring the level of gene mRNA in a sample obtained from the subject.
  • the gene expression profile is determined by measuring the level of mRNA transcripts of one or more genes of the pan-tumor inflammation gene panel. Any method known in the art can be used to measure the level of the gene mRNA.
  • the gene mRNA is measured using reverse transcriptase PCR.
  • the gene mRNA is measured using a nuclease protection assay.
  • the gene mRNA is measured using next-generation sequencing (NGS).
  • NGS next-generation sequencing
  • the gene mRNA is measured using RNA in situ hybridization.
  • the expression of a gene is determined by measuring the level of protein encoded by the gene in a sample obtained from the subject.
  • the gene expression profile is determined by measuring the level of protein encoded by one or more genes of the pan-tumor inflammation gene panel in a sample obtained from the subject. Any method known in the art can be used to measure the level of the protein.
  • the gene expression profile is measured using an immunohistochemistry (IHC) assay.
  • the IHC is an automated IHC.
  • the expression of one or more of the genes of the pan-tumor inflammation gene panel is normalized relative to the expression of one or more housekeeping genes.
  • the one or more housekeeping genes are made up of genes that have relatively consistent expression across various tumor types in various subjects.
  • raw gene expression values are normalized following standard gene expression profiling protocols.
  • a gene expression profile can be calculated as the median or average of the log2 -transformed normalized and scaled expression values across all of the target genes in the signature, and presented on a linear scale.
  • profiles have positive or negative values, depending on whether gene expression is up- or down-regulated under a particular condition.
  • increased/decreased expression is characterized by an expression level that is greater/less than the expression of the same gene or genes in a reference sample.
  • the reference sample comprises a non-tumor tissue of the same subject.
  • the reference sample comprises a corresponding non-tumor tissue of the same subject.
  • the reference sample comprises a corresponding tissue in a subject that does not have a tumor.
  • the reference sample comprises more than one tumor tissue sample from more than one other subject, e.g., the increased expression is relative to an average expression level across more than one other tumor samples.
  • the increased/decreased expression is characterized by an expression level that is greater/less than a reference expression level.
  • the reference expression level is an average expression level.
  • the average expression level is determined by measuring the expression of the gene (or the multiple genes) present in the gene panel in tumor samples obtained from a population of subjects, and calculating the average for the population of subjects.
  • each member of the population of subjects is afflicted with the same tumor as the subject being administered the 1-0 therapy, e.g., an anti-PD- 1/PD-L1 antagonist.
  • increased expression of the upregulated genes is characterized by an expression level that is at least about 5%, at least about 10%, at least about 15%, at least about
  • increased expression is characterized by an expression level that is at least about 25% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 30% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 35% higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 40% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 45% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 50% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 55% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 60% higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 65% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 70% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 75% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 80% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 85% higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 90% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 95% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 100% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 125% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 150% higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 175% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 200% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 225% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 250% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 275% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 300% higher than an expression level in a reference sample or than an average expression level.
  • increased expression of the upregulated genes is characterized by an expression level that is at least about 1.25-fold, at least about 1.30-fold, at least about 1.35- fold, at least about 1.40-fold, at least about 1.45-fold, at least about 1.50-fold, at least about 1.55- fold, at least about 1.60-fold, at least about 1.65-fold, at least about 1.70-fold, at least about 1.75- fold, at least about 1.80-fold, at least about 1.85-fold, at least about 1.90-fold, at least about 1.95- fold, at least about 2-fold, at least about 2.25-fold, at least about 2.50-fold, at least about 2.75- fold, at least about 3-fold, at least about 3.25-fold, at least about 3.50-fold, at least about 3.75- fold, or at least about 400-fold higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 1.25-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.30-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.35-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.40-fold higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 1.45- fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.50-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.55-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.60-fold higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 1.65-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.70-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.75-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.80-fold higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 1.85- fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.90-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.95-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2-fold higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 2.25-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2.50-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2.75-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3.25- fold higher than an expression level in a reference sample or than an average expression level.
  • increased expression is characterized by an expression level that is at least about 3.50-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3.75-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 4-fold higher than an expression level in a reference sample or than an average expression level.
  • decreased expression of the down-regulated genes is characterized by an expression level that is less than a reference expression level.
  • decreased expression is characterized by an expression level that is at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 225%, at least about 250%, at least about 275%, or at least about 300% lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 25% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 30% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 35% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 40% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 45% lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 50% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 55% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 60% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 65% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 70% lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 75% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 80% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 85% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 90% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 95% lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 100% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 125% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 150% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 175% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 200% lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 225% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 250% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 275% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 300% lower than an expression level in a reference sample or than an average expression level.
  • decreased expression of the down-regulated genes is characterized by an expression level that is at least about 1.25-fold, at least about 1.30-fold, at least about 1.35-fold, at least about 1.40-fold, at least about 1.45-fold, at least about 1.50-fold, at least about 1.55-fold, at least about 1.60-fold, at least about 1.65-fold, at least about 1.70-fold, at least about 1.75-fold, at least about 1.80-fold, at least about 1.85-fold, at least about 1.90-fold, at least about 1.95-fold, at least about 2-fold, at least about 2.25-fold, at least about 2.50-fold, at least about 2.75-fold, at least about 3-fold, at least about 3.25-fold, at least about 3.50-fold, at least about 3.75-fold, or at least about 400-fold lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 1.25-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.30-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.35-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.40- fold lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 1.45-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.50-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.55-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.60-fold lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 1.65-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.70-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.75-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.80- fold lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 1.85-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.90-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.95-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2-fold lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 2.25-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2.50-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2.75-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3.25- fold lower than an expression level in a reference sample or than an average expression level.
  • decreased expression is characterized by an expression level that is at least about 3.50-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3.75-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 4-fold lower than an expression level in a reference sample or than an average expression level.
  • Certain aspects of the present disclosure are directed to methods of identifying a subject suitable for a therapy and then administering the therapy to the suitable subject.
  • the methods of identifying a suitable subject described herein can be used in advance of any immuno-oncology (I-O) therapy.
  • the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds a protein selected from PD-1, PD-L1, CTLA-4, LAG-3, TIGIT, TIM3, CSF1R, NKG2a, 0X40, ICOS, CD137, KIR, TGFp, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, MICA, MICB, and any combination thereof.
  • a protein selected from PD-1, PD-L1, CTLA-4, LAG-3, TIGIT, TIM3, CSF1R, NKG2a, 0X40, ICOS, CD137, KIR, TGFp, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR,
  • the method of identifying a patient in need of an 1-0 therapy comprises (a) obtaining a tumor sample from the patient, and (b) analyzing the expression level of CCL4, CCL5, CD27, CD276, CD3D, CDS A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HLA- DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSFJ8, and any combination thereof, according to the methods disclosed herein.
  • the gene panel consists of CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSFJ8.
  • the method further comprises isolating mRNA from the tumor sample prior to analyzing the expression level of the one or more genes.
  • the expression level of the one or more genes in the gene panel is analyzed by measuring an mRNA level of the one or more genes in the gene panel in the tumor sample.
  • the expression level is measured a nuclease protection assay.
  • the expression level is measured using next-generation sequencing.
  • the expression level is measured using reverse transcriptase polymerase chain reaction (RT-PCR).
  • the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigenbinding fragment thereof that specifically binds CTLA-4. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds LAG-3. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds TIGIT.
  • the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds TIM3. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds GITR. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds MICA. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigenbinding fragment thereof that specifically binds MICB. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds CSF1R.
  • the suitable subject is to be administered and/or subsequently administered more than one antibody or antigen-binding fragment thereof disclosed herein. In some aspects, the suitable subject is to be administered and/or subsequently administered at least two antibodies or antigen-binding fragments thereof. In some aspects, the suitable subject is to be administered and/or subsequently administered at least three antibodies or antigen-binding fragments thereof. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1 and an antibody or antigen-binding fragment thereof that specifically binds CTLA-4.
  • the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1 and an antibody or antigen-binding fragment thereof that specifically binds CTLA-4. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1 and an antibody or antigen-binding fragment thereof that specifically binds CSF1R. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1 and an antibody or antigen-binding fragment thereof that specifically binds CSF1R.
  • the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1 and an antibody or antigen-binding fragment thereof that specifically binds LAG-3. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1 and an antibody or antigenbinding fragment thereof that specifically binds LAG-3.
  • the therapy is administered to the suitable subject after the gene expression profile has been measured.
  • the measuring is in vitro.
  • the measuring is in vivo.
  • the therapy is administered at least about 1 day, at least about 2 days, at least about 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 7 days, at least about 8 days, at least about 9 days, at least about 10 days, at least about 11 days, at least about 12 days, at least about 13 days, or at least about 14 days after the gene expression profile has been measured.
  • the particular therapy to be administered and/or subsequently administered to the suitable subject is dependent on the gene expression profile.
  • a first therapy is administered if a subject has a first type of gene expression profile.
  • a second therapy is administered if a subject has a second type of gene expression profile.
  • the first and second therapy are different.
  • the first therapy, the second therapy, or both comprises an 1-0 monotherapy.
  • a subject identified as having a high pan-tumor gene signature score is administered an 1-0 therapy as a first therapy and/or a second therapy.
  • the first therapy, the second therapy, or both comprises a combination therapy comprising (i) an 1-0 therapy and (ii) one or more additional anti-cancer agent.
  • a subject identified as having a moderate pantumor gene signature score is administered a combination therapy comprising (i) an 1-0 therapy and (ii) one or more additional anti-cancer agent as a first therapy and/or a second therapy.
  • the first therapy comprises an 1-0 monotherapy and the second therapy comprises a combination therapy comprising (i) an 1-0 therapy and (ii) one or more additional anti-cancer agent.
  • the present disclosure is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist, for use in a method of identifying a human subject suitable for the 1-0 therapy, e.g., anti-PD-l/PD-Ll antagonist, wherein the method comprises measuring expression of CCL4, CCL5, CD27, CD276, CD3D, CDS A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HIA-DRA, HLA-DRB1, I.GAI.S9, NKG7, TMEM173 (STING1), and TNFSF18, according to the methods disclosed herein.
  • the subject is identified as being suitable for an 1-0 therapy when the tumor sample exhibits a high pan-tumor gene signature score. In some aspects, the subject is identified as being suitable for a combination therapy comprising (i) an 1-0 therapy and (ii) an additional anticancer agent, when the tumor sample exhibits a moderate pan-tumor gene signature score. In some aspects, the subject is identified as being suitable for an anticancer therapy when the tumor sample exhibits a low pan-tumor gene signature score, wherein the anticancer therapy does not comprise an 1-0 therapy.
  • the present disclosure provides, in some aspects, a pharmaceutical composition comprising an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist, for use in a method of treating a human subject afflicted with a tumor, wherein a tumor sample obtained from the subject exhibits a high pan-tumor gene signature score.
  • an 1-0 therapy e.g., an anti-PD-l/PD-Ll antagonist
  • the disclosure provides a method of identifying a human subject suitable for an 1-0 therapy, e.g. an anti-PD-l/PD-Ll antagonist therapy, comprising measuring expression of CCI.4, CCL5, CD27, CD276, CD3D, CDS A, CXCI.10, CXCI.9, CXCR1, HLA- DMB, HIA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18, according to the methods disclosed herein.
  • the measuring is in vitro. In other aspects, the measuring is in vivo.
  • the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an 1-0 therapy described herein. In some aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an anti-PD-l/PD-Ll antagonist. In some aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an anti-PD-1 antibody. In some aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an anti-PD-Ll antibody.
  • the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an 1-0 therapy described herein and (ii) one or more additional anti-cancer agents.
  • the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an anti- PD-1/PD-L1 antagonist and (ii) one or more additional anti-cancer agents.
  • the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an anti-PD-1 antibody and (ii) one or more additional anti-cancer agents.
  • the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an anti-PD-Ll antibody and (ii) one or more additional anti-cancer agents.
  • the present disclosure are directed to methods of treating a suitable subject, as determined according to a method disclosed herein, using an 1-0 therapy.
  • Any 1-0 therapy known in the art can be used in the methods described herein.
  • the 1-0 therapy comprises administering to the suitable subject an antibody or an antigen-binding fragment thereof the specifically binds a protein selected from PD-1, PD-L1, CTLA-4, LAG-3, TIGIT, TIM3, CSF1R, NKG2a, 0X40, ICOS, CD137, KIR, TGFp, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, MICA, MICB, and any combination thereof.
  • the subject is administered a single 1-0 therapy, i.e., a monotherapy.
  • the subject is administered an anti-PD-1 antibody monotherapy.
  • the subject is administered a combination therapy comprising a first 1-0 therapy and a second 1-0 therapy.
  • the subject is administered a combination therapy comprising administering a first 1-0 therapy and an additional anti-cancer agent.
  • the additional anti-cancer agent comprises a second 1-0 therapy, a chemotherapy, a standard of care therapy, or any combination thereof.
  • the subject is administered a combination therapy comprising an anti-PD-1 antibody and a second anti-cancer agent. In certain aspects, the subject is administered a combination therapy comprising an anti-PD-1 antibody and an anti-CTLA-4 antibody. In certain aspects, the subject is administered a combination therapy comprising an anti-PD-1 antibody and an anti-CSFIR antibody.
  • the subject is administered a combination therapy comprising an anti-PD-Ll antibody and a second anti-cancer agent.
  • the subject is administered a combination therapy comprising an anti-PD-Ll antibody and an anti-CTLA-4 antibody.
  • the subject is administered a combination therapy comprising an anti-PD-Ll antibody and an anti-CSFIR antibody.
  • Anti-PD-1 antibodies that are known in the art can be used in the presently described compositions and methods.
  • Various human monoclonal antibodies that bind specifically to PD-1 with high affinity have been disclosed in U.S. Patent No. 8,008,449.
  • Anti-PD-1 antibodies usable in the present disclosure include monoclonal antibodies that bind specifically to human PD-1 and exhibit at least one, in some aspects, at least five, of the preceding characteristics.
  • Other anti-PD-1 monoclonal antibodies have been described in, for example, U.S. Patent Nos. 6,808,710, 7,488,802, 8,168,757 and 8,354,509, US Publication No. 2016/0272708, and PCT Publication Nos. WO 2012/145493, WO 2008/156712, WO 2015/112900, WO 2012/145493, WO 2015/112800, WO 2014/206107, WO 2015/35606, WO 2015/085847, WO
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab (also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538), pembrolizumab (Merck; also known as KEYTRUDA®, lambrolizumab, and MK-3475; see WO2008/156712), PDR001 (Novartis; see WO 2015/112900), MEDI-0680 (AstraZeneca; also known as AMP-514; see WO 2012/145493), cemiplimab (Regeneron; also known as REGN- 2810; see WO 2015/112800), JS001 (TAIZHOU JUNSHI PHARMA; also known as toripalimab; see Si-Yang Liu et al., J.
  • nivolumab also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538
  • BGB-A317 Beigene; also known as Tislelizumab; see WO 2015/35606 and US 2015/0079109
  • INCSHR1210 Jiangsu Hengrui Medicine; also known as SHR-1210; see WO 2015/085847; Si-Yang Liu et al., J. Hematol. Oncol. 70: 136 (2017)
  • TSR-042 Tesaro Biopharmaceutical; also known as ANB011; see WO2014/179664)
  • GLS-010 Wangi/Harbin Gloria Pharmaceuticals; also known as WBP3055; see Si-Yang Liu et al., J. Hematol.
  • the anti-PD-1 antibody is nivolumab.
  • Nivolumab is a fully human IgG4 (S228P) PD-1 immune checkpoint inhibitor antibody that selectively prevents interaction with PD-1 ligands (PD-L1 and PD-L2), thereby blocking the down-regulation of antitumor T-cell functions (U.S. Patent No. 8,008,449; Wang et al., 2014 Cancer Immunol Res. 2(9/846-56).
  • the anti-PD-1 antibody is pembrolizumab.
  • Pembrolizumab is a humanized monoclonal IgG4 (S228P) antibody directed against human cell surface receptor PD- 1 (programmed death-1 or programmed cell death-1). Pembrolizumab is described, for example, in U.S. Patent Nos. 8,354,509 and 8,900,587.
  • Anti-PD-1 antibodies usable in the disclosed compositions and methods also include isolated antibodies that bind specifically to human PD-1 and cross-compete for binding to human PD-1 with any anti-PD-1 antibody disclosed herein, e.g., nivolumab (see, e.g., U.S. Patent No.
  • the anti-PD-1 antibody binds the same epitope as any of the anti-PD-1 antibodies described herein, e.g, nivolumab.
  • the ability of antibodies to cross-compete for binding to an antigen indicates that these monoclonal antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other cross-competing antibodies to that particular epitope region.
  • These cross-competing antibodies are expected to have functional properties very similar those of the reference antibody, e.g, nivolumab, by virtue of their binding to the same epitope region of PD-1.
  • Cross-competing antibodies can be readily identified based on their ability to cross-compete with nivolumab in standard PD-1 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • the antibodies that cross-compete for binding to human PD-1 with, or bind to the same epitope region of human PD-1 antibody, nivolumab are monoclonal antibodies.
  • these cross-competing antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies.
  • Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
  • Anti-PD-1 antibodies usable in the compositions and methods of the disclosed disclosure also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • Anti-PD-1 antibodies suitable for use in the disclosed compositions and methods are antibodies that bind to PD-1 with high specificity and affinity, block the binding of PD-L1 and or PD-L2, and inhibit the immunosuppressive effect of the PD-1 signaling pathway.
  • an anti-PD-1 "antibody” includes an antigenbinding portion or fragment that binds to the PD-1 receptor and exhibits the functional properties similar to those of whole antibodies in inhibiting ligand binding and up-regulating the immune system.
  • the anti-PD-1 antibody or antigen-binding portion thereof crosscompetes with nivolumab for binding to human PD-1.
  • the anti-PD-1 antibody is administered at a dose ranging from 0.1 mg/kg to 20.0 mg/kg body weight once every 2, 3, 4, 5, 6, 7, or 8 weeks, e.g., 0.1 mg/kg to 10.0 mg/kg body weight once every 2, 3, or 4 weeks. In other aspects, the anti-PD-1 antibody is administered at a dose of about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, or 10 mg/kg body weight once every 2 weeks.
  • the anti-PD-1 antibody is administered at a dose of about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, or 10 mg/kg body weight once every 3 weeks.
  • the anti-PD-1 antibody is administered at a dose of about 5 mg/kg body weight about once every 3 weeks.
  • the anti-PD-1 antibody e.g., nivolumab
  • the anti-PD-1 antibody e.g., pembrolizumab
  • the anti-PD-1 antibody useful for the present disclosure can be administered as a flat dose.
  • the anti-PD-1 antibody is administered at a flat dose of from about 100 to about 1000 mg, from about 100 mg to about 900 mg, from about 100 mg to about 800 mg, from about 100 mg to about 700 mg, from about 100 mg to about 600 mg, from about 100 mg to about 500 mg, from about 200 mg to about 1000 mg, from about 200 mg to about 900 mg, from about 200 mg to about 800 mg, from about 200 mg to about 700 mg, from about 200 mg to about 600 mg, from about 200 mg to about 500 mg, from about 200 mg to about 480 mg, or from about 240 mg to about 480 mg,
  • the anti-PD-1 antibody is administered as a flat dose of at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about
  • the anti-PD-1 antibody is administered as a flat dose of about 200 mg to about 800 mg, about 200 mg to about 700 mg, about 200 mg to about 600 mg, about 200 mg to about 500 mg, at a dosing interval of about 1, 2, 3, or 4 weeks.
  • the anti-PD-1 antibody is administered as a flat dose of about 200 mg at about once every 3 weeks. In other aspects, the anti-PD-1 antibody is administered as a flat dose of about 200 mg at about once every 2 weeks. In other aspects, the anti-PD-1 antibody is administered as a flat dose of about 240 mg at about once every 2 weeks. In certain aspects, the anti-PD-1 antibody is administered as a flat dose of about 480 mg at about once every 4 weeks. [0176] In some aspects, nivolumab is administered at a flat dose of about 240 mg once about every 2 weeks. In some aspects, nivolumab is administered at a flat dose of about 240 mg once about every 3 weeks. In some aspects, nivolumab is administered at a flat dose of about 360 mg once about every 3 weeks. In some aspects, nivolumab is administered at a flat dose of about 480 mg once about every 4 weeks.
  • pembrolizumab is administered at a flat dose of about 200 mg once about every 2 weeks. In some aspects, pembrolizumab is administered at a flat dose of about 200 mg once about every 3 weeks. In some aspects, pembrolizumab is administered at a flat dose of about 400 mg once about every 4 weeks.
  • the PD-1 inhibitor is a small molecule. In some aspects, the PD-1 inhibitor comprises a millamolecule. In some aspects, the PD-1 inhibitor comprises a macrocyclic peptide. In certain aspects, the PD-1 inhibitor comprises BMS-986189. In some aspects, the PD-1 inhibitor comprises an inhibitor disclosed in International Publication No. WO2014/151634, which is incorporated by reference herein in its entirety. In some aspects, the PD-1 inhibitor comprises INCMGA00012 (Insight Pharmaceuticals). In some aspects, the PD-1 inhibitor comprises a combination of an anti -PD-1 antibody disclosed herein and a PD-1 small molecule inhibitor.
  • an anti-PD-Ll antibody is substituted for the anti-PD-1 antibody in any of the methods disclosed herein.
  • Anti-PD-Ll antibodies that are known in the art can be used in the compositions and methods of the present disclosure.
  • Examples of anti-PD-Ll antibodies useful in the compositions and methods of the present disclosure include the antibodies disclosed in US Patent No. 9,580,507.
  • 9,580,507 have been demonstrated to exhibit one or more of the following characteristics: (a) bind to human PD-L1 with a KD of 1 x 10' 7 M or less, as determined by surface plasmon resonance using a Biacore biosensor system; (b) increase T-cell proliferation in a Mixed Lymphocyte Reaction (MLR) assay; (c) increase interferon-y production in an MLR assay; (d) increase IL-2 secretion in an MLR assay; (e) stimulate antibody responses; and (f) reverse the effect of T regulatory cells on T cell effector cells and/or dendritic cells.
  • MLR Mixed Lymphocyte Reaction
  • Anti- PD-Ll antibodies usable in the present disclosure include monoclonal antibodies that bind specifically to human PD-L1 and exhibit at least one, in some aspects, at least five, of the preceding characteristics.
  • the anti-PD-Ll antibody is selected from the group consisting of BMS-936559 (also known as 12A4, MDX-1105; see, e.g, U.S. Patent No. 7,943,743 and WO 2013/173223), atezolizumab (Roche; also known as TECENTRIQ®; MPDL3280A, RG7446; see US 8,217,149; see, also, Herbst et al.
  • the PD-L1 antibody is atezolizumab (TECENTRIQ®).
  • Atezolizumab is a fully humanized IgGl monoclonal anti-PD-Ll antibody.
  • the PD-L1 antibody is durvalumab (IMFINZITM).
  • Durvalumab is a human IgGl kappa monoclonal anti-PD-Ll antibody.
  • the PD-L1 antibody is avelumab (BAVENCIO®).
  • Avelumab is a human IgGl lambda monoclonal anti-PD-Ll antibody.
  • Anti-PD-Ll antibodies usable in the disclosed compositions and methods also include isolated antibodies that bind specifically to human PD-L1 and cross-compete for binding to human PD-L1 with any anti-PD-Ll antibody disclosed herein, e.g., atezolizumab, durvalumab, and/or avelumab.
  • the anti-PD-Ll antibody binds the same epitope as any of the anti-PD-Ll antibodies described herein, e.g., atezolizumab, durvalumab, and/or avelumab.
  • antibodies to cross-compete for binding to an antigen indicates that these antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other crosscompeting antibodies to that particular epitope region.
  • These cross-competing antibodies are expected to have functional properties very similar those of the reference antibody, e.g., atezolizumab and/or avelumab, by virtue of their binding to the same epitope region of PD-L1.
  • Cross-competing antibodies can be readily identified based on their ability to cross-compete with atezolizumab and/or avelumab in standard PD-L1 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • the antibodies that cross-compete for binding to human PD-L1 with, or bind to the same epitope region of human PD-L1 antibody as, atezolizumab, durvalumab, and/or avelumab are monoclonal antibodies.
  • these cross-competing antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies.
  • Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
  • Anti-PD-Ll antibodies usable in the compositions and methods of the disclosed disclosure also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • Anti-PD-Ll antibodies suitable for use in the disclosed compositions and methods are antibodies that bind to PD-L1 with high specificity and affinity, block the binding of PD-1, and inhibit the immunosuppressive effect of the PD-1 signaling pathway.
  • an anti-PD-Ll "antibody” includes an antigen-binding portion or fragment that binds to PD-L1 and exhibits the functional properties similar to those of whole antibodies in inhibiting receptor binding and up-regulating the immune system.
  • the anti-PD-Ll antibody or antigen-binding portion thereof cross-competes with atezolizumab, durvalumab, and/or avelumab for binding to human PD-L1.
  • the anti-PD-Ll antibody useful for the present disclosure can be any PD-L1 antibody that specifically binds to PD-L1, e.g., antibodies that cross-compete with durvalumab, avelumab, or atezolizumab for binding to human PD-1, e.g., an antibody that binds to the same epitope as durvalumab, avelumab, or atezolizumab.
  • the anti-PD-Ll antibody is durvalumab.
  • the anti-PD-Ll antibody is avelumab.
  • the anti-PD-Ll antibody is atezolizumab.
  • the anti-PD-Ll antibody is administered at a dose ranging from about 0.1 mg/kg to about 20.0 mg/kg body weight, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 11 mg/kg, about 12 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 16 mg/kg, about 17 mg/kg, about 18 mg/kg, about 19 mg/kg, or about 20 mg/kg, about once every 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the anti-PD-Ll antibody is administered at a dose of about 15 mg/kg body weight at about once every 3 weeks. In other aspects, the anti-PD-Ll antibody is administered at a dose of about 10 mg/kg body weight at about once every 2 weeks.
  • the anti-PD-Ll antibody useful for the present disclosure is a flat dose.
  • the anti-PD-Ll antibody is administered as a flat dose of from about 200 mg to about 1600 mg, about 200 mg to about 1500 mg, about 200 mg to about 1400 mg, about 200 mg to about 1300 mg, about 200 mg to about 1200 mg, about 200 mg to about 1100 mg, about 200 mg to about 1000 mg, about 200 mg to about 900 mg, about 200 mg to about 800 mg, about 200 mg to about 700 mg, about 200 mg to about 600 mg, about 700 mg to about 1300 mg, about 800 mg to about 1200 mg, about 700 mg to about 900 mg, or about 1100 mg to about 1300 mg.
  • the anti-PD-Ll antibody is administered as a flat dose of at least about 240 mg, at least about 300 mg, at least about 320 mg, at least about 400 mg, at least about 480 mg, at least about 500 mg, at least about 560 mg, at least about 600 mg, at least about 640 mg, at least about 700 mg, at least 720 mg, at least about 800 mg, at least about 840 mg, at least about 880 mg, at least about 900 mg, at least 960 mg, at least about 1000 mg, at least about 1040 mg, at least about 1100 mg, at least about 1120 mg, at least about 1200 mg, at least about 1280 mg, at least about 1300 mg, at least about 1360 mg, or at least about 1400 mg, at a dosing interval of about 1, 2, 3, or 4 weeks.
  • the anti-PD-Ll antibody is administered as a flat dose of about 1200 mg at about once every 3 weeks. In other aspects, the anti-PD-Ll antibody is administered as a flat dose of about 800 mg at about once every 2 weeks. In other aspects, the anti-PD-Ll antibody is administered as a flat dose of about 840 mg at about once every 2 weeks. [0192] In some aspects, atezolizumab is administered as a flat dose of about 1200 mg once about every 3 weeks. In some aspects, atezolizumab is administered as a flat dose of about 800 mg once about every 2 weeks. In some aspects, atezolizumab is administered as a flat dose of about 840 mg once about every 2 weeks.
  • avelumab is administered as a flat dose of about 800 mg once about every 2 weeks.
  • durvalumab is administered at a dose of about 10 mg/kg once about every 2 weeks. In some aspects, durvalumab is administered as a flat dose of about 800 mg/kg once about every 2 weeks. In some aspects, durvalumab is administered as a flat dose of about 1200 mg/kg once about every 3 weeks.
  • the PD-L1 inhibitor is a small molecule. In some aspects, the PD- L1 inhibitor comprises a millamolecule. In some aspects, the PD-L1 inhibitor comprises a macrocyclic peptide. In certain aspects, the PD-L1 inhibitor comprises BMS-986189.
  • the PD-L1 inhibitor comprises a millamolecule having a formula set forth in formula (II): wherein R x -R 13 are amino acid side chains, R a -R n are hydrogen, methyl, or form a ring with a vicinal R group, and R 14 is -C(O)NHR 15 , wherein R 15 is hydrogen, or a glycine residue optionally substituted with additional glycine residues and/or tails which can improve pharmacokinetic properties.
  • the PD-L1 inhibitor comprises a compound disclosed in International Publication No. WO2014/151634, which is incorporated by reference herein in its entirety.
  • the PD-L1 inhibitor comprises a compound disclosed in International Publication No. WO2016/039749, WO2016/149351, WO2016/077518, W02016/100285, WO2016/100608, WO2016/126646, WO2016/057624, W02017/151830, WO20 17/176608, W02018/085750, WO2018/237153, or WO2019/070643, each of which is incorporated by reference herein in its entirety.
  • the PD-L1 inhibitor comprises a small molecule PD-L1 inhibitor disclosed in International Publication No. WO2015/034820, WO2015/160641, WO2018/044963, WO20 17/066227, W02018/009505, WO2018/183171, WO2018/118848, WO2019/147662, or WO2019/169123, each of which is incorporated by reference herein in its entirety.
  • the PD-L1 inhibitor comprises a combination of an anti-PD-Ll antibody disclosed herein and a PD-L1 small molecule inhibitor disclosed herein.
  • Anti-CTLA-4 antibodies that are known in the art can be used in the compositions and methods of the present disclosure.
  • Anti-CTLA-4 antibodies of the instant disclosure bind to human CTLA-4 so as to disrupt the interaction of CTLA-4 with a human B7 receptor. Because the interaction of CTLA-4 with B7 transduces a signal leading to inactivation of T-cells bearing the CTLA-4 receptor, disruption of the interaction effectively induces, enhances or prolongs the activation of such T cells, thereby inducing, enhancing or prolonging an immune response.
  • 6,984,720 have been demonstrated to exhibit one or more of the following characteristics: (a) binds specifically to human CTLA-4 with a binding affinity reflected by an equilibrium association constant (K a ) of at least about 10 7 M' 1 , or about 10 9 M' 1 , or about 10 10 M' 1 to 10 11 M' 1 or higher, as determined by Biacore analysis; (b) a kinetic association constant (U of at least about 10 3 , about 10 4 , or about 10 5 m' 1 s' 1 ; (c) a kinetic disassociation constant (k ⁇ ) of at least about 10 3 , about 10 4 , or about 10 5 m' 1 s' 1 ; and (d) inhibits the binding of CTLA-4 to B7-1 (CD80) and B7-2 (CD86).
  • Anti-CTLA-4 antibodies useful for the present disclosure include monoclonal antibodies that bind specifically to human CTLA-4 and exhibit at least one, at least two, or at least three of the preceding characteristics.
  • the CTLA-4 antibody is selected from the group consisting of ipilimumab (also known as YERVOY®, MDX-010, 10D1; see U.S. Patent No. 6,984,720), MK- 1308 (Merck), AGEN-1884 (Agenus Inc.; see WO 2016/196237), and tremelimumab (AstraZeneca; also known as ticilimumab, CP-675,206; see WO 2000/037504 and Ribas, Update Cancer Ther. 2(3): 133-39 (2007)).
  • the anti-CTLA-4 antibody is ipilimumab.
  • the CTLA-4 antibody is ipilimumab for use in the compositions and methods disclosed herein.
  • Ipilimumab is a fully human, IgGl monoclonal antibody that blocks the binding of CTLA-4 to its B7 ligands, thereby stimulating T cell activation and improving overall survival (OS) in patients with advanced melanoma.
  • the CTLA-4 antibody is tremelimumab.
  • the CTLA-4 antibody is MK-1308.
  • CTLA-4 antibody is AGEN-1884.
  • Anti-CTLA-4 antibodies usable in the disclosed compositions and methods also include isolated antibodies that bind specifically to human CTLA-4 and cross-compete for binding to human CTLA-4 with any anti-CTLA-4 antibody disclosed herein, e.g., ipilimumab and/or tremelimumab.
  • the anti-CTLA-4 antibody binds the same epitope as any of the anti-CTLA-4 antibodies described herein, e.g., ipilimumab and/or tremelimumab.
  • the ability of antibodies to cross-compete for binding to an antigen indicates that these antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other crosscompeting antibodies to that particular epitope region.
  • cross-competing antibodies are expected to have functional properties very similar those of the reference antibody, e.g., ipilimumab and/or tremelimumab, by virtue of their binding to the same epitope region of CTLA-4.
  • Cross-competing antibodies can be readily identified based on their ability to crosscompete with ipilimumab and/or tremelimumab in standard CTLA-4 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • the antibodies that cross-compete for binding to human CTLA- 4 with, or bind to the same epitope region of human CTLA-4 antibody as, ipilimumab and/or tremelimumab are monoclonal antibodies.
  • these crosscompeting antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies.
  • Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
  • Anti-CTLA-4 antibodies usable in the compositions and methods of the disclosed disclosure also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
  • Anti-CTLA-4 antibodies suitable for use in the disclosed methods or compositions are antibodies that bind to CTLA-4 with high specificity and affinity, block the activity of CTLA-4, and disrupt the interaction of CTLA-4 with a human B7 receptor.
  • an anti-CTLA-4 "antibody” includes an antigenbinding portion or fragment that binds to CTLA-4 and exhibits the functional properties similar to those of whole antibodies in inhibiting the interaction of CTLA-4 with a human B7 receptor and up-regulating the immune system.
  • the anti-CTLA-4 antibody or antigenbinding portion thereof cross-competes with ipilimumab and/or tremelimumab for binding to human CTLA-4.
  • the anti-CTLA-4 antibody or antigen-binding portion thereof is administered at a dose ranging from 0.1 mg/kg to 10.0 mg/kg body weight once every 2, 3, 4, 5, 6, 7, or 8 weeks. In some aspects, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered at a dose of 1 mg/kg or 3 mg/kg body weight once every 3, 4, 5, or 6 weeks. In one aspect, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered at a dose of 3 mg/kg body weight once every 2 weeks. In another aspect, the anti-PD-1 antibody or antigenbinding portion thereof is administered at a dose of 1 mg/kg body weight once every 6 weeks.
  • the anti-CTLA-4 antibody or antigen-binding portion thereof is administered as a flat dose.
  • the anti-CTLA-4 antibody is administered at a flat dose of from about 10 to about 1000 mg, from about 10 mg to about 900 mg, from about 10 mg to about 800 mg, from about 10 mg to about 700 mg, from about 10 mg to about 600 mg, from about 10 mg to about 500 mg, from about 100 mg to about 1000 mg, from about 100 mg to about 900 mg, from about 100 mg to about 800 mg, from about 100 mg to about 700 mg, from about 100 mg to about 100 mg, from about 100 mg to about 500 mg, from about 100 mg to about 480 mg, or from about 240 mg to about 480 mg.
  • the anti-CTLA-4 antibody or antigenbinding portion thereof is administered as a flat dose of at least about 60 mg, at least about 80 mg, at least about 100 mg, at least about 120 mg, at least about 140 mg, at least about 160 mg, at least about 180 mg, at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, at least about 400 mg, at least about 420 mg, at least about 440 mg, at least about 460 mg, at least about 480 mg, at least about 500 mg, at least about 520 mg at least about 540 mg, at least about 550 mg, at least about 560 mg, at least about 580 mg, at least about 600 mg, at least about 620 mg, at least about 640 mg, at least about 660 mg, at least about 680 mg, at least about 700 mg, or at least about 720
  • ipilimumab is administered at a dose of about 3 mg/kg once about every 3 weeks. In some aspects, ipilimumab is administered at a dose of about 10 mg/kg once about every 3 weeks. In some aspects, ipilimumab is administered at a dose of about 10 mg/kg once about every 12 weeks. In some aspects, the ipilimumab is administered for four doses.
  • Anti-LAG-3 antibodies of the instant disclosure bind to human LAG-3. Antibodies that bind to LAG-3 have been disclosed in Int'l Publ. No. WO/2015/042246 and U.S. Publ. Nos. 2014/0093511 and 2011/0150892, each of which is incorporated by reference herein in its entirety.
  • An exemplary LAG-3 antibody useful in the present disclosure is 25F7 (described in U.S. Publ. No. 2011/0150892).
  • An additional exemplary LAG-3 antibody useful in the present disclosure is BMS-986016.
  • an anti-LAG-3 antibody useful for the composition cross-competes with 25F7 or BMS-986016.
  • an anti -LAG-3 antibody useful for the composition binds to the same epitope as 25F7 or BMS-986016.
  • an anti- LAG-3 antibody comprises six CDRs of 25F7 or BMS-986016.
  • the anti-LAG- 3 antibody is IMP731 (H5L7BW), MK-4280 (28G-10), REGN3767, humanized BAP050, IMP-
  • WO2016/028672 W02017/106129, WO2017/062888, W02009/044273, WO2018/069500, WO2016/126858, WO2014/179664, WO2016/200782, WO2015/200119, WO2017/019846, WO2017/198741, WO2017/220555, WO2017/220569, WO2018/071500, W02017/015560, WO2017/025498, WO2017/087589, WO2017/087901, W02018/083087, WO2017/149143, WO2017/219995,
  • Anti-CD137 antibodies specifically bind to and activate CD137-expressing immune cells, stimulating an immune response, in particular a cytotoxic T cell response, against tumor cells.
  • Antibodies that bind to CD137 have been disclosed in U.S. Publ. No. 2005/0095244 and U.S. Pat. Nos. 7,288,638, 6,887,673, 7,214,493, 6,303,121, 6,569,997, 6,905,685, 6,355,476, 6,362,325, 6,974,863, and 6,210,669, each of which is incorporated by reference herein in its entirety.
  • the anti-CD137 antibody is urelumab (BMS-663513), described in U.S. Pat. No. 7,288,638 (20H4.9-IgG4 [10C7 or BMS-663513]).
  • the anti- CD137 antibody is BMS-663031 (20H4.9-IgGl), described in U.S. Pat. No. 7,288,638.
  • the anti-CD137 antibody is 4E9 or BMS-554271, described in U.S. Pat. No. 6,887,673.
  • the anti-CD137 antibody is an antibody disclosed in U.S. Pat. Nos.
  • the anti-CD137 antibody is 1D8 or BMS-469492; 3H3 or BMS-469497; or 3E1, described in U.S. Pat. No. 6,362,325.
  • the anti-CD137 antibody is an antibody disclosed in issued U.S. Pat. No. 6,974,863 (such as 53A2).
  • the anti-CD137 antibody is an antibody disclosed in issued U.S. Pat. No. 6,210,669 (such as 1D8, 3B8, or 3E1).
  • the antibody is Pfizer's PF- 05082566 (PF-2566).
  • an anti-CD137 antibody useful for the methods disclosed herein cross-competes with the anti-CD137 antibodies disclosed herein.
  • an anti- CD137 antibody binds to the same epitope as the anti-CD137 antibody disclosed herein.
  • an anti-CD137 antibody useful in the disclosure comprises six CDRs of the anti-CD137 antibodies disclosed herein.
  • KIR Killer-cell immunoglobulin-like receptors
  • NK cells NK cells
  • Blocking these receptors facilitates activation of NK cells and, potentially, destruction of tumor cells by the latter.
  • Examples of anti-KIR antibodies have been disclosed in Int'l Publ. Nos. WO/2014/055648, WO 2005/003168, WO 2005/009465, WO 2006/072625, WO 2006/072626, WO 2007/042573, WO 2008/084106, WO 2010/065939, WO 2012/071411 and WO/2012/160448, each of which is incorporated by reference herein in its entirety.
  • One anti-KIR antibody useful in the present disclosure is lirilumab (also referred to as BMS-986015, IPH2102, or the S241P variant of 1-7F9), first described in Int'l Publ. No. WO 2008/084106.
  • An additional anti-KIR antibody useful in the present disclosure is 1-7F9 (also referred to as IPH2101), described in Int'l Publ. No. WO 2006/003179.
  • an anti- KIR antibody for the present composition cross competes for binding to KIR with lirilumab or I- 7F9.
  • an anti-KIR antibody binds to the same epitope as lirilumab or I-7F9.
  • an anti-KIR antibody comprises six CDRs of lirilumab or I-7F9.
  • Anti-GITR antibodies useful in the methods disclosed herein include any anti- GITR antibody that binds specifically to human GITR target and activates the glucocorticoid- induced tumor necrosis factor receptor (GITR).
  • GITR is a member of the TNF receptor superfamily that is expressed on the surface of multiple types of immune cells, including regulatory T cells, effector T cells, B cells, natural killer (NK) cells, and activated dendritic cells ("anti-GITR agonist antibodies"). Specifically, GITR activation increases the proliferation and function of effector T cells, as well as abrogating the suppression induced by activated T regulatory cells.
  • GITR stimulation promotes anti-tumor immunity by increasing the activity of other immune cells such as NK cells, antigen presenting cells, and B cells.
  • anti-GITR antibodies have been disclosed in Int'l Publ. Nos. WO/2015/031667, WO2015/184,099, WO2015/026,684, WO11/028683 and WO/2006/105021, U.S. Pat. Nos. 7,812,135 and 8,388,967 and U.S. Publ. Nos. 2009/0136494, 2014/0220002, 2013/0183321 and 2014/0348841, each of which is incorporated by reference herein in its entirety.
  • an anti-GITR antibody useful in the present disclosure is TRX518 (described in, for example, Schaer et al. Curr Opin Immunol. (2012) Apr; 24(2): 217-224, and WO/2006/105021).
  • the anti-GITR antibody is selected from MK4166, MK1248, and antibodies described in WO11/028683 and U.S. 8,709,424, and comprising, e.g., a VH chain comprising SEQ ID NO: 104 and a VL chain comprising SEQ ID NO: 105 (wherein the SEQ ID NOs are from WO11/028683 or U.S. 8,709,424).
  • an anti-GITR antibody is an anti-GITR antibody that is disclosed in WO2015/031667, e.g., an antibody comprising VH CDRs 1-3 comprising SEQ ID NOs: 31, 71 and 63 of WO2015/031667, respectively, and VL CDRs 1-3 comprising SEQ ID NOs: 5, 14 and 30 of WO2015/031667.
  • an anti-GITR antibody is an anti-GITR antibody that is disclosed in WO2015/184099, e.g., antibody Hum231#l or Hum231#2, or the CDRs thereof, or a derivative thereof (e.g., pabl967, pabl975 or pabl979).
  • an anti-GITR antibody is an anti- GITR antibody that is disclosed in JP2008278814, W009/009116, WO2013/039954, US20140072566, US20140072565, US20140065152, or WO2015/026684, or is INBRX-110 (INHIBRx), LKZ-145 (Novartis), or MEDI-1873 (Medlmmune).
  • an anti-GITR antibody is an anti-GITR antibody that is described in PCT/US2015/033991 (e.g., an antibody comprising the variable regions of 28F3, 18E10 or 19D3).
  • the anti-GITR antibody cross-competes with an anti-GITR antibody described herein, e.g., TRX518, MK4166 or an antibody comprising a VH domain and a VL domain amino acid sequence described herein.
  • the anti-GITR antibody binds the same epitope as that of an anti-GITR antibody described herein, e.g., TRX518 or MK4166.
  • the anti-GITR antibody comprises the six CDRs of TRX518 or MK4166.
  • any anti-TIM3 antibody or antigen binding fragment thereof known in the art can be used in the methods described herein.
  • the anti-TIM3 antibody is be selected from the anti-TIM3 antibodies disclosed in Int'l Publ. Nos.W02018013818, WO/2015/117002 (e.g., MGB453, Novartis), WO/2017/161270 (e.g., TSR-022, Tesaro/AnaptysBio),
  • WO201 1155607, WO2016/144803 e.g., STI-600, Sorrento Therapeutics
  • WO2016/0714408 WO17055399; W017055404, WO17178493, WO18036561, W018039020 (e.g., Ly-3221367, Eli Lilly), WO2017205721, WO17079112; WO17079115; WO17079116, WO11159877, W013006490, W02016068802 W02016068803, WO2016/111947, and WO/2017/031242, each of which is incorporated by reference herein in its entirety. II.C.9.
  • any antibody or antigen-binding fragment thereof that specifically binds 0X40 can be used in the methods disclosed herein.
  • the anti-OX40 antibody is BMS-986178 (Bristol-Myers Squibb Company), described in Int'l Publ. No. WO20160196228.
  • the anti- 0X40 antibody is selected from the anti-OX40 antibodies described in Int'l Publ. Nos.
  • NKG2A is a member of the C-type lectin receptor family that is expressed on natural killer (NK) cells and a subset of T lymphocytes. Specifically, NKG2A primarily expressed on tumor infiltrating innate immune effector NK cells, as well as on some CD8+ T cells. Its natural ligand human leukocyte antigen E (HLA-E) is expressed on solid and hematologic tumors. NKG2A is an inhibitory receptor that blinds HLA-E.
  • NK natural killer
  • HLA-E human leukocyte antigen E
  • the anti-NKG2A antibody may be BMS-986315, a human monoclonal antibody that blocks the interaction of NKG2A to its ligand HLA-E, thus allowing activation of an anti-tumor immune response.
  • the anti-NKG2A antibody is a checkpoint inhibitor that activates T cells, NK cells, and/or tumor-infiltrating immune cells.
  • the anti-NKG2A antibody is selected from the anti-NKG2A antibodies described in, for example, WO 2006/070286 (Innate Pharma S.A.; University of Genova); U.S. Patent No.
  • ICOS Anti-ICOS Antibodies
  • ICOS is an immune checkpoint protein that is a member of the CD28-superfamily.
  • ICOS is a 55-60 kDa type I transmembrane protein that is expressed on T cells after T cell activation and co-stimulates T-cell activation after binding its ligand, ICOS-L (B7H2).
  • ICOS is also known as inducible T-cell co-stimulator, CVID1, AILIM, inducible costimulator, CD278, activation-inducible lymphocyte immunomediatory molecule, and CD278 antigen.
  • the anti-ICOS antibody is BMS-986226, a humanized IgG monoclonal antibody that binds to and stimulates human ICOS.
  • the anti-ICOS antibody is selected from anti-ICOS antibodies described in, for example, WO 2016/154177 (Jounce Therapeutics, Inc.), WO 2008/137915 (Medlmmune), WO 2012/131004 (INSERM, French National Institute of Health and Medical Research), EP3147297 (INSERM, French National Institute of Health and Medical Research), WO 2011/041613 (Memorial Sloan Kettering Cancer Center), EP 2482849 (Memorial Sloan Kettering Cancer Center), WO 1999/15553 (Robert Koch Institute), U.S.
  • Patent Nos. 7,259,247 and 7,722,872 Robot Kotch Institute
  • WO 1998/038216 Japanese Tobacco Inc.
  • US. Patents. Nos. 7,045,615; 7,112,655, and 8,389,690 Japan Tobacco Inc.
  • U.S. Patent Nos. 9,738,718 and 9,771,424 GaxoSmithKline
  • WO 2017/220988 Kelmab Limited
  • the anti-TIGIT antibody is BMS- 986207.
  • the anti-TIGIT antibody is clone 22G2, as described in WO 2016/106302.
  • the anti-TIGIT antibody is MTIG7192A/RG6058/RO7092284, or clone 4.1D3, as described in WO 2017/053748.
  • the anti-TIGIT antibody is selected from the anti-TIGIT antibodies described in, for example, WO 2016/106302 (Bristol- Myers Squibb Company) and WO 2017/053748 (Genentech).
  • the anti-CSFIR antibody is an antibody species disclosed in any of international publications WO2013/132044, W02009/026303, WO2011/140249, or W02009/112245, such as cabiralizumab, RG7155 (emactuzumab), AMG820, SNDX 6352 (UCB 6352), CXIIG6, IMC-CS4, JNJ-40346527, MCS1 10, or the anti-CSFIR antibody in the methods is replaced with an anti-CSFIR inhibitor or anti-CSFl inhibitor such as BLZ-945, pexidartinib (PLX3397, PLX108-01), AC-708, PLX-5622, PLX7486, ARRY-382, or PLX-73086.
  • an anti-CSFIR inhibitor or anti-CSFl inhibitor such as BLZ-945, pexidartinib (PLX3397, PLX108-01), AC-708, PLX-5622, PLX7486
  • the methods disclosed herein comprise administering an 1-0 therapy, e.g., an anti-PD-1 antibody or an anti-PD-Ll antibody, and one or more additional anti-cancer therapies.
  • the method comprising administering (i) a first 1-0 therapy, e.g., an anti-PD-1 antibody or an anti-PD-Ll antibody), (ii) a second 1-0 therapy, e.g., an anti-CTLA-4 antibody or an anti-CSFIR antibody, and (iii) one or more additional anti-cancer therapies.
  • the methods dislosed herein comprise administering an anti-cancer therapy to a subject identified as having a low pan-tumor gene signature score, wherein the anti-cancer agent does not comprise an 1-0 therapy.
  • the anti-cancer therapy can comprise any therapy known in the art for the treatment of a tumor in a subject and/or any standard-of-care therapy, as disclosed herein.
  • the anti-cancer therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof.
  • the anti-cancer therapy comprises a chemotherapy, including any chemotherapy disclosed herein.
  • the chemotherapy is a platinum based-chemotherapy.
  • Platinum-based chemotherapies are coordination complexes of platinum.
  • the platinum-based chemotherapy is a platinum-doublet chemotherapy.
  • the chemotherapy is administered at the approved dose for the particular indication.
  • the chemotherapy is administered at any dose disclosed herein.
  • the platinum-based chemotherapy is cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, Nedaplatin, Triplatin, Lipoplatin, or combinations thereof.
  • the platinum-based chemotherapy is any other platinum-based chemotherapy known in the art.
  • the chemotherapy is the nucleotide analog gemcitabine.
  • the chemotherapy is a folate antimetabolite.
  • the folate antimetabolite is pemetrexed.
  • the chemotherapy is a taxane.
  • the taxane is paclitaxel.
  • the chemotherapy is any other chemotherapy known in the art.
  • at least one, at least two or more chemotherapeutic agents are administered in combination with the 1-0 therapy.
  • the 1-0 therapy is administered in combination with gemcitabine and cisplatin.
  • the 1-0 therapy is administered in combination with pemetrexed and cisplatin.
  • the 1-0 therapy is administered in combination with gemcitabine and pemetrexed.
  • the 1-0 therapy is administered in combination with paclitaxel and carboplatin.
  • an I- O therapy is additionally administered.
  • the additional anti-cancer therapy e.g., administered to a subject having a moderate pan-tumor gene signature score, comprises an immunotherapy.
  • the additional anti-cancer therapy comprises administration of an antibody or antigenbinding portion thereof that specifically binds LAG-3, TIGIT, TIM3, NKG2a, CSF1R, 0X40, ICOS, MICA, MICB, CD137, KIR, TGFp, IL-10, IL-8, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, or any combination thereof.
  • the tumor is derived from a cancer selected from the group consisting of hepatocellular cancer, gastroesophageal cancer, melanoma, bladder cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC)), kidney cancer, renal cell carcinoma, head and neck cancer (e.g., squamous cell carcinoma of the head and neck), pancreatic cancer, gastric cancer, ovarian cancer, prostate cancer, colon cancer, colorectal cancer, urothelial carcinoma, and any combination thereof.
  • the tumor is derived from a hepatocellular cancer, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a gastroesophageal cancer, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a melanoma, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a bladder cancer, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a lung cancer, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a kidney cancer, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a head and neck cancer, wherein the tumor has a high inflammatory signature score.
  • the tumor is derived from a colon cancer, wherein the tumor has a high inflammatory signature score.
  • the subject has received one, two, three, four, five or more prior cancer treatments.
  • the subject is treatment-naive.
  • the subject has progressed on other cancer treatments.
  • the prior cancer treatment comprised an immunotherapy.
  • the prior cancer treatment comprised a chemotherapy.
  • the tumor has reoccurred.
  • the tumor is metastatic.
  • the tumor is not metastatic.
  • the tumor is locally advanced.
  • the subject has received a prior therapy to treat the tumor and the tumor is relapsed or refractory.
  • the at least one prior therapy comprises a standard-of-care therapy.
  • the at least one prior therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof.
  • the at least one prior therapy comprises a chemotherapy.
  • the subject has received a prior immuno-oncology (I-O) therapy to treat the tumor and the tumor is relapsed or refractory.
  • I-O immuno-oncology
  • the subject has received more than one prior therapy to treat the tumor and the subject is relapsed or refractory.
  • the subject has received either an anti-PD-1 or anti-PD-Ll antibody therapy.
  • the previous line of therapy comprises a chemotherapy.
  • the chemotherapy comprises a platinum-based therapy.
  • the platinumbased therapy comprises a platinum-based antineoplastic selected from the group consisting of cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and any combination thereof.
  • the platinum-based therapy comprises cisplatin.
  • the platinum-based therapy comprises carboplatin.
  • the at least one prior therapy is selected from a therapy comprising administration of an anti-cancer agent selected from the group consisting of a platinum agent (e.g., cisplatin, carboplatin), a taxanes agent (e.g., paclitaxel, albumin-bound paclitaxel, docetaxel), vinorelbine, vinblastine, etoposide, pemetrexed, gemcitabine, bevacizumab (AVASTIN®), erlotinib (TARCEVA®), crizotinib (XALKORI®), cetuximab (ERBITUX®), and any combination thereof.
  • the at least one prior therapy comprises a platinum-based doublet chemotherapy.
  • the subject has experienced disease progression after the at least one prior therapy.
  • the subject has received at least two prior therapies, at least three prior therapies, at least four prior therapies, or at least five prior therapies.
  • the subject has received at least two prior therapies.
  • the at least two prior therapies comprises a first prior therapy and a second prior therapy, wherein the subject has experienced disease progression after the first prior therapy and/or the second prior therapy, and wherein the first prior therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof; and wherein the second prior therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof.
  • the first prior therapy comprises a platinum-based doublet chemotherapy
  • the second prior therapy comprises a single-agent chemotherapy.
  • the single-agent chemotherapy comprises docetaxel.
  • Therapeutic agents of the present disclosure can be constituted in a composition, e.g., a pharmaceutical composition containing an antibody and/or a cytokine and a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier for a composition containing an antibody is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion), whereas the carrier for a composition containing an antibody and/or a cytokine is suitable for non-parenteral, e.g., oral, administration.
  • the subcutaneous injection is based on Halozyme Therapeutics’ ENHANZE® drug-delivery technology (see U.S. Patent No. 7,767,429, which is incorporated by reference herein in its entirety).
  • ENHANZE® uses a coformulation of an antibody with recombinant human hyaluronidase enzyme (rHuPH20), which removes traditional limitations on the volume of biologies and drugs that can be delivered subcutaneously due to the extracellular matrix (see U.S. Patent No. 7,767,429).
  • a pharmaceutical composition of the disclosure can include one or more pharmaceutically acceptable salts, antioxidant, aqueous and non-aqueous carriers, and/or adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Therefore, in some aspects, the pharmaceutical composition for the present disclosure can further comprise recombinant human hyaluronidase enzyme, e.g., rHuPH20.
  • nivolumab monotherapy dosing up to 10 mg/kg every two weeks has been achieved without reaching the maximum tolerated does (MTD)
  • MTD maximum tolerated does
  • the significant toxi cities reported in other trials of checkpoint inhibitors plus anti-angiogenic therapy support the selection of a nivolumab dose lower than 10 mg/kg.
  • the antibodies disclosed herein are administered at doses that are significantly lower than the approved dosage, i.e., a subtherapeutic dosage, of the agent.
  • the antibody can be administered at the dosage that has been shown to produce the highest efficacy as monotherapy in clinical trials, e.g., about 3 mg/kg of nivolumab administered once every three weeks (Topalian et al., 2012a; Topalian et al., 2012), or at a significantly lower dose, i.e., at a subtherapeutic dose.
  • Dosage and frequency vary depending on the half-life of the antibody in the subject. In general, human antibodies show the longest half-life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies.
  • the dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is typically administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • compositions of the present disclosure can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being unduly toxic to the patient.
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present disclosure employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a composition of the present disclosure can be administered via one or more routes of administration using one or more of a variety of methods well known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
  • kits comprising (a) an anti-PD- 1 antibody or an anti-PD-Ll antibody for therapeutic uses.
  • Kits typically include a label indicating the intended use of the contents of the kit and instructions for use.
  • the term label includes any writing, or recorded material supplied on or with the kit, or which otherwise accompanies the kit.
  • this disclosure provides a kit for treating a subject afflicted with a tumor, the kit comprising: (a) a dosage ranging from 0.1 to 10 mg/kg body weight of an anti-PD-1 antibody or a dosage ranging from 0.1 to 20 mg/kg body weight of an anti-PD-Ll antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-Ll antibody in the methods disclosed herein.
  • kits for treating a subject afflicted with a tumor comprising: (a) a dosage ranging from about 4 mg to about 500 mg of an anti-PD-1 antibody or a dosage ranging from about 4 mg to about 2000 mg of an anti-PD-Ll antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-Ll antibody in the methods disclosed herein.
  • this disclosure provides a kit for treating a subject afflicted with a tumor, the kit comprising: (a) a dosage ranging from 200 mg to 800 mg of an anti-PD-1 antibody or a dosage ranging from 200 mg to 1800 mg of an anti-PD-Ll antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-Ll antibody in the methods disclosed herein.
  • the kit comprises an anti-human PD-1 antibody disclosed herein, e.g., nivolumab or pembrolizumab.
  • the kit comprises an anti-human PD-L1 antibody disclosed herein, e.g., atezolizumab, durvalumab, or avelumab.
  • the kit further comprises an anti-CTLA-4 antibody.
  • the kit comprises an anti-human CTLA-4 antibody disclosed herein, e.g., ipilimumab, tremelimumab, MK-1308, or AGEN-1884.
  • the kit further includes a gene panel assay disclosed herein. In some aspects, the kit further includes instructions to administer the anti-PD-1 antibody or the anti-PD-Ll antibody to a suitable subject according to the methods disclosed herein.
  • TME tumor microenvironment
  • IHC immunohistochemistry
  • RNA sequencing RNA-seq
  • GEP panels targeting select sets of genes or pathways.
  • cross-platform consistency remains to be determined.
  • CD8 signature A 16-gene expression signature (CD8 signature) has been developed as an investigational use only assay that correlated with pathology-assisted digital scoring of CD8 expression by IHC (CD8 IHC) across multiple tumor types (Szabo et al. J Clin Oncol 2019;37(suppl) Abstract 2593).
  • the CD8 signature was associated with clinical efficacy in post- hoc analyses of samples from patients with urothelial carcinoma treated with nivolumab in a clinical trial for the treatment of urothelial carcinoma (Szabo et al. J Immunother Cancer 2019;7(suppl 1) Abstract Pl 09).
  • GEP panels include a 2-gene T-cell abundance (TCA) signature, which can be used as a measure of CD8+ T-cell infiltration in the TME (see Danaher et al. J Immunother Cancer 2017;5(l 8)) and an 18-gene tumor inflammation signature (TIS), which was associated with inflammatory responses and clinical efficacy of 1-0 therapy in post-hoc analyses across multiple tumor types (see Danaher et al. J Immunother Cancer 2018;6(63); Cristescu et al. Science 2018;362:eaar3593).
  • TCA 2-gene T-cell abundance
  • TIS 18-gene tumor inflammation signature
  • the present example presents data from a study designed to assess correlations between gene expression signatures and other predictive biomarkers of response to immune checkpoint inhibitors.
  • this study compared CD8 IHC with three different gene expression signatures to assess their suitability as surrogate biomarkers of inflammation; investigated the relationship between gene expression signatures and programmed death ligand 1 (PD-L1) expression on tumor cells, assessed using IHC; and evaluated the consistency of the CD8 signature derived from tumor samples using RNA-seq and four different gene expression panels.
  • PD-L1 programmed death ligand 1
  • FFPE paraffin-embedded
  • CD8 signature scores were derived using data from RNA-seq and the four GEP panels.
  • RNA-seq was used as a reference to derive Pearson’s correlation coefficients (r) for cross-platform comparisons.
  • the TCA (Danaher P, et al. J Immunother Cancer 2017;5(l 8)) and TIS (Danaher P, et al. J Immunother Cancer 2018;6(63)) scores were derived using data from the IO 360 panel.
  • CD8 IHC was performed using a monoclonal CD8 antibody.
  • CD8+ T cells were quantified using pathologist-supervised, digital scoring-based analysis of 20* whole slide images. Scores were calculated from CD8+ T cells as a percentage of total cells in the tumor parenchyma, tumor stroma, and tumor margin.
  • Melanoma (MEL) samples were labelled with an alkaline phosphatase (AP)-linked secondary antibody (PowerVision Poly-AP Anti-Mouse IgG; Leica Biosystems, Buffalo Grove, IL) and visualized using Fast Red chromogen (Leica). All other tumor types were labeled with Envision FLEX+ Mouse LINKER antibody and horseradish peroxidase polymer (Agilent) and visualized with 3, 3 '-diaminobenzidine (DAB).
  • AP alkaline phosphatase
  • Tissues stained using Fast Red and DAB were evaluated using a Nuclear v9 Mosaic Red-Melanin MTM UT220-vl and Nuclear v9 MTM UT 220 algorithms, respectively (both Aperio).
  • CD8 IHC scores were square-root transformed to meet linear model distribution assumptions.
  • PD-L1 expression on tumor cells was assessed using the Dako PD-L1 IHC 28-8 pharmDx assay (Agilent Technologies, Santa Clara, CA).
  • PD-Ll-positive (PD-L1+) and PD-L1 -negative (PD-L1-) status were determined around a cutoff of PD-L1 expression on 1% of tumor cells.
  • Pearson’s correlation coefficients (r) were derived from comparisons between gene signature scores and CD8 IHC or PD-L1 IHC.
  • a region of interest was defined by a pathologist to include tumor parenchyma and contiguous tumor-associated stroma within 500 pm of the edge of the tumor.
  • Sample characteristics including tumor grade, biopsy type, tumor content, percent necrosis
  • Tissue outside the ROI was then removed, and remaining samples were scraped into sterile 1.7 mL nuclease-free microcentrifuge tubes using a sterile razor blade.
  • Inflammation gene signatures were determined using RNA-seq and 3 alternative GEP panel assays (Tumor Immunology Panel [TIP], PanCancer IO 360TM Panel, and OncomineTM Immune Response Research Assay) (Table XX).
  • GEP gene expression profiling
  • NGS next-generation sequencing
  • RNA-seq RNA sequencing.
  • TMM Trimmed Mean of M-values
  • the CD8 signature was developed and analytically validated as an IUO assay using the custom TIP.
  • This EdgeSeq (HTG Molecular Diagnostics, Arlington, AZ) panel comprises 95 probes, including genes related to tumor inflammation, other 1-0 genes of interest, housekeeping genes, and control probes. mRNA expression levels of all 95 probes on the panel were determined using an automated, extraction-free, nuclease protection assay, combined with NGS.
  • a minimum sample area of 7.2 mm 2 macrodissected tissue from an unstained section was required for GEP using the TIP (tissue from more than one section was used in some cases).
  • Tissues were lysed in lysis buffer (HTG Molecular Diagnostics; 50 pL per 7.2 mm 2 tissue) and incubated for 15-20 minutes at 95°C.
  • Proteinase K was added to a final concentration of 1 mg/mL, and samples were incubated for 3 hours at 50°C with shaking. Each sample was then added to a well of a 96-well plate. Nuclease protection probes were added to the lysed samples and the nuclease protection step was performed using an HTG EdgeSeq Processor with controller software version v6.1.653.2205.
  • SI nuclease was then added to degrade nonhybridized probes and nontargeted RNA, followed by heat inactivation (85°C, 20 minutes, pH 9). Samples were then individually barcoded using a 16-cycle PCR to add adapters and molecular barcodes. Barcoded samples were individually purified using Agencourt AMPure XP beads (Beckman Coulter, Indianapolis, IN; catalog number A63880) and quantitated using a KAPA Library Quantification kit (Kapa Biosciences, Wilmington, MA; catalog number KK4873). PCR- enriched libraries were sequenced using a MiSeqDx with controller software version v2.5.05 (Illumina).
  • NGS data were processed using HTG EdgeSeq Host System Software (HTG Molecular Diagnostics). Demultiplexed FASTQ files were parsed and aligned to probe FASTQ files and raw counts were transformed to log2 counts per million (CPM). No additional normalization was applied. Normalized data were filtered based on quality control metrics examined in the following order: input RNA quality (samples with reads for positive control probes ⁇ 21% of total reads passed this filter), sequencing read depth (cutoff varied by number of samples in the assay plate), and samples with minimum expression variability (samples passed if the relative standard deviation of log2 CPM among 77 probes was > 0.19).
  • TIS Tumor Inflammation Signature
  • TCA 2-gene CD8+ T-cell abundance
  • GEP was performed on the same pool of extracted RNA as for RNA-seq, using the OncomineTM Immune Response Research Assay (Thermo Fisher Scientific, Waltham, MA), targeting 395 genes according to manufacturer’s instructions. Absolute digital gene expression counts were generated using Torrent Suite software (v5.0.2) with the immuneResponseRNA plugin (Thermo Fisher Scientific). TMM normalization was applied using the EdgeSeq Host System Software (HTG Molecular Diagnostics), and normalized data were transformed to log2 CPM.
  • a machine-learning method utilized a multivariate regression model to develop a classifier with 63 probes on the TIP as the predictors and CD8 IHC as the outcome variable.
  • Tumor-specific signatures were derived independently for MEL and SCCHN samples.
  • a similar approach was then applied to pooled MEL and SCCHN samples to derive a combined MEL/SCCHN signature.
  • the least absolute shrinkage and selection operator (LASSO) technique with optimal shrinkage parameters for lambda.min and lambda.
  • CD8 signature multitumor 16-gene expression model
  • TCA T-cell abundance
  • TIS tumor inflammation signature
  • CD8+ T-cell abundance was assessed by GEP and IHC in parallel using 1633 matched samples from a total of 12 tumor types.
  • Reasons for sample attrition included insufficient RNA, insufficient read depth, or low variation in gene expression.
  • CD8 expression via CD8 IHC and the GEP-derived CD8 signature score, was consistent across most of the tumor types investigated (FIG. 2).
  • MEL, SCCHN, NSCLC, and GC samples showed higher median CD8 scores and wider ranges by both IHC and GEP.
  • Lower ranges and median scores by GEP were observed in tumor types with lower CD8 expression by IHC (SCLC, OVC, and PC) (FIG. 2).
  • FIGs. 4A-4L shows the proportion of samples with CD8 IHC scores or CD8 signature scores above cutoff values that varied from 0-40.
  • CD8 signature scores were assessed in 15 replicates from 8 samples per tumor type. Samples that were representative of the dynamic range of inflammation seen in each of the 12 tumor types were selected. Across all tumor types, % CV decreased as a function of increasing CD8 signature score, and variation was reduced to ⁇ 25 in samples with CD8 signature scores > 5 (FIG. 5).
  • FIGs. 6A-6Q show correlations between gene expression data for the 16 genes in the CD8 signature, derived using RNA-seq vs one of 3 different GEP panels. Median Pearson’s r and interquartile ranges for individual gene expression were comparable across the 3 GEP panels [0.87 (0.78-0.94), 0.82 (0.66-0.86), and 0.71 (0.44-0.83) for the PanCancer IO 360TM Panel, OncomineTM Immune Response Research Assay, and TIP, respectively].
  • the CD8 signature was determined in pretreatment platinum-resistant metastatic UC samples from patients subsequently treated with nivolumab (3 mg/kg intravenously every 2 weeks). Of 270 samples from patients enrolled in CheckMate 275 (NCT02387996), 263 (97%) were evaluable for CD8 IHC, 205 (76%) were evaluable for the CD8 signature by GEP, and 203 (75%) were evaluable for both CD8 IHC and the CD8 signature. Reasons for sample attrition included insufficient tumor biopsy available for GEP or insufficient RNA quality.
  • Table XX Baseline Characteristics in Patients From CheckMate 275.
  • Prolonged OS was also observed in patients with CD8 signature scores in the upper tertile (22.14 months, 95% CI, 9.99-30.72) compared with patients with CD8 signature scores in the lower two tertiles (5.72 months, 95% CI, 4.37-11.43, and 6.24 months, 95% CI, 4.53-9.00, for patients with scores in the middle and lower tertiles, respectively; FIG. 1 IB).
  • CD8+ T cells Infiltration of CD8+ T cells, often assessed by IHC, can be used as a robust marker of inflammation that, for some tumor types, is associated with prognosis and/or response to treatment.
  • the CD8 signature was developed to predict CD8+ T-cell abundance across multiple tumor types and analytically validated across 1633 commercial samples. Inflammation status determined by the CD8 signature score correlated with that derived by CD8 IHC (r > 0.82) in all tumor types tested except pancreatic and prostate cancer, indicating that this gene signature may be utilized as an accurate surrogate measure of inflammation across a range of tumor types.
  • CD8-positivity was similar for GEP -based and IHC -based methods, suggesting that a CD8 signature assay has the potential to be utilized prospectively in a clinical trial setting to determine inflammation-positive patients.
  • CD8+ T-cell abundance was lower than observed in other tumor types, and a high correlation between CD8 signature and CD8 IHC scores was observed.
  • a linear increase in CD8 signature scores resulted in fewer samples falling within the 95% CI for CD8 signature vs CD8 IHC correlations than seen for other tumor types and in increased variability in the assessment of inflammation prevalence.
  • the sample processing, pathological evaluation, and data processing steps employed in this IUO assay have enabled reliable GEP -based surrogate assessment of CD8+ T- cell abundance in the TME.
  • the IUO CD8 signature assay demonstrated high reproducibility when using the TIP to determine CD8 signature scores across 12 tumor types. Assay precision (CV ⁇ 25%) was demonstrated for samples with CD8 signature scores > 5.
  • CD8 signature scores correlated with CD8 IHC scores on all platforms tested, demonstrating platform-independent consistency for the CD8 signature as a surrogate measure of CD8+ T-cell abundance. Similar results were observed for select alternative GEP-derived tumor inflammation signatures, demonstrating the robust nature of inflammation gene expression signatures and potential feasibility of utilizing multiple GEP platforms for their assessment.
  • the TIP assay combines extraction-free RNase protection with NGS in an automated EdgeSeq system.
  • This system can generate high-quality sequencing data from small amounts of FFPE samples (as little as 7.2 mm 2 of a 4-5 pm section), overcoming some of the limitations experienced with alternative GEP methods, including sample acquisition and assay standardization.
  • CD8 signature scores did not correlate with PD-L1 expression on tumor cells, although a trend for increased CD8 signature score was observed in PD-L1 -positive patients. Additional studies are warranted to assess the relationship between CD8 signature scores and PD-L1 expression on tumor cells, immune cells, or both. The lack of correlation between potential biomarkers of response to ICIs suggests that some or all of these may reflect distinct, nonoverlapping, biological aspects of inflammation in the TME and provides a rationale for additional studies to evaluate the potential of multiple biomarkers in combination.

Abstract

The present disclosure provides methods of identifying a subject suitable for an immuno-oncology (I-O) therapy comprising measuring the expression of a pan-tumor inflammation gene panel. In some aspects, the method further comprises administering an 1-0 therapy to the subject. In some aspects, the I-O therapy comprises administering an anti -PD-1 antibody or antigen-binding portion thereof or an anti-PD-Ll antibody or antigen-binding portion thereof to the subject.

Description

MULTI-TUMOR GENE SIGNATURES AND USES THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This PCT application claims the priority benefit of U.S. Provisional Application No. 63/121,137, filed December 3, 2020, which is incorporated herein by reference in its entirety.
FIELD OF THE DISCLOSURE
[0002] The present disclosure provides a method for treating a subject afflicted with a tumor using an immunotherapy.
BACKGROUND OF THE DISCLOSURE
[0003] Human cancers harbor numerous genetic and epigenetic alterations, generating neoantigens potentially recognizable by the immune system (Sjoblom et al., Science (2006) 314(5797):268-274). The adaptive immune system, comprised of T and B lymphocytes, has powerful anti-cancer potential, with a broad capacity and exquisite specificity to respond to diverse tumor antigens. Further, the immune system demonstrates considerable plasticity and a memory component. The successful harnessing of all these attributes of the adaptive immune system would make immunotherapy unique among all cancer treatment modalities.
[0004] In the past decade, intensive efforts to develop specific immune checkpoint pathway inhibitors have begun to provide new immunotherapeutic approaches for treating cancer, including the development of antibodies that block the inhibitory Programmed Death- 1 (PD-1)/Programmed Death ligand 1 (PD-L1) pathway such as nivolumab and pembrolizumab (formerly lambrolizumab; USAN Council Statement, 2013) that bind specifically to the PD-1 receptor and atezolizumab, durvalumab, and avelumab that bind specifically to PD-L1 (Topalian et al., 2012a, b; Topalian et al., 2014; Hamid et al., 2013; Hamid and Carvajal, 2013; McDermott and Atkins, 2013).
[0005] The immune system and response to immuno-therapy have shown to be complex. Additionally, anti-cancer agents can vary in their effectiveness based on the unique patient characteristics. Accordingly, there is a need for targeted therapeutic strategies that identify patients who are more likely to respond to a particular anti-cancer agent and, thus, improve the clinical outcome for patients diagnosed with cancer.
SUMMARY OF THE DISCLOSURE
[0006] Some aspects of the present disclosure are directed to a pharmaceutical composition comprising an immune-oncology (I-O) therapy for use in a method of treating a tumor in a subject in need thereof, wherein the subject has been identified as having a high pantumor gene signature score; wherein the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and wherein the pan-tumor gene signature score is determined according to the following formula: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein: (a) = any real number between 0.01 and 0.05; (b) = any real number between 0.10 and 0.15; (c) = any real number between 0.01 and 0.05; (d) = any real number between -0.10 and -0.15; (e) = any real number between 0.03 and 0.09; (f) = any real number between 0.35 and 0.45; (g) = any real number between 0.07 and 0.13; (h) = any real number between 0.02 and 0.08; (i) = any real number between -0.01 and -0.05; (j) = any real number between -0.0001 and -0.0010; (k) = any real number between 0.01 and 0.06; (1) = any real number between 0.02 and 0.08; (m) = any real number between -0.008 and -0.040; (n) = any real number between 0.01 and 0.050; (o) = any real number between -0.03 and -0.080; and (p) = any real number between -0.005 and -0.010. In some aspects, the high pan-tumor gene signature score is characterized by a score of greater than about 12.6.
[0007] Some aspects of the present disclosure are directed to a pharmaceutical composition comprising an anticancer agent for use in a method of treating a tumor in a subject in need thereof, wherein the anticancer agent does not comprise an 1-0 therapy, wherein the subject has been identified as having a low pan-tumor gene signature score; wherein the pantumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HL A- DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and wherein the pan-tumor gene signature score is determined according to the following formula: pan-tumor signature score = [CCL4] *
(a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA- DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein: (a) = any real number between 0.01 and 0.05; (b) = any real number between 0.10 and 0.15; (c) = any real number between 0.01 and 0.05; (d) = any real number between -0.10 and - 0.15; (e) = any real number between 0.03 and 0.09; (f) = any real number between 0.35 and 0.45; (g) = any real number between 0.07 and 0.13; (h) = any real number between 0.02 and 0.08; (i) = any real number between -0.01 and -0.05; (j) = any real number between -0.0001 and -0.0010; (k) = any real number between 0.01 and 0.06; (1) = any real number between 0.02 and 0.08; (m) = any real number between -0.008 and -0.040; (n) = any real number between 0.01 and 0.050; (o) = any real number between -0.03 and -0.080; and (p) = any real number between -0.005 and -0.010. In some aspects, the low pan-tumor gene signature score is characterized by a score of less than about 7.9.
[0008] Some aspects of the present disclosure are directed to a pharmaceutical composition comprising an 1-0 therapy for use in a method of treating a tumor in a subject in need thereof, wherein the 1-0 therapy is to be administered in combination with an additional anticancer agent to a subject that has been identified as having a moderate pan-tumor gene signature score; wherein the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and wherein the pan-tumor gene signature score is determined according to the following formula: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA- DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein: (a) = any real number between 0.01 and 0.05;
(b) = any real number between 0.10 and 0.15; (c) = any real number between 0.01 and 0.05; (d) = any real number between -0.10 and -0.15; (e) = any real number between 0.03 and 0.09; (f) = any real number between 0.35 and 0.45; (g) = any real number between 0.07 and 0.13; (h) = any real number between 0.02 and 0.08; (i) = any real number between -0.01 and -0.05; (j) = any real number between -0.0001 and -0.0010; (k) = any real number between 0.01 and 0.06; (1) = any real number between 0.02 and 0.08; (m) = any real number between -0.008 and -0.040; (n) = any real number between 0.01 and 0.050; (o) = any real number between -0.03 and -0.080; and (p) = any real number between -0.005 and -0.010. In some aspects, the moderate pan-tumor gene signature score is characterized by a score of from about 7.9 to about 12.6.
[0009] Some aspects of the present disclosure are directed to a pan-tumor gene panel for use in a method of identifying a subject suitable for an 1-0 therapy comprising calculating a pantumor gene signature score for a tumor sample obtained from the subject, wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the pantumor gene panel in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying the following formula to the measured expression values: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein: (a) = any real number between 0.01 and 0.05; (b) = any real number between 0.10 and 0.15; (c) = any real number between 0.01 and 0.05; (d) = any real number between -0.10 and -0.15; (e) = any real number between 0.03 and 0.09; (f) = any real number between 0.35 and 0.45; (g) = any real number between 0.07 and 0.13; (h) = any real number between 0.02 and 0.08; (i) = any real number between -0.01 and -0.05; (j) = any real number between -0.0001 and -0.0010; (k) = any real number between 0.01 and 0.06; (1) = any real number between 0.02 and 0.08; (m) = any real number between -0.008 and -0.040; (n) = any real number between 0.01 and 0.050; (o) = any real number between -0.03 and -0.080; and (p) = any real number between -0.005 and -0.010. In some aspects, the subject is identified as being suitable for an 1-0 therapy if the subject has a high pantumor gene signature score.
[0010] Some aspects of the present disclosure are directed to a pan-tumor gene panel for use in a method of stratifying a subject in need of an 1-0 therapy, comprising determining a pantumor gene signature score for a tumor sample obtained from the subject, wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying the following formula to the measured expression values: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA- DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein: (a) = any real number between 0.01 and 0.05; (b) = any real number between 0.10 and 0.15; (c) = any real number between 0.01 and 0.05; (d) = any real number between -0.10 and -0.15; (e) = any real number between 0.03 and 0.09; (f) = any real number between 0.35 and 0.45; (g) = any real number between 0.07 and 0.13; (h) = any real number between 0.02 and 0.08; (i) = any real number between -0.01 and -0.05; (j) = any real number between -0.0001 and -0.0010; (k) = any real number between 0.01 and 0.06; (1) = any real number between 0.02 and 0.08; (m) = any real number between -0.008 and -0.040; (n) = any real number between 0.01 and 0.050; (o) = any real number between -0.03 and -0.080; and (p) = any real number between -0.005 and -0.010; and wherein the subject is identified as having a high pan-tumor gene signature score if the score greater than about 12.6; wherein the subject is identified as having a low pan-tumor gene signature score if the score is less than about 7.9; and wherein the subject is identified as having a moderate pan-tumor gene signature score if the score is from about 7.9 to about 12.6.
[0011] In some aspects, the subject identified as having a high gene signature score is to be administered an 1-0 therapy.
[0012] In some aspects, the subject identified as having a low gene signature score is to be administered an anticancer agent other than an 1-0 therapy.
[0013] In some aspects, the subject identified as having a moderate gene signature score is to be administered an 1-0 therapy in combination with an additional anticancer agent.
[0014] In some aspects, (a) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.05. In some aspects, (a) = about 0.03 to about 0.04. In some aspects, (a) = about 0.03447.
[0015] In some aspects, (b) = about 0.1, about 0.105, about 0.11, about 0.115, about 0.12, about 0.125, about 0.13, about 0.135, about 0.14, about 0.145, or about 0.15. In some aspects, (b) = about 0.13 to about 0.14. In some aspects, (b) = about 0.13825.
[0016] In some aspects, (c) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.05. In some aspects, (c) = about 0.02 to about 0.03. In some aspects, (c) = about 0.02756.
[0017] In some aspects, (d) = about -0.1, about -0.105, about -0.11, about -0.115, about - 0.12, about -0.125, about -0.13, about -0.135, about -0.14, about -0.145, or about -0.15. In some aspects, (d) = about -0.10 to about -0.15. In some aspects, (d) = about -0.13089. [0018] In some aspects, (e) = about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, about 0.08, about 0.085, or about 0.09. In some aspects, (e) = about 0.06 to about 0.07. In some aspects, (e) = about 0.06368. [0019] In some aspects, (f) = about 0.35, about 0.36, about 0.37, about 0.38, about 0.39, about 0.40, about 0.41, about 0.42, about 0.43, about 0.44, or about 0.45. In some aspects, (f) = about 0.39 to about 0.40. In some aspects, (f) = about 0.39185.
[0020] In some aspects, (g) = about 0.07, about 0.075, about 0.08, about 0.085, about 0.09, about 0.095, about 0.10, about 0.105, about 0.11, about 0.115, about 0.12, about 0.125, or about 0.13. In some aspects, (g) = about 0.10 to about 0.11. In some aspects, (g) = about 0.10976. [0021] In some aspects, (h) = about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.08. In some aspects, (h) = about 0.04 to about 0.05. In some aspects, (h) = about 0.04913. [0022] In some aspects, (i) = about -0.01, about -0.015, about -0.02, about -0.025, about - 0.03, about -0.035, about -0.04, about -0.045, or about -0.05. In some aspects, (i) = about -0.02 to about -0.03. In some aspects, (i) = about -0.02537.
[0023] In some aspects, (j) = about -0.0005, about -0.00055, about -0.0006, about - 0.00065, about -0.0007, about -0.00075, about -0.0008, about -0.00085, about -0.0009, about - 0.00095, or about -0.0010. In some aspects, (j) = about -0.0008 to about -0.0009. In some aspects, (j) = about -0.00081.
[0024] In some aspects, (k) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, or about 0.06. In some aspects, (k) = about 0.03 to about 0.04. In some aspects, (k) = about 0.03654.
[0025] In some aspects, (1) = about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.8. In some aspects, (1) = about 0.04 to about 0.05. In some aspects, (1) = about 0.04797.
[0026] In some aspects, (m) = about -0.008, about -0.010, about -0.015, about -0.020, about -0.025, about -0.030, about -0.035, or about -0.0400. In some aspects, (m) = about -0.01 to about -0.02. In some aspects, (m) = about -0.01751.
[0027] In some aspects, (n) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.050. In some aspects, (n) = about 0.02 to about 0.03. In some aspects, (n) = about 0.02254. [0028] In some aspects, (o) = about -0.03, about -0.035, about -0.04, about -0.045, about - 0.05, about -0.055, about -0.06, about -0.065, about -0.07, about -0.075, or about -0.08. In some aspects, (o) = about -0.05 to about -0.06. In some aspects, (o) = about -0.05317.
[0029] In some aspects, (p) = about -0.005, about -0.0055, about -0.006, about -0.0065, about -0.007, about -0.0075, about -0.008, about -0.0085, about -0.009, about -0.0095, or about - 0.010. In some aspects, (p) = about 0.007 to about 0.008. In some aspects, (p) = about -0.00710. [0030] In some aspects, (a) = about 0.03447; (b) = about 0.13825; (c) = about 0.02756;
(d) = about -0.13089; (e) = about 0.06368; (f) = about 0.39185; (g) = about 0.10976; (h) = about 0.04913; (i) = about -0.02537; (j) = about -0.00081; (k) = about 0.03654; (1) = about 0.04797; (m) = about -0.01751; (n) = about 0.02254; (o) = about -0.05317; and (p) = about -0.00710.
[0031] In some aspects, the gene panel further comprises one or more housekeeping genes. In some aspects, the gene panel further comprises one or more housekeeping genes selected from the group consisting of ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof.
[0032] In some aspects, the gene panel further comprises one or more control genes. In some aspects, the gene panel further comprises one or more control genes selected from the group consisting of ANTI, ANT2, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4, PCL- 5, PCL-6, PCL-7, PCL-8, PCL-9, POS1, POS2, POS3, POS4, and any combination thereof.
[0033] In some aspects, the 1-0 therapy comprises an immune checkpoint inhibitor. In some aspects, the 1-0 therapy comprises an anti-PDl/PD-L2 antagonist. In some aspects, the 1-0 therapy comprises an antibody that specifically binds a protein selected from PD-1, PD-L1, LAG-3, TIGIT, TIM3, NKG2a, CSF1R, 0X40, ICOS, MICA, MICB,CD137, KIR, TGFp, IL-10, IL-8, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, or any combination thereof. In some aspects, the 1-0 comprises an anti-PD-1 antibody. In some aspects, the 1-0 therapy comprises an anti -PD -LI antibody.
[0034] In some aspects, the tumor sample is not subjected to CD8 immunohi stochemi stry .
[0035] Some aspects of the present disclosure are directed to a method of identifying a patient in need of an 1-0 therapy, (a) obtaining a tumor sample from the patient; (b) analyzing the expression level of one or more genes in a gene panel selected from CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSF18, and any combination thereof; and (c) determining a pan-tumor gene signature score according to any method disclosed herein. In some aspects, the method further comprises isolating mRNA from the tumor sample prior to analyzing the expression level of the one or more genes.
[0036] Some aspects of the present disclosure are directed to a method of treating a subject in need thereof, comprising: (a) obtaining a tumor sample from the subject; (b) isolating mRNA from the tumor sample; (c) analyzing the expression level of one or more genes in a gene panel selected from CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSF18, and any combination thereof; (d) determining a pan-tumor gene signature score according to any method disclosed herein; and (e) administering to the subject an 1-0 therapy
[0037] In some aspects, the expression level of the one or more genes in the gene panel is analyzed by measuring an mRNA level of the one or more genes in the gene panel in the tumor sample. In some aspects, the expression level is measured using a nuclease protection assay. In some aspects, the expression level is measured using next-generation sequencing. In some aspects, the expression level is measured using reverse transcriptase polymerase chain reaction (RT-PCR).
BRIEF DESCRIPTION OF THE DRAWINGS
[0038] FIGs. 1A-1C are schematic representations, illustrating the methods used to develop a multi-tumor inflammatory signature based on GEP. FIG. ID is a table listing the weighing coefficient for each gene in the CD8 signature gene panel, disclosed herein. FIG. IE is a representative image of immunohistical (IHC) staining of a tumor sample to detect CD8 localization. FIG. IF shows the comparison of the CD8 signature score and the CD8 IHC score (as determined by examining IHC staining of tumor samples). MEL, melanoma; SCCHN, squamous cell carcinoma of the head and neck; + and - indicate the range of CD8 positivity, based on scoring of each sample in the dataset by digital pathology.
[0039] FIG. 2 is a scatter plot, showing CD8 expression by IHC (1) and CD8 signature score (2) across tumor types. CD8 score represents the CD8 IHC or CD8 signature scores. Each box represents the 25%-75% interquartile range (IQR) and the line is the median value. Whiskers represent values 1.5 x the upper and lower limits of the IQR. CRC, colorectal cancer; GC, gastric cancer; HCC, hepatocellular carcinoma; MEL, melanoma; NSCLC, non-small cell lung cancer; OVC, ovarian cancer; PANC, pancreatic cancer; PC, prostate cancer; RCC, renal cell carcinoma; SCCHN, squamous cell carcinoma of the head and neck; SCLC, small cell lung cancer; UC, urothelial carcinoma.
[0040] FIGs. 3A-3L are scatter plots, illustrating the correlation of CD8 IHC and CD8 signature scores across tumor types. MEL, melanoma (FIG. 3A); SCCHN, squamous cell carcinoma of the head and neck (FIG. 3B); UC, urothelial carcinoma (FIG. 3C); CRC, colorectal cancer (FIG. 3D); GC, gastric cancer (FIG. 3E); HCC, hepatocellular carcinoma (FIG. 3F); NSCLC, non-small cell lung cancer (FIG. 3G); OVC, ovarian cancer (FIG. 3H); PANC, pancreatic cancer (FIG. 31); PC, prostate cancer (FIG. 3J); RCC, renal cell carcinoma (FIG. 3K); and SCLC, small cell lung cancer (FIG. 3L).
[0041] FIGs. 4A-4L are graphical representations, showing the prevalence of inflammation by CD8 IHC and CD8 signature scores across tumor types. CD8 score represents the CD8 IHC or CD8 signature scores. UC, urothelial carcinoma (FIG. 4A); CRC, colorectal cancer (FIG. 4B); GC, gastric cancer (FIG. 4C); HCC, hepatocellular carcinoma (FIG. 4D); MEL, melanoma (FIG. 4E); NSCLC, non-small cell lung cancer (FIG. 4F); OVC, ovarian cancer (FIG. 4G); PANC, pancreatic cancer (FIG. 4H); PC, prostate cancer (FIG. 41); RCC, renal cell carcinoma (FIG. 4J); SCCHN, squamous cell carcinoma of the head and neck (FIG. 4K); and SCLC, small cell lung cancer (FIG. 4L).
[0042] FIG. 5 is a scatter plot, illustrating the precision of the CD8 signature, and showing that the precision improves with increasing score across multiple tumor types. Precision of CD8 signature scores was assessed using 8 samples with 15 replicates per tumor type. CRC, colorectal cancer; GC, gastric cancer; HCC, hepatocellular carcinoma; MEL, melanoma; NSCLC, non-small cell lung cancer; OVC, ovarian cancer; PANC, pancreatic cancer; PC, prostate cancer; RCC, renal cell carcinoma; SCCHN, squamous cell carcinoma of the head and neck; SCLC, small cell lung cancer; UC, urothelial carcinoma.
[0043] FIGs. 6A-6P are gene-by-gene scatter plots of RNA-seq data compared to target panel signature scores ("1" = IO 360; "2" = Oncomine IRRA; "3" TIP; and "4" = IOv2) for CCL4 (FIG. 6A), CCL5 (FIG. 6B), CD27 (FIG. 6C), CD276 (FIG. 6D), CD3D (FIG. 6E), CD8A (FIG. 6F), CXCL10 (FIG. 6G), CXCL9 (FIG. 6H), CXCR1 (FIG. 61), HLA-DMB (FIG. 6 J), HLA- DRA (FIG. 6K), HLA-DRB1 (FIG. 6L), LGALS9 (FIG. 6M), NKG7 (FIG. 6N), TMEM173 (STING1) (FIG. 60), TNFSF18 and (FIG. 6P). FIG. 6Q is a chart summarizing the correlation with RNA-seq (Pearson's r) for each of the genes of FIGs. 6A-6P as compared to each of the GEP panels (IO 360, Oncomine IRRA, TIP, and IOv2), as indicated. [0044] FIGs. 7A-7C are scatter plots showing the correlation of CD8 signature scores derived using the IO 360 (FIG. 7A), the Oncomine IRRA (FIG. 7B), and the TIP (FIG. 7C) GEP panels as compared to RNA-seq. Pearson’s r is shown for comparisons of CD8 signature scores derived using each gene panel vs RNA-seq, and "n" is the number of commercial samples analyzed (FIGs. 7A-7C).
[0045] FIGs. 8A-8C are scatter plots showing the correlation between gene expression signature score and CD8 immunohistochemistry (H4C) score for the CD8 signature (TIP; FIG. 8A), TCA signature (FIG. 8B), and TIS signature (IO 360; FIG. 8C). FIGs. 8D-8F are scatter plots showing the correlation between gene expression signature score and PDL-1 expression on tumor cells (%) for the CD8 signature (TIP; FIG. 8D), TCA signature (FIG. 8E), and TIS signature (IO 360; FIG. 8F). Pearson's r values are indicated on each graph (FIGs. 8A-8F). FIGs. 8G-8I are box plots showing the distribution of gene signature scores relative to PD-L1 status (negative or positive) for the CD8 signature (TIP; FIG. 8G), TCA signature (FIG. 8H), and TIS signature (IO 360; FIG. 81).
[0046] FIGs. 9A-9X are scatter plots showing correlations between CD8 IHC (FIGs. 9A- 9L) or CD8 signature scores (FIGs. 9M-9X) and PD-L1 expression on tumor cells by IHC. Data are shown for melanoma (FIGs. 9A and 9M), SSCHN (FIGs. 9B and 9N), NSCLC (FIGs. 9C and 90), pancreatic cancer (FIGs. 9D and 9P), gastric cancer (FIGs. 9E and 9Q), RCC (FIGs. 9F and 9R), colorectal cancer (FIGs. 9G and 9S), urothelial cancer (FIGs. 9H and 9T), hepatocellular cancer (FIGs. 91 and 9U), ovarian cancer (FIGs. 9J and 9V), SCLC (FIGs. 9K and 9W), and prostate cancer (FIGs. 9L and 9X). Pearson’s r is shown for each comparison.
[0047] FIG. 10 is a scatter plot showing the correlation between CD8 signature scores and CD8 IHC in UC samples.
[0048] FIGs. 11A-11B are Kaplan-Meier curves showing the association of CD8 signature scores with PFS (FIG. 11 A) and OS (FIG. 11B) with nivolumab in patients with UC, grouped by CD8 signature score tertile, defined as low (< 7.9), medium (7.9-12.6), or high (> 12.6). OS, overall survival; PFS, progression-free survival; UC, urothelial carcinoma.
DETAILED DESCRIPTION OF THE DISCLOSURE
[0049] Certain aspects of the present disclosure are directed to methods of identifying a human subject suitable for an immune-oncology (1-0) therapy, e.g., an anti-PD-l/PD-Ll antagonist therapy. In some aspects, the present disclosure is directed to a method of identifying a human subject suitable for an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist therapy, comprising measuring expression of CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9 (Galectin-9) , NKG7, TM 1X1173 (STING1), and TNFSF18 (GITRL). A gene panel comprising the identified genes of the present disclosure and the gene signature are useful to identify a subject suitable for and/or responsive to an 1-0 therapy, especially in predicting an inflammatory phenotype in the tumor microenvironment (TME) across multiple tumor types. Therefore, in some aspects, the gene panel and its use can replace the inconvenient and burdensome CD8+ immunohistochemistry.
I. Terms
[0050] In order that the present disclosure can be more readily understood, certain terms are first defined. As used in this application, except as otherwise expressly provided herein, each of the following terms shall have the meaning set forth below. Additional definitions are set forth throughout the application.
[0051] It is understood that wherever aspects are described herein with the language "comprising," otherwise analogous aspects described in terms of "consisting of and/or "consisting essentially of are also provided.
[0052] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure is related. For example, the Concise Dictionary of Biomedicine and Molecular Biology, Juo, Pei-Show, 2nd ed., 2002, CRC Press; The Dictionary of Cell and Molecular Biology, 3rd ed., 1999, Academic Press; and the Oxford Dictionary Of Biochemistry And Molecular Biology, Revised, 2000, Oxford University Press, provide one of skill with a general dictionary of many of the terms used in this disclosure.
[0053] Units, prefixes, and symbols are denoted in their Systeme International de Unites (SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Where a range of values is recited, it is to be understood that each intervening real number value, and each fraction thereof, between the recited upper and lower limits of that range is also specifically disclosed, along with each subrange between such values. The upper and lower limits of any range can independently be included in or excluded from the range, and each range where either, neither or both limits are included is also encompassed within the disclosure. Thus, ranges recited herein are understood to be shorthand for all of the values within the range, inclusive of the recited endpoints. For example, a range of 1 to 10 is understood to include any number, combination of numbers, or sub-range from the group consisting of 1, 2, 3, 4, 5, 6, 7, 8, 9, and 10.
[0054] Where a value is explicitly recited, it is to be understood that values which are about the same quantity or amount as the recited value are also within the scope of the disclosure. Where a combination is disclosed, each subcombination of the elements of that combination is also specifically disclosed and is within the scope of the disclosure. Conversely, where different elements or groups of elements are individually disclosed, combinations thereof are also disclosed. Where any element of a disclosure is disclosed as having a plurality of alternatives, examples of that disclosure in which each alternative is excluded singly or in any combination with the other alternatives are also hereby disclosed; more than one element of a disclosure can have such exclusions, and all combinations of elements having such exclusions are hereby disclosed.
[0055] "Administering" refers to the physical introduction of a composition comprising a therapeutic agent to a subject, using any of the various methods and delivery systems known to those skilled in the art. Preferred routes of administration for the immunotherapy, e.g., the anti- PD-1 antibody or the anti-PD-Ll antibody, include intravenous, intramuscular, subcutaneous, intraperitoneal, spinal or other parenteral routes of administration, for example by injection or infusion. The phrase "parenteral administration" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal injection and infusion, as well as in vivo electroporation. Other non-parenteral routes include an oral, topical, epidermal or mucosal route of administration, for example, intranasally, vaginally, rectally, sublingually or topically. Administering can also be performed, for example, once, a plurality of times, and/or over one or more extended periods.
[0056] An "adverse event" (AE) as used herein is any unfavorable and generally unintended or undesirable sign (including an abnormal laboratory finding), symptom, or disease associated with the use of a medical treatment. For example, an adverse event can be associated with activation of the immune system or expansion of immune system cells (e.g., T cells) in response to a treatment. A medical treatment can have one or more associated AEs and each AE can have the same or different level of severity. Reference to methods capable of "altering adverse events" means a treatment regime that decreases the incidence and/or severity of one or more AEs associated with the use of a different treatment regime.
[0057] An "antibody" (Ab) shall include, without limitation, a glycoprotein immunoglobulin which binds specifically to an antigen and comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds, or an antigen-binding portion thereof. Each H chain comprises a heavy chain variable region (abbreviated herein as Vzz) and a heavy chain constant region. The heavy chain constant region comprises three constant domains, Cm, Cm and Cm. Each light chain comprises a light chain variable region (abbreviated herein as Vz) and a light chain constant region. The light chain constant region is comprises one constant domain, CL. The Vzz and Vz regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FRs). Each Vzz and Vz comprises three CDRs and four FRs, arranged from amino-terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies can mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system. Therefore, the term "anti-PD-1 antibody" includes a full antibody having two heavy chains and two light chains that specifically binds to PD-1 and antigen-binding portions of the full antibody. Non limiting examples of the antigen-binding portions are shown elsewhere herein. [0058] An immunoglobulin can derive from any of the commonly known isotypes, including but not limited to IgA, secretory IgA, IgG and IgM. IgG subclasses are also well known to those in the art and include but are not limited to human IgGl, IgG2, IgG3 and IgG4. "Isotype" refers to the antibody class or subclass (e.g., IgM or IgGl) that is encoded by the heavy chain constant region genes. The term "antibody" includes, by way of example, both naturally occurring and non-naturally occurring antibodies; monoclonal and polyclonal antibodies; chimeric and humanized antibodies; human or nonhuman antibodies; wholly synthetic antibodies; and single chain antibodies. A nonhuman antibody can be humanized by recombinant methods to reduce its immunogenicity in man. Where not expressly stated, and unless the context indicates otherwise, the term "antibody" also includes an antigen-binding fragment or an antigenbinding portion of any of the aforementioned immunoglobulins, and includes a monovalent and a divalent fragment or portion, and a single chain antibody. [0059] An "isolated antibody" refers to an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., an isolated antibody that binds specifically to PD-1 is substantially free of antibodies that bind specifically to antigens other than PD-1). An isolated antibody that binds specifically to PD-1 may, however, have cross-reactivity to other antigens, such as PD-1 molecules from different species. Moreover, an isolated antibody can be substantially free of other cellular material and/or chemicals.
[0060] The term "monoclonal antibody" (mAb) refers to a non-naturally occurring preparation of antibody molecules of single molecular composition, i.e., antibody molecules whose primary sequences are essentially identical, and which exhibits a single binding specificity and affinity for a particular epitope. A monoclonal antibody is an example of an isolated antibody. Monoclonal antibodies can be produced by hybridoma, recombinant, transgenic or other techniques known to those skilled in the art.
[0061] A "human antibody" (HuMAb) refers to an antibody having variable regions in which both the framework and CDR regions are derived from human germline immunoglobulin sequences. Furthermore, if the antibody contains a constant region, the constant region also is derived from human germline immunoglobulin sequences. The human antibodies of the disclosure can include amino acid residues not encoded by human germline immunoglobulin sequences e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo). However, the term "human antibody," as used herein, is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species, such as a mouse, have been grafted onto human framework sequences. The terms "human antibody" and "fully human antibody" and are used synonymously.
[0062] A "humanized antibody" refers to an antibody in which some, most or all of the amino acids outside the CDRs of a non-human antibody are replaced with corresponding amino acids derived from human immunoglobulins. In one aspect of a humanized form of an antibody, some, most or all of the amino acids outside the CDRs have been replaced with amino acids from human immunoglobulins, whereas some, most or all amino acids within one or more CDRs are unchanged. Small additions, deletions, insertions, substitutions or modifications of amino acids are permissible as long as they do not abrogate the ability of the antibody to bind to a particular antigen. A "humanized antibody" retains an antigenic specificity similar to that of the original antibody.
[0063] A "chimeric antibody" refers to an antibody in which the variable regions are derived from one species and the constant regions are derived from another species, such as an antibody in which the variable regions are derived from a mouse antibody and the constant regions are derived from a human antibody.
[0064] An "anti-antigen antibody" refers to an antibody that binds specifically to the antigen. For example, an anti-PD-1 antibody binds specifically to PD-1, an anti-PD-Ll antibody binds specifically to PD-L1, and an anti-CTLA-4 antibody binds specifically to CTLA-4.
[0065] An "antigen-binding portion" of an antibody (also called an "antigen-binding fragment") refers to one or more fragments of an antibody that retain the ability to bind specifically to the antigen bound by the whole antibody. It has been shown that the antigenbinding function of an antibody can be performed by fragments of a full-length antibody. Examples of binding fragments encompassed within the term "antigen-binding portion" of an antibody, e.g., an anti- PD-1 antibody or an anti-PD-Ll antibody described herein, include (i) a Fab fragment (fragment from papain cleavage) or a similar monovalent fragment consisting of the VL, VH, LC and CHI domains; (ii) a F(ab')2 fragment (fragment from pepsin cleavage) or a similar bivalent fragment comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CHI domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341 :544-546), which consists of a VH domain; (vi) an isolated complementarity determining region (CDR) and (vii) a combination of two or more isolated CDRs which can optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see, e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883). Such single chain antibodies are also intended to be encompassed within the term "antigen-binding portion" of an antibody. These antibody fragments are obtained using conventional techniques known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. Antigen-binding portions can be produced by recombinant DNA techniques, or by enzymatic or chemical cleavage of intact immunoglobulins.
[0066] Antibodies useful in the methods and compositions described herein include, but are not limited to, antibodies and antigen-binding portions thereof that specifically bind a protein selected from the group consisting of Inducible T cell Co-Stimulator (ICOS), CD137 (4-1BB), CD 134 (0X40), NKG2A, CD27, CD96, Glucocorticoid-Induced TNFR-Related protein (GITR), and Herpes Virus Entry Mediator (HVEM), Programmed Death- 1 (PD-1), Programmed Death Ligand- 1 (PD-L1), Cytotoxic T-Lymphocyte Antigen-4 (CTLA-4), B and T Lymphocyte Attenuator (BTLA), T cell Immunoglobulin and Mucin domain-3 (TIM-3), Lymphocyte Activation Gene-3 (LAG-3), adenosine A2a receptor (A2aR), Killer cell Lectin-like Receptor G1 (KLRG-1), Natural Killer Cell Receptor 2B4 (CD244), CD 160, T cell Immunoreceptor with Ig and ITIM domains (TIGIT), and the receptor for V-domain Ig Suppressor of T cell Activation (VISTA), KIR, TGFp, IL- 10, IL-8, IL-2, B7-H4, Fas ligand, CXCR4, CSF1R, mesothelin, CEACAM-1, CD52, HER2, MICA, MICB, , and any combination thereof.
[0067] A "cancer" refers a broad group of various diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division and growth divide and grow results in the formation of malignant tumors that invade neighboring tissues and can also metastasize to distant parts of the body through the lymphatic system or bloodstream.
[0068] The term "immunotherapy" refers to the treatment of a subject afflicted with, or at risk of contracting or suffering a recurrence of, a disease by a method comprising inducing, enhancing, suppressing or otherwise modifying an immune response. "Treatment" or "therapy" of a subject refers to any type of intervention or process performed on, or the administration of an active agent to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting, slowing down or preventing the onset, progression, development, severity or recurrence of a symptom, complication or condition, or biochemical indicia associated with a disease.
[0069] "Programmed Death-1" (PD-1) refers to an immunoinhibitory receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo, and binds to two ligands, PD-L1 and PD-L2. The term "PD-1" as used herein includes human PD-1 (hPD-1), variants, isoforms, and species homologs of hPD-1, and analogs having at least one common epitope with hPD-1. The complete hPD-1 sequence can be found under GenBank Accession No. U64863.
[0070] "Programmed Death Ligand-1" (PD-L1) is one of two cell surface glycoprotein ligands for PD-1 (the other being PD-L2) that downregulate T cell activation and cytokine secretion upon binding to PD-1. The term "PD-L1" as used herein includes human PD-L1 (hPD- Ll), variants, isoforms, and species homologs of hPD-Ll, and analogs having at least one common epitope with hPD-Ll. The complete hPD-Ll sequence can be found under GenBank Accession No. Q9NZQ7. The human PD-L1 protein is encoded by the human CD274 gene (NCBI Gene lD: 29126). [0071] As used herein, a PD-1 or PD-L1 "inhibitor," refers to any molecule capable of blocking, reducing, or otherwise limiting the interaction between PD-1 and PD-L1 and/or the activity of PD-1 and/or PD-L1. In some aspects, the inhibitor is an antibody or an antigenbinding fragment of an antibody. In other aspects, the inhibitor comprises a small molecule.
[0072] "T-Cell surface glycoprotein CD8 alpha chain" or "CD8A" as used herein refers to an integral membrane glycoprotein that is involved in the immune response and that serves multiple functions in responses against both external and internal offenses. In T-cells, CD8a functions primarily as a co-receptor for MHC class I molecule / peptide complex. CD8A interacts simultaneously with the T-cell receptor (TCR) and the MHC class I proteins presented by antigen presenting cells (APCs). In turn, CD8a recruits the Src kinase LCK to the vicinity of the TCR- CD3 complex. LCK then initiates different intracellular signaling pathways by phosphorylating various substrates ultimately leading to lymphokine production, motility, adhesion and activation of cytotoxic T-lymphocytes (CTLs). This mechanism enables CTLs to recognize and eliminate infected cells and tumor cells. In NK-cells, the presence of CD8A homodimers at the cell surface provides a survival mechanism allowing conjugation and lysis of multiple target cells. CD8A homodimer molecules also promote the survival and differentiation of activated lymphocytes into memory CD8 T-cells. The complete CD8a amino acid sequence can be found under UniProtKB identification number P01732. The human CD8a protein is encoded by the human CD8a gene (NCBI Gene ID: 925).
[0073] "Lymphocyte Activation Gene-3," "LAG3," "LAG-3," or "CD223," as used herein, refers to a type I transmembrane protein that is expressed on the cell surface of activated CD4+ and CD8+ T cells and subsets of NK and dendritic cells. LAG-3 protein is closely related to CD4, which is a co-receptor for T helper cell activation. Both molecules have four extracellular Ig-like domains and require binding to their ligand, major histocompatibility complex (MHC) class II, for their functional activity. LAG-3 protein is only expressed on the cell surface of activated T cells and its cleavage from the cell surface terminates LAG-3 signaling. LAG-3 can also be found as a soluble protein, which does not bind to MHC class II. LAG-3 also plays an important role in promoting regulatory T cell (Treg) activity and in negatively regulating T cell activation and proliferation. Both natural and induced Treg express increased LAG-3, which is required for their maximal suppressive function. The complete human LAG-3 amino acid sequence can be found under UniProtKB identification number Pl 8627. The human LAG-3 protein is encoded by the human LAG3 gene (NCBI Gene ID: 3902). [0074] "Signal transducer and activator of transcription 1-alpha/beta" or "STAT1," as used herein, refers to a signal transducer and transcription activator that mediates cellular responses to interferons (IFNs), cytokine KITLG/SCF, and other cytokines and other growth factors. Following type I IFN (IFN-alpha and IFN-beta) binding to cell surface receptors, signaling via protein kinases leads to activation of Jak kinases (TYK2 and JAK1) and to tyrosine phosphorylation of STAT1 and STAT2. The phosphorylated STATs dimerize and associate with ISGF3G/IRF-9 to form a complex termed ISGF3 transcription factor, which enters the nucleus. ISGF3 binds to the IFN stimulated response element (ISRE) to activate the transcription of IFN- stimulated genes (ISG), which drive the cell in an antiviral state. In response to type II IFN (IFN- gamma), STAT1 is tyrosine- and serine-phosphorylated. It then forms a homodimer termed IFN- gamma-activated factor (GAF), migrates into the nucleus and binds to the IFN gamma activated sequence (GAS) to drive the expression of the target genes, inducing a cellular antiviral state. STAT1 becomes activated in response to KITLG/SCF and KIT signaling. STAT1 may also mediate cellular responses to activated FGFR1, FGFR2, FGFR3, and FGFR4. The complete human STAT1 amino acid sequence can be found under UniProtKB identification number P42224. The human STAT1 protein is encoded by the human STAT1 gene (NCBI Gene ID: 6772).
[0075] "Cytotoxic T-Lymphocyte Antigen-4" (CTLA-4) refers to an immunoinhibitory receptor belonging to the CD28 family. CTLA-4 is expressed exclusively on T cells in vivo, and binds to two ligands, CD80 and CD86 (also called B7-1 and B7-2, respectively). The term "CTLA-4" as used herein includes human CTLA-4 (hCTLA-4), variants, isoforms, and species homologs of hCTLA-4, and analogs having at least one common epitope with hCTLA-4. The complete hCTLA-4 sequence can be found under GenBank Accession No. AAB59385.
[0076] A "subject" includes any human or nonhuman animal. The term "nonhuman animal" includes, but is not limited to, vertebrates such as nonhuman primates, sheep, dogs, and rodents such as mice, rats and guinea pigs. In preferred aspects, the subject is a human. The terms, "subject" and "patient" are used interchangeably herein.
[0077] A "therapeutically effective amount" or "therapeutically effective dosage" of a drug or therapeutic agent is any amount of the drug that, when used alone or in combination with another therapeutic agent, protects a subject against the onset of a disease or promotes disease regression evidenced by a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction. The ability of a therapeutic agent to promote disease regression can be evaluated using a variety of methods known to the skilled practitioner, such as in human subjects during clinical trials, in animal model systems predictive of efficacy in humans, or by assaying the activity of the agent in in vitro assays.
[0078] By way of example, an "anti-cancer agent" promotes cancer regression in a subject. In preferred aspects, a therapeutically effective amount of the drug promotes cancer regression to the point of eliminating the cancer. "Promoting cancer regression" means that administering an effective amount of the drug, alone or in combination with an anti -neoplastic agent, results in a reduction in tumor growth or size, necrosis of the tumor, a decrease in severity of at least one disease symptom, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction. In addition, the terms "effective" and "effectiveness" with regard to a treatment includes both pharmacological effectiveness and physiological safety. Pharmacological effectiveness refers to the ability of the drug to promote cancer regression in the patient. Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (adverse effects) resulting from administration of the drug.
[0079] As used herein, an "immuno-oncology" therapy or an "I-O" therapy refers to a therapy that comprises utilizing an immune response to target and treat a tumor in a subject. As such, as used herein, an 1-0 therapy is a type of anti-cancer therapy. In some aspects, and 1-0 therapy comprises administering an antibody or an antigen-binding fragment thereof to a subject. In some aspects, an 1-0 therapy comprises administering to a subject an immune cell, e.g., a T cell, e.g., a modified T cell, e.g., a T cell modified to express a chimeric antigen receptor or an particular T cell receptor. In some aspects, the 1-0 therapy comprises administering a therapeutic vaccine to a subject. In some aspects, the 1-0 therapy comprises administering a cytokine or a chemokine to a subject. In some aspects, the 1-0 therapy comprises administering an interleukin to a subject. In some aspects, the 1-0 therapy comprises administering an interferon to a subject. In some aspects, the 1-0 therapy comprises administering a colony stimulating factor to a subject. [0080] By way of example for the treatment of tumors, a therapeutically effective amount of an anti-cancer agent preferably inhibits cell growth or tumor growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects. In other preferred aspects of the disclosure, tumor regression can be observed and continue for a period of at least about 20 days, more preferably at least about 40 days, or even more preferably at least about 60 days. Notwithstanding these ultimate measurements of therapeutic effectiveness, evaluation of immunotherapeutic drugs must also make allowance for immune-related response patterns.
[0081] An "immune response" is as understood in the art, and generally refers to a biological response within a vertebrate against foreign agents or abnormal, e.g., cancerous cells, which response protects the organism against these agents and diseases caused by them. An immune response is mediated by the action of one or more cells of the immune system (for example, a T lymphocyte, B lymphocyte, natural killer (NK) cell, macrophage, eosinophil, mast cell, dendritic cell or neutrophil) and soluble macromolecules produced by any of these cells or the liver (including antibodies, cytokines, and complement) that results in selective targeting, binding to, damage to, destruction of, and/or elimination from the vertebrate's body of invading pathogens, cells or tissues infected with pathogens, cancerous or other abnormal cells, or, in cases of autoimmunity or pathological inflammation, normal human cells or tissues. An immune reaction includes, e.g., activation or inhibition of a T cell, e.g., an effector T cell, a Th cell, a CD4+ cell, a CD8+ T cell, or a Treg cell, or activation or inhibition of any other cell of the immune system, e.g., NK cell.
[0082] An "immune-related response pattern" refers to a clinical response pattern often observed in cancer patients treated with immunotherapeutic agents that produce antitumor effects by inducing cancer-specific immune responses or by modifying native immune processes. This response pattern is characterized by a beneficial therapeutic effect that follows an initial increase in tumor burden or the appearance of new lesions, which in the evaluation of traditional chemotherapeutic agents would be classified as disease progression and would be synonymous with drug failure. Accordingly, proper evaluation of immunotherapeutic agents can require longterm monitoring of the effects of these agents on the target disease.
[0083] The terms "treat," "treating," and "treatment," as used herein, refer to any type of intervention or process performed on, or administering an active agent to, the subject with the objective of reversing, alleviating, ameliorating, inhibiting, or slowing down or preventing the progression, development, severity or recurrence of a symptom, complication, condition or biochemical indicia associated with a disease or enhancing overall survival. Treatment can be of a subject having a disease or a subject who does not have a disease (e.g., for prophylaxis).
[0084] The term "effective dose" or "effective dosage" is defined as an amount sufficient to achieve or at least partially achieve a desired effect. A "therapeutically effective amount" or "therapeutically effective dosage" of a drug or therapeutic agent is any amount of the drug that, when used alone or in combination with another therapeutic agent, promotes disease regression evidenced by a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, an increase in overall survival (the length of time from either the date of diagnosis or the start of treatment for a disease, such as cancer, that patients diagnosed with the disease are still alive), or a prevention of impairment or disability due to the disease affliction. A therapeutically effective amount or dosage of a drug includes a "prophylactically effective amount" or a "prophylactically effective dosage", which is any amount of the drug that, when administered alone or in combination with another therapeutic agent to a subject at risk of developing a disease or of suffering a recurrence of disease, inhibits the development or recurrence of the disease. The ability of a therapeutic agent to promote disease regression or inhibit the development or recurrence of the disease can be evaluated using a variety of methods known to the skilled practitioner, such as in human subjects during clinical trials, in animal model systems predictive of efficacy in humans, or by assaying the activity of the agent in in vitro assays.
[0085] By way of example, an anti-cancer agent is a drug that promotes cancer regression in a subject. In some aspects, a therapeutically effective amount of the drug promotes cancer regression to the point of eliminating the cancer. "Promoting cancer regression" means that administering an effective amount of the drug, alone or in combination with an antineoplastic agent, results in a reduction in tumor growth or size, necrosis of the tumor, a decrease in severity of at least one disease symptom, an increase in frequency and duration of disease symptom-free periods, an increase in overall survival, a prevention of impairment or disability due to the disease affliction, or otherwise amelioration of disease symptoms in the patient. In addition, the terms "effective" and "effectiveness" with regard to a treatment includes both pharmacological effectiveness and physiological safety. Pharmacological effectiveness refers to the ability of the drug to promote cancer regression in the patient. Physiological safety refers to the level of toxicity, or other adverse physiological effects at the cellular, organ and/or organism level (adverse effects) resulting from administration of the drug.
[0086] By way of example for the treatment of tumors, a therapeutically effective amount or dosage of the drug inhibits cell growth or tumor growth by at least about 20%, by at least about 40%, by at least about 60%, or by at least about 80% relative to untreated subjects. In some aspects, a therapeutically effective amount or dosage of the drug completely inhibits cell growth or tumor growth, i.e., inhibits cell growth or tumor growth by 100%. The ability of a compound to inhibit tumor growth can be evaluated using an assay described herein. Alternatively, this property of a composition can be evaluated by examining the ability of the compound to inhibit cell growth, such inhibition can be measured in vitro by assays known to the skilled practitioner. In some aspects described herein, tumor regression can be observed and continue for a period of at least about 20 days, at least about 40 days, or at least about 60 days.
[0087] A "cancer gene panel," "hereditary cancer panel," "comprehensive cancer panel," or "multigene cancer panel," as used herein, refers to a method of sequencing a subset of targeted cancer genes, including coding regions, introns, promoters, and/or mRNA transcripts. In some aspects, the CGP comprises sequencing at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, or at least about 50 targeted cancer genes.
[0088] The term "microsatellite instability" or "MSI" refers to a change that occurs in the DNA of certain cells (such as tumor cells) in which the number of repeats of microsatellites (short, repeated sequences of DNA) is different than the number of repeats that was in the DNA when it was inherited. MSI can be high microsatellite instability (MSI-H) or low microsatellite instability (MSI-L). Microsatellites are short tandem DNA repeat sequences of 1-6 bases. These are prone to DNA replication errors, which are repaired by mismatch repair (MMR). Hence microsatellites are good indicators of genome instability, especially deficient mismatch repair (dMMR). MSI is usually diagnosed by screening 5 microsatellite markers (BAT-25, BAT-26, NR21, NR24, and NR27). MSI-H represents the presence of at least 2 unstable markers among 5 microsatellite markers analyzed (or >30% of the markers if a larger panel is used). MSI-L means instability of 1 MSI marker (or 10%-30% of markers in larger panels). MSS means the absence of an unstable microsatellite marker.
[0089] The term "biological sample" as used herein refers to biological material isolated from a subject. The biological sample can contain any biological material suitable for determining target gene expression, for example, by sequencing nucleic acids in the tumor (or circulating tumor cells) and identifying a genomic alteration in the sequenced nucleic acids. The biological sample can be any suitable biological tissue or fluid such as, for example, tumor tissue, blood, blood plasma, and serum. In one aspect, the sample is a tumor sample. In some aspects, the tumor sample can be obtained from a tumor tissue biopsy, e.g., a formalin-fixed, paraffin- embedded (FFPE) tumor tissue or a fresh-frozen tumor tissue or the like. In another aspect, the biological sample is a liquid biopsy that, in some aspects, comprises one or more of blood, serum, plasma, circulating tumor cells, exoRNA, ctDNA, and cfDNA.
[0090] A "tumor sample," as used herein, refers to a biological sample that comprises tumor tissue. In some aspects, a tumor sample is a tumor biopsy. In some aspects, a tumor sample comprises tumor cells and one or more non-tumor cell present in the tumor microenvironment (TME). For the purposes of the present disclosure, the TME is made up of at least two regions. The tumor "parenchyma" is a region of the TME that includes predominantly tumor cells, e.g., the part (or parts) of the TME that includes the bulk of the tumor cells. The tumor parenchyma does not necessarily consist of only tumor cells, rather other cells such as stromal cells and/or lymphocytes can also be present in the parenchyma. The "stromal" region of the TME includes the adjacent non-tumor cells. In some aspects, the tumor sample comprises all or part of the tumor parenchyma and one or more cells of the stroma. In some aspects, the tumor sample is obtained from the parenchyma. In some aspects the tumor sample is obtained from the stroma. In other aspects, the tumor sample is obtained from the parenchyma and the stroma.
[0091] The use of the alternative (e.g., "or") should be understood to mean either one, both, or any combination thereof of the alternatives. As used herein, the indefinite articles "a" or "an" should be understood to refer to "one or more" of any recited or enumerated component.
[0092] The terms "about" or "comprising essentially of' refer to a value or composition that is within an acceptable error range for the particular value or composition as determined by one of ordinary skill in the art, which will depend in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, "about" or "comprising essentially of' can mean within 1 or more than 1 standard deviation per the practice in the art. Alternatively, "about" or "comprising essentially of' can mean a range of up to 10%. Furthermore, particularly with respect to biological systems or processes, the terms can mean up to an order of magnitude or up to 5-fold of a value. When particular values or compositions are provided in the application and claims, unless otherwise stated, the meaning of "about" or "comprising essentially of' should be assumed to be within an acceptable error range for that particular value or composition.
[0093] As described herein, any concentration range, percentage range, ratio range or real number range is to be understood to include the value of any real number within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an real number), unless otherwise indicated.
[0094] Various aspects of the disclosure are described in further detail in the following subsections.
II. Methods of the Disclosure [0095] Some aspects of the present disclosure are directed to methods of identifying a human subject suitable for an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist therapy, comprising calculating a pan-tumor gene signature score for a tumor sample obtained from the subject. Some aspects of the present disclosure are directed to methods of treating a tumor in a subject in need thereof, comprising administering an 1-0 therapy to a subject that has been identidfied as having a high pan-tumor gene signature score. Some aspects of the present disclosure are directed to methods of treating a tumor in a subject in need thereof, comprising administering an anti-cancer therapy to a subject that has been identidfied as having a low pantumor gene signature score, wherein the anti-cancer therapy does not comprise an 1-0 therapy. Some aspects of the present disclosure are directed to methods of treating a tumor in a subject in need thereof, comprising administering an 1-0 therapy in combination with an additional anticancer agent to a subject that has been identidfied as having a moderate pan-tumor gene signature score.
[0096] Inflammation in the TME can be an indicator of potential responsiveness to an 1-0 therapy. However, contemporary methods for measuring inflammation in a tumor require the laborious process of immunohistochemistry to detect and analyze CD8 expression in a tumor biopsy. It was surprisingly found that the expression pattern of a relatively small number of genes (in some aspects, at least about 16 genes) correlates with inflammation in a tumor microenvironment. In some aspects, the methods described herein can replace the need for timeconsuming IHC. In some aspects the measuring is conducted in vitro.
[0097] In some aspects, the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18. In some aspects, the pan tumor gene signature score is determined according to the following formula (I): pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p)
(I).
[0098] Some aspects of the present disclosure are directed to a pan-tumor gene panel for use in a method of identifying a subject suitable for an 1-0 therapy comprising calculating a pan- tumor gene signature score for a tumor sample obtained from the subject, wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the pantumor gene panel in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying Formula 1. In some aspects, the subject is identified as being suitable for an 1-0 therapy if the subject has a high pan-tumor gene signature score.
[0099] Some aspects of the present disclosure are directed to a pan-tumor gene panel for use in a method of stratifying a subject in need of an 1-0 therapy, comprising determining a pantumor gene signature score for a tumor sample obtained from the subject,
[0100] wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA- DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying Formula 1 to the measured expression values. In some aspects, the subject is stratified as either having a high pan-tumor gene signature score, a moderate pan-tumor gene signature score, or a low pan-tumor gene signature score.
[0101] In some aspects, (a) is any real number between 0.01 and 0.05. In some aspects, (b) is any real number between 0.10 and 0.15. In some aspects, (c) is any real number between 0.01 and 0.05. In some aspects, (d) is any real number between -0.10 and -0.15. In some aspects, (e) is any real number between 0.03 and 0.09. In some aspects, (f) is any real number between 0.35 and 0.45. In some aspects, (g) is any real number between 0.07 and 0.13. In some aspects, (h) is any real number between 0.02 and 0.08. In some aspects, (i) is any real number between - 0.01 and -0.05. In some aspects, (j) is any real number between -0.0001 and -0.0010. In some aspects, (k) is any real number between 0.01 and 0.06. In some aspects, (1) is any real number between 0.02 and 0.08. In some aspects, (m) is any real number between -0.008 and -0.040. In some aspects, (n) is any real number between 0.01 and 0.050. In some aspects, (o) is any real number between -0.03 and -0.080. In some aspects, (p) is any real number between -0.005 and - 0.010. In some aspects, (a) is any real number between 0.01 and 0.05; (b) is any real number between 0.10 and 0.15; (c) is any real number between 0.01 and 0.05; (d) is any real number between -0.10 and -0.15; (e) is any real number between 0.03 and 0.09; (f) is any real number between 0.35 and 0.45; (g) is any real number between 0.07 and 0.13; (h) is any real number between 0.02 and 0.08; (i) is any real number between -0.01 and -0.05; (j) is any real number between -0.0001 and -0.0010; (k) is any real number between 0.01 and 0.06; (1) is any real number between 0.02 and 0.08; (m) is any real number between -0.008 and -0.040; (n) is any real number between 0.01 and 0.050; (o) is any real number between -0.03 and -0.080; and (p) is any real number between -0.005 and -0.010.
[0102] In some aspects, (a) is about 0.01, about 0.015, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.026, about 0.027, about 0.028, about 0.029, about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, about 0.04, about 0.041, about 0.042, about 0.043, about 0.044, about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, or about 0.05. In some aspects, (a) is about 0.02 to about 0.05, about 0.02 to about 0.04, about 0.03 to about 0.05, or about 0.03 to about 0.04. In some aspects, (a) is about 0.03. In some aspects, (a) is about 0.031. In some aspects, (a) is about 0.032. In some aspects, (a) is about 0.033. In some aspects, (a) is about 0.034. In some aspects, (a) is about 0.035. In some aspects, (a) is about 0.036. In some aspects, (a) is about 0.037. In some aspects, (a) is about 0.038. In some aspects, (a) is about 0.039. In some aspects, (a) is about 0.04. In some aspects, (a) is about 0.041. In some aspects, (a) is about 0.042. In some aspects, (a) is about 0.043. In some aspects, (a) is about 0.044. In some aspects,
(a) is about 0.045. In some aspects, (a) is about 0.046. In some aspects, (a) is about 0.047. In some aspects, (a) is about 0.048. In some aspects, (a) is about 0.049. In some aspects, (a) is about 0.05. In certain aspects, (a) is about 0.03447.
[0103] In some aspects, (b) is about 0.1, about 0.105, about 0.11, about 0.115, about 0.12, about 0.125, about 0.13, about 0.131, about 0.132, about 0.133, about 0.134, about 0.135, about 0.136, about 0.137, about 0.138, about 0.139, about 0.14, about 0.141, about 0.142, about 0.143, about 0.144, about 0.145, about 0.146, about 0.147, about 0.148, about 0.149, or about 0.15. In some aspects, (b) is about 0.11 to about 0.15, about 0.11 to about 0.14, about 0.12 to about 0.15, about 0.12 to about 0.14, or about 0.13 to about 0.14. In some aspects, (b) is about 0.131. In some aspects, (b) is about 0.132. In some aspects, (b) is about 0.133. In some aspects, (b) is about 0.134. In some aspects, (b) is about 0.135. In some aspects, (b) is about 0.136. In some aspects,
(b) is about 0.137. In some aspects, (b) is about 0.138. In some aspects, (b) is about 0.139. In some aspects, (b) is about 0.14. In certain aspects, (b) is about 0.13825.
[0104] In some aspects, (c) is about 0.01, about 0.015, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.026, about 0.027, about 0.028, about 0.029, about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, about 0.04, about 0.045, or about 0.05. In some aspects (c) is about 0.01 to about 0.05, about 0.01 to about 0.04, about 0.01 to about 0.03, about 0.02 to about 0.05, about 0.02 to about 0.04, or about 0.02 to about 0.03. In some aspects, (c) is about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.026, about 0.027, about 0.028, about 0.029, about 0.03. In certain aspects (c) is about 0.02756.
[0105] In some aspects, (d) is about -0.05, about -0.1, about -0.105, about -0.11, about - 0.115, about -0.12, about -0.125, about -0.126, about -0.127, about -0.128, about -0.129, about - 0.13, about -0.131, about -0.132, about -0.133, about -0.134, about -0.135, about -0.14, about - 0.145, or about -0.15. In some aspects, (d) is about -0.10 to about -0.15, about -0.10 to about - 0.14, about -0.10 to about -0.13, about -0.11 to about -0.15, about -0.11 to about -0.14, about - 0.11 to about -0.13, about -0.12 to about -0.15, about -0.12 to about -0.14, about -0.12 to about - 0.13, about -0.13 to about -0.15, about -0.13 to about -0.14, about -0.13 to about -0.139, about - 0.13 to about -0.138, about -0.13 to about -0.137, about -0.13 to about -0.136, about -0.13 to about -0.135, about -0.13 to about -0.134, about -0.13 to about -0.133, about -0.13 to about - 0.132, or about -0.13 to about -0.131. In certain aspects, (d) is about -0.13089.
[0106] In some aspects, (e) is about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.061, about 0.062, about 0.063, about 0.064, about 0.065, about 0.066, about 0.067, about 0.068, about 0.069, about 0.07, about 0.075, about 0.08, about 0.085, or about 0.09. In some aspects, (e) is about 0.04 to about 0.09, about 0.04 to about 0.08, about 0.05 to about 0.09, about 0.05 to about 0.08, about 0.06 to about 0.09, about 0.06 to about 0.08, about 0.06 to about 0.07, about 0.06 to about 0.069, about 0.06 to about 0.068, about 0.06 to about 0.067, about 0.06 to about 0.066, about 0.06 to about 0.065, about 0.06 to about 0.064, about 0.061 to about 0.069, about 0.061 to about 0.068, about 0.061 to about 0.067, about 0.061 to about 0.066, about 0.061 to about 0.065, about 0.061 to about 0.064, about 0.062 to about 0.069, about 0.062 to about 0.068, about 0.062 to about 0.067, about 0.062 to about 0.066, about 0.062 to about 0.065, about 0.062 to about 0.064, about 0.063 to about 0.069, about 0.063 to about 0.068, about 0.063 to about 0.067, about 0.063 to about 0.066, about 0.063 to about 0.065, or about 0.063 to about 0.064. In some aspects, (e) is about 0.06368.
[0107] In some aspects, (f) is about 0.35, about 0.36, about 0.37, about 0.38, about 0.385, about 0.386, about 0.387, about 0.388, about 0.389, about 0.39, about 0.391, about 0.392, about 0.393, about 0.394, about 0.395, about 0.396, about 0.397, about 0.398, about 0.399, about 0.40, about 0.41, about 0.42, about 0.43, about 0.44, or about 0.45. In some aspects, (f) is about 0.37 to about 0.42, about 0.37 to about 0.41, about 0.37 to about 0.40, about 0.38 to about 0.42, about 0.38 to about 0.41, about 0.38 to about 0.40, about 0.39 to about 0.42, about 0.39 to about 0.41, about 0.39 to about 0.40, about 0.39 to about 0.399, about 0.39 to about 0.398, about 0.39 to about 0.397, about 0.39 to about 0.396, about 0.39 to about 0.395, about 0.39 to about 0.394, about 0.39 to about 0.393, about 0.39 to about 0.392, or about 0.391 to about 0.392. In certain aspects, (f) is about 0.39185.
[0108] In some aspects, (g) is about 0.07, about 0.075, about 0.08, about 0.085, about 0.09, about 0.095, about 0.10, about 0.105, about 0.106, about 0.107, about 0.108, about 0.109, about 0.11, about 0.112, about 0.113, about 0.114, about 0.115, about 0.12, about 0.125, or about 0.13. In some aspects, (g) is about 0.08 to about 0.13, about 0.08 to about 0.12, about 0.08 to about 0.11, about 0.09 to about 0.13, about 0.09 to about 0.12, about 0.09 to about 0.11, about 0.09 to about 0.13, about 0.10 to about 0.13, about 0.10 to about 0.12, about 0.10 to about 0.11, about 0.105 to about 0.115, about 0.105 to about 0.115. In some aspects, (g) is about 0.105, about 0.106, about 0.107, about 0.108, about 0.109, about 0.11, about 0.112, about 0.113, about 0.114, or about 0.115. In certain aspects, (g) is about 0.10976.
[0109] In some aspects, (h) is about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.08. In some aspects, (h) is about 0.02 to about 0.07, about 0.02 to about 0.06, about 0.02 to about 0.05, about 0.03 to about 0.07, about 0.03 to about 0.06, about 0.03 to about 0.05, about 0.04 to about 0.07, about 0.04 to about 0.06, about 0.04 to about 0.055, about 0.04 to about 0.05, about 0.045 to about 0.055, about 0.045 to about 0.054, about 0.045 to about 0.053, about 0.045 to about 0.052, about 0.045 to about 0.051, about 0.045 to about 0.050, about 0.045 to about 0.049, about 0.046 to about 0.054, about 0.047 to about 0.053, about 0.048 to about 0.052, about 0.049 to about 0.051, or about 0.049 to about 0.050. In some aspects, (h) is about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, or about 0.055. In certain aspects, (h) is about 0.04913.
[0110] In some aspects, (i) is about -0.01, about -0.015, about -0.02, about -0.021, about - 0.022, about -0.023, about -0.024, about -0.025, about -0.026, about -0.027, about -0.028, about - 0.029, about -0.03, about -0.035, about -0.04, about -0.045, or about -0.05. In some aspects, (i) is about -0.02 to about -0.05, about -0.02 to about -0.04, about -0.02 to about -0.03, about -0.02 to about -0.029, about -0.02 to about -0.028, about -0.02 to about -0.027, about -0.02 to about - 0.026, about -0.021 to about -0.029, about -0.021 to about -0.028, about -0.021 to about -0.027, about -0.021 to about -0.026, about -0.022 to about -0.029, about -0.022 to about -0.028, about - 0.022 to about -0.027, about -0.022 to about -0.026, about -0.023 to about -0.029, about -0.023 to about -0.028, about -0.023 to about -0.027, about -0.023 to about -0.026, about -0.024 to about - 0.029, about -0.024 to about -0.028, about -0.024 to about -0.027, about -0.024 to about -0.026, about -0.025 to about -0.029, about -0.025 to about -0.028, about -0.025 to about -0.027, or about -0.025 to about -0.026. In some aspects, (i) is about -0.02, about -0.021, about -0.022, about - 0.023, about -0.024, about -0.025, about -0.026, about -0.027, about -0.028, or about -0.029. In certain aspects, (i) is about -0.02537.
[0111] In some aspects, (j) is about -0.0005, about -0.00055, about -0.0006, about - 0.00065, about -0.0007, about -0.00075, about -0.000756, about -0.000757, about -0.000758, about -0.000759, about -0.0008, about -0.00081, about -0.00082, about -0.00083, about -0.00084, about -0.00085, about -0.0009, about -0.00095, or about -0.0010. In some aspects, (j) is about -
0.0006 to about -0.001, about -0.0006 to about -0.0009, about -0.0006 to about -0.0008, about -
0.0007 to about -0.001, about -0.0007 to about -0.0009, about -0.0007 to about -0.0008, about -
0.0008 to about -0.001, about -0.0008 to about -0.0009. In some aspects, (j) is about -0.00075, about -0.000756, about -0.000757, about -0.000758, about -0.000759, about -0.0008, about - 0.00081, about -0.00082, about -0.00083, about -0.00084, or about -0.00085. In certain aspects, (j) = about -0.00081.
[0112] In some aspects, (k) is about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, about 0.04, about 0.045, about 0.05, about 0.055, or about 0.06.
[0113] In some aspects, (k) is about 0.01 to about 0.06, about 0.01 to about 0.05, about 0.01 to about 0.04, about 0.02 to about 0.06, about 0.02 to about 0.05, about 0.02 to about 0.04, about 0.03 to about 0.06, about 0.03 to about 0.05, about 0.03 to about 0.04, about 0.031 to about 0.039, about 0.032 to about 0.038, about 0.033 to about 0.037, about 0.034 to about 0.037, about 0.035 to about 0.037, or about 0.036 to about 0.037. In some aspects, (k) is about 0.03, about 0.031, about 0.032, about 0.033, about 0.034, about 0.035, about 0.036, about 0.037, about 0.038, about 0.039, or about 0.04. In certain aspects, (k) is about 0.03654.
[0114] In some aspects, (1) is about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.8.
[0115] In some aspects, (1) is about 0.02 to about 0.07, about 0.02 to about 0.06, about 0.02 to about 0.05, about 0.03 to about 0.07, about 0.03 to about 0.06, about 0.03 to about 0.05, about 0.04 to about 0.07, about 0.04 to about 0.06, about 0.04 to about 0.05, about 0.045 to about 0.055, about 0.046 to about 0.054, about 0.047 to about 0.053, about 0.047 to about 0.052, about 0.047 to about 0.051, about 0.047 to about 0.050, about 0.047 to about 0.049, or about 0.047 to about 0.048. In some aspects, (1) is about 0.045, about 0.046, about 0.047, about 0.048, about 0.049, about 0.05, about 0.051, about 0.052, about 0.053, about 0.054, or about 0.055. In certain aspects, (1) is about 0.04797.
[0116] In some aspects, (m) is about -0.008, about -0.010, about -0.015, about -0.016, about -0.017, about -0.018, about -0.019, about -0.020, about -0.021, about -0.022, about -0.023, about -0.024, about -0.025, about -0.030, about -0.035, or about -0.0400. In some aspects, (m) is about -0.05 to about -0.03, about -0.05 to about -0.025, about -0.01 to about -0.03, about -0.01 to about -0.025, about -0.01 to about -0.02, about -0.01 to about -0.019, about -0.01 to about -0.018, about -0.01 to about -0.017, about -0.015 to about -0.02, about -0.015 to about -0.019, about - 0.015 to about -0.018, about -0.015 to about -0.017, about -0.016 to about -0.02, about -0.016 to about -0.019, about -0.016 to about -0.018, about -0.016 to about -0.017, about -0.017 to about - 0.02, about -0.017 to about -0.019, or about -0.017 to about -0.018. In some aspects, (m) is about -0.015, about -0.016, about -0.017, about -0.018, about -0.019, about -0.020, about -0.021, about -0.022, about -0.023, about -0.024, or about -0.025. In certain aspects, (m) is about -0.01751.
[0117] In some aspects, (n) is about 0.01, about 0.015, about 0.016, about 0.017, about 0.018, about 0.019, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.050.
[0118] In some aspects, (n) is about 0.01 to about 0.05, about 0.01 to about 0.04, about 0.01 to about 0.03, about 0.015 to about 0.05, about 0.015 to about 0.04, about 0.015 to about 0.03, about 0.02 to about 0.05, about 0.02 to about 0.04, about 0.02 to about 0.03, about 0.02 to about 0.025, about 0.021 to about 0.024, or about 0.022 to about 0.023. In some aspects, (n) is about 0.015, about 0.016, about 0.017, about 0.018, about 0.019, about 0.02, about 0.021, about 0.022, about 0.023, about 0.024, or about 0.025. In certain aspects, (n) is about 0.02254.
[0119] In some aspects, (o) is about -0.03, about -0.035, about -0.04, about -0.045, about -0.05, about -0.051, about -0.052, about -0.053, about -0.054, about -0.055, about -0.056, about - 0.057, about -0.058, about -0.059, about -0.06, about -0.065, about -0.07, about -0.075, or about - 0.08.
[0120] In some aspects, (o) is about -0.03 to about -0.08, about -0.03 to about -0.07, about -0.03 to about -0.06, about -0.04 to about -0.08, about -0.04 to about -0.07, about -0.04 to about -0.06, about -0.05 to about -0.08, about -0.05 to about -0.07, about -0.05 to about -0.06, about -0.051 to about -0.059, about -0.052 to about -0.058, about -0.053 to about -0.059, about - 0.053 to about -0.058, about -0.053 to about -0.057, about -0.053 to about -0.056, about -0.053 to about -0.055, or about -0.053 to about -0.054. In some aspects, (o) is about -0.045, about -0.05, about -0.051, about -0.052, about -0.053, about -0.054, about -0.055, about -0.056, about -0.057, about -0.058, about -0.059, or about -0.06. In certain aspects, (o) is about -0.05317.
[0121] In some aspects, (p) is about -0.005, about -0.0055, about -0.006, about -0.0065, about -0.0066, about -0.0067, about -0.0068, about -0.0069, about -0.007, about -0.0071, about - 0.0072, about -0.0073, about -0.0074, about -0.0075, about -0.008, about -0.0085, about -0.009, about -0.0095, or about -0.010.
[0122] In some aspects, (p) is about 0.005 to about 0.010, about 0.005 to about 0.009, about 0.005 to about 0.008, about 0.006 to about 0.010, about 0.006 to about 0.009, about 0.006 to about 0.008, about 0.007 to about 0.010, about 0.007 to about 0.009, about 0.007 to about 0.008, about 0.0065 to about 0.0075, about 0.0066 to about 0.0075, about 0.0067 to about 0.0075, about 0.0068 to about 0.0074, about 0.0069 to about 0.0073, about 0.0070 to about 0.0072, about 0.0071 to about 0.0072, or about 0.0070 to about 0.0071. In some aspects, (p) is about -0.0065, about -0.0066, about -0.0067, about -0.0068, about -0.0069, about -0.007, about - 0.0071, about -0.0072, about -0.0073, about -0.0074, or about -0.0075. In certain aspects, (p) is about -0.00710.
[0123] In some aspects, (a) is about 0.03447; (b) is about 0.13825; (c) is about 0.02756; (d) is about -0.13089; (e) is about 0.06368; (f) is about 0.39185; (g) is about 0.10976; (h) is about 0.04913; (i) is about -0.02537; (j) is about -0.00081; (k) is about 0.03654; (1) is about 0.04797; (m) is about -0.01751; (n) is about 0.02254; (o) is about -0.05317; and (p) is about -0.00710.
[0124] In some aspects, the methods disclosed herein comprise administering an 1-0 therapy to a subject identified as having a high pan-tumor gene signature score. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12, greater than about 12.1, greater than about 12.2, greater than about 12.3, greater than about 12.4, greater than about 12.5, greater than about 12.6, greater than about 12.7, greater than about 12.8, greater than about 12.9, greater than about 13, greater than about 14, greater than about 15, greater than about 16, greater than about 17, greater than about 18, greater than about 19, greater than about 20. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.1. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.2. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.3. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.4. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.5. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.6. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.7. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.8. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 12.9. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 13. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 14. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 15. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 16. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 17. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 18. In some aspects, a high pan-tumor gene signature score is characterized by a score of greater than about 19. In some aspects, a high pantumor gene signature score is characterized by a score of greater than about 20.
[0125] In some aspects, the methods disclosed herein comprise administering an 1-0 therapy in combination with an additional anticancer agent to a subject identified as having a moderate pan-tumor gene signature score. In some aspects, a moderate pan-tumor gene signature score is characterized by a score from about 7.0 to about 13.5, about 7.1 to about 13.4, about 7.2 to about 13.3, about 7.3 to about 13.2, about 7.4 to about 13.1, about 7.5 to about 13.0, about 7.6 to about 12.9, about 7.8 to about 12.8, about 7.9 to about 12.7, or about 7.9 to about 12.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score from about 7.9 to about 12.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.5. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 8.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 8.9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 9.9. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 10. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 10.9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.1. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 11.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.4. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.6. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.7. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.8. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 11.9. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.1. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.2. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.3. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.4. In some aspects, a moderate pantumor gene signature score is characterized by a score of about 12.5. In some aspects, a moderate pan-tumor gene signature score is characterized by a score of about 12.6.
[0126] In some aspects, the methods disclosed herein comprise administering an anticancer therapy to a subject identified as having a low pan-tumor gene signature score, wherein the anticancer therapy does not comprise an 1-0 therapy. In some aspects, a low pantumor gene signature score is characterized by a score of less than about 12, less than about 12.1, less than about 11, less than about 10, less than about 9, less than about 8, less than about 7.9, less than about 7.8, less than about 7.7, less than about 7.6, less than about 7.5, less than about
7.4, less than about 7.3, less than about 7.2, less than about 7.1, less than about 7.0, less than about 6.9, less than about 6.8, less than about 6.7, less than about 6.6, less than about 6.5, less than about 6.4, less than about 6.3, less than about 6.2, less than about 6.1, less than about 6.0, less than about 5.9, less than about 5.8, less than about 5.7, less than about 5.6, less than about
5.5, less than about 5.4, less than about 5.3, less than about 5.2, less than about 5.1, or less than about 5.0. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 8. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.9. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.8. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.6. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 17.5. In some aspects, a low pantumor gene signature score is characterized by a score of less than about 7.4. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.3. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.2. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7.1. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 7. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.9. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.8. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.7. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.6. In some aspects, a low pantumor gene signature score is characterized by a score of less than about 6.5. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.4. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.3. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.2. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.1. In some aspects, a low pan-tumor gene signature score is characterized by a score of less than about 6.
[0127] In some aspects, the gene panel further comprises one or more additional genes. In some aspects, the gene panel comprises at least 2 to at least about 100 genes. In some aspects, the gene panel comprises at least 2 to at least about 95, at least 2 to at least about 90, at least 2 to at least about 85, at least 2 to at least about 80, at least 2 to at least about 75, at least 2 to at least about 70, at least 2 to at least about 65, at least 2 to at least about 60, at least 2 to at least about 55, at least 2 to at least about 50, at least 2 to at least about 45, at least 2 to at least about 40, at least 2 to at least about 35, at least 2 to at least about 30, at least 2 to at least about 25, at least 2 to at least about 20, at least 2 to at least about 15, at least 2 to at least about 10, at least 2 to at least about 9, at least 2 to at least about 8, at least 2 to at least about 7, at least 2 to at least about 6, at least 2 to at least about 5, or at least 2 to at least about 4 genes.
[0128] In some aspects, the gene panel comprises at least 2, at least about 3, at least about 4, at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, at least about 10, at least about 11, at least about 12, at least about 13, at least about 14, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, at least about 50, at least about 55, at least about 60, at least about 65, at least about 70, at least about 75, at least about 80, at least about 85, at least about 90, at least about 95, or at least about 100 genes.
[0129] In certain aspects, the gene panel comprises at least 2 genes. In certain aspects, the gene panel comprises at least 3 genes. In certain aspects, the gene panel comprises at least 4 genes. In certain aspects, the gene panel comprises at least 5 genes. In certain aspects, the gene panel comprises at least 6 genes. In certain aspects, the gene panel comprises at least 7 genes. In certain aspects, the gene panel comprises at least 8 genes. In certain aspects, the gene panel comprises at least 9 genes. In certain aspects, the gene panel comprises at least 10 genes. In certain aspects, the gene panel comprises at least 11 genes. In certain aspects, the gene panel comprises at least 12 genes. In certain aspects, the gene panel comprises at least 13 genes. In certain aspects, the gene panel comprises at least 14 genes. In certain aspects, the gene panel comprises at least 15 genes. In certain aspects, the gene panel comprises at least 16 genes. In certain aspects, the gene panel comprises at least 17 genes. In certain aspects, the gene panel comprises at least 18 genes. In certain aspects, the gene panel comprises at least 19 genes. In certain aspects, the gene panel comprises at least 20 genes.
[0130] TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173
(STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173
(STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173 (STING1) TMEM173
(STINGl)In some aspects, the gene panel comprises CCL4, CCL5, CD27, CD276, CD3D,
CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSF18, and one or more additional gene. In some aspects, the gene panel comprises at least about 5, at least about 6, at least about 7, at least about 8, at least about 9, at least about 10, at least about 15, at least about 20, at least about 25, at least about 30, at least about 35, at least about 40, at least about 45, at least about 50, at least about 55, at least about 60, at least about 65, at least about 70, or at least about 75 additional genes. In some aspects, the gene panel comprises less than 95 genes, wherein the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18. In some aspects, the gene panel comprises less than 90 genes, less than about 85 genes, less than about 80 genes, less than about 75 genes, less than about 70 genes, less than about 65 genes, less than about 60 genes, less than about 55 genes, less than about 50 genes, less than about 45 genes, less than about 40 genes, less than about 35 genes, less than about 30 genes, less than about 25 genes, or less than about 20, wherein the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18. In some aspects, the gene panel comprises 16 to 90 genes, 16 to about 85 genes, 16 to about 70 genes, 16 to about 65 genes, 16 to about 60 genes, 16 to about 55 genes, 16 to about 50 genes, 16 to about 45 genes, 16 to about 40 genes, 16 to about 35 genes, 16 to about 30 genes, 16 to about 25 genes, 16 to about 20 genes, wherein the genes comprise CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (SHNG1), and TNFSF18.
[0131] In some aspects, the one or more additional genes are selected from CCL2, CCL3, CCR2, CCR5, CD274, CD28, CDS, CD73, CD80, CD86, CMKLR1, CSF1R, CTLA-4, CXCL11, CXCR2, CXCR6, EP 4, GITR, GZMA, GZMK, HLA-DMA, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-E, IGOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, 0X40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof.
[0132] In some aspects, the panel of genes comprises at least one housekeeping gene. As used herein, a housekeeping gene is a gene that is expressed at a nearly constant level across various cell types. As such, the expression of a house keeping gene can be relative to the number of cells in a sample, acting as a means of normalizing the expression of other variable genes. In some aspects, the panel of genes comprises at least 2 housekeeping genes, at least 3 housekeeping genes, at least housekeeping genes, at least 4 housekeeping genes, at least 5 housekeeping genes, at least 6 housekeeping genes, at least 7 housekeeping genes, at least 8 housekeeping genes, at least 9 housekeeping genes, at least 10 housekeeping genes, at least 11 housekeeping genes, at least 12 housekeeping genes, at least 13 housekeeping genes, at least 14 housekeeping genes, at least 15 housekeeping genes, at least 16 housekeeping genes, at least 17 housekeeping genes, at least 18 housekeeping genes, at least 19 housekeeping genes, at least 20 housekeeping genes, at least 21 housekeeping genes, at least 22 housekeeping genes, at least 23 housekeeping genes, at least 24 housekeeping genes, at least 25 housekeeping genes, at least 26 housekeeping genes, at least housekeeping genes, at least 27 housekeeping genes, at least 28 housekeeping genes, at least 29 housekeeping genes, at least 30 housekeeping genes, at least 35 housekeeping genes, at least 40 housekeeping genes, at least 45 housekeeping genes, at least 50 housekeeping genes, at least 55 housekeeping genes, at least 60 housekeeping genes, at least 65 housekeeping genes, at least 70 housekeeping genes, at least 75 housekeeping genes, at least 80 housekeeping genes, at least 85 housekeeping genes, at least 85 housekeeping genes, at least 90 housekeeping genes, at least 95 housekeeping genes, or at least 100 housekeeping genes. In some aspects, the panel of genes comprises at least 2 housekeeping genes. In some aspects, the panel of genes comprises at least 3 housekeeping genes. In some aspects, the panel of genes comprises at least 4 housekeeping genes. In some aspects, the panel of genes comprises at least 5 housekeeping genes. In some aspects, the panel of genes comprises at least 6 housekeeping genes. In some aspects, the panel of genes comprises at least 7 housekeeping genes. In some aspects, the panel of genes comprises at least 8 housekeeping genes. In some aspects, the panel of genes comprises at least 9 housekeeping genes. In some aspects, the panel of genes comprises at least 10 housekeeping genes. In some aspects, the panel of genes comprises at least 11 housekeeping genes. In some aspects, the panel of genes comprises at least 12 housekeeping genes. In some aspects, the panel of genes comprises at least 13 housekeeping genes. In some aspects, the panel of genes comprises at least 14 housekeeping genes. In some aspects, the panel of genes comprises at least 15 housekeeping genes.
[0133] Any housekeeping genes known in the art can be used in the panel of genes disclosed herein. In some aspects, the housekeeping genes are selected from the group consisting of ACTB, ATP5F1, DDX5, EEF 1G, GAPDH, NCL, OAZ1, PPIA, RPI.38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof
[0134] In some aspects, the panel of genes comprises at least one control gene. In some aspects, the panel of genes comprises at least 2 control genes, at least 3 control genes, at least control genes, at least 4 control genes, at least 5 control genes, at least 6 control genes, at least 7 control genes, at least 8 control genes, at least 9 control genes, at least 10 control genes, at least 11 control genes, at least 12 control genes, at least 13 control genes, at least 14 control genes, at least 15 control genes, at least 16 control genes, at least 17 control genes, at least 18 control genes, at least 19 control genes, at least 20 control genes, at least 21 control genes, at least 22 control genes, at least 23 control genes, at least 24 control genes, at least 25 control genes, at least 26 control genes, at least control genes, at least 27 control genes, at least 28 control genes, at least 29 control genes, at least 30 control genes, at least 35 control genes, at least 40 control genes, at least 45 control genes, at least 50 control genes, at least 55 control genes, at least 60 control genes, at least 65 control genes, at least 70 control genes, at least 75 control genes, at least 80 control genes, at least 85 control genes, at least 85 control genes, at least 90 control genes, at least 95 control genes, or at least 100 control genes. In some aspects, the panel of genes comprises at least 2 control genes. In some aspects, the panel of genes comprises at least 3 control genes. In some aspects, the panel of genes comprises at least 4 control genes. In some aspects, the panel of genes comprises at least 5 control genes. In some aspects, the panel of genes comprises at least 6 control genes. In some aspects, the panel of genes comprises at least 7 control genes. In some aspects, the panel of genes comprises at least 8 control genes. In some aspects, the panel of genes comprises at least 9 control genes. In some aspects, the panel of genes comprises at least 10 control genes. In some aspects, the panel of genes comprises at least 11 control genes. In some aspects, the panel of genes comprises at least 12 control genes. In some aspects, the panel of genes comprises at least 13 control genes. In some aspects, the panel of genes comprises at least 14 control genes. In some aspects, the panel of genes comprises at least 15 control genes. In some aspects, the panel of genes comprises at least 16 control genes. In some aspects, the panel of genes comprises at least 17 control genes. In some aspects, the panel of genes comprises at least 18 control genes. In some aspects, the panel of genes comprises at least 19 control genes. In some aspects, the panel of genes comprises at least 20 control genes. In some aspects, the control genes are selected from the group consisting of ANTI, ANTS, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4, PCL-5, PCL-6, PCL-7, PCL-8, PCL-9, POS1, POS2, POS3, POS4, and any combination thereof.
[0135] In some aspects, the one or more additional genes are selected from CCL2, CCL3, CCR2, CCR5, CD274, CD28, CDS, CD73, CD80, CD86, CMKLR1, CSF1R, CTLA-4, CXCL11, CXCR2, CXCR6, EP 4, GITR, GZMA, GZMK, HLA-DMA, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAGS, Nectin-2, NKG2D, 0X40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIMS, ANTI, ANT2, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4, PCL- 5, PCL-6, PCL-7, PCL-8, PCL-9, POS1, POS2, POS3, POS4, ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof.
[0136] In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene involved in inflammation, e.g., at least one inflammatory gene. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene that is a T cell marker. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene that is a macrophage marker. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene that is an HLA gene. In some aspects, the HLA gene is an MHC class I gene. In some aspects, the HLA gene is an MHC class II gene. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene is an interferon gene. In some aspects, the one or more genes of the pan-tumor inflammation panel comprises at least one gene encodes a protein that is part of the interferon gamma pathway. In some aspects, the one or more genes of the pantumor inflammation panel comprises at least one gene encodes a protein that is a checkpoint inhibitor.
ILA. Gene Expression Profiling
[0137] Gene expression profiling, as used herein, is a measurement of the combined expression level of the one or more genes of the pan-tumor inflammation gene panel disclosed herein. In some aspects, the measurement is made using a sample obtained from a subject. In certain aspects, the sample is a tumor sample. Any biological sample comprising one or more tumor cell can be used in the methods disclosed herein. In some aspects, the sample is selected from a tumor biopsy, a blood sample, a serum sample, or any combination thereof. In certain aspects, the sample is a tumor biopsy collected from the subject prior to administration of a therapy described herein, e.g., an 1-0 therapy, e.g., an anti-PD-l/PD-Ll agonist. In particular aspects, the sample obtained from the subject is a formalin-fixed tumor biopsy. In some aspects, the sample obtained from the subject is a paraffin-embedded tumor biopsy. In some aspects, the sample obtained from the subject is a fresh-frozen tumor biopsy.
[0138] Any method known in the art for measuring the expression of a particular gene or a panel of genes can be used in the methods of the present disclosure. In some aspects, the expression of one or more of the genes in the pan-tumor inflammation gene panel is determined by detecting the presence of mRNA transcribed from the gene, the presence of a protein encoded by the gene, or both.
[0139] In some aspects, the expression of a gene is determined by measuring the level of gene mRNA in a sample obtained from the subject. In certain aspects, the gene expression profile is determined by measuring the level of mRNA transcripts of one or more genes of the pan-tumor inflammation gene panel. Any method known in the art can be used to measure the level of the gene mRNA. In some aspects, the gene mRNA is measured using reverse transcriptase PCR. In some aspects, the gene mRNA is measured using a nuclease protection assay. In some aspects, the gene mRNA is measured using next-generation sequencing (NGS). In some aspects, the gene mRNA is measured using RNA in situ hybridization.
[0140] In some aspects, the expression of a gene is determined by measuring the level of protein encoded by the gene in a sample obtained from the subject. In certain aspects, the gene expression profile is determined by measuring the level of protein encoded by one or more genes of the pan-tumor inflammation gene panel in a sample obtained from the subject. Any method known in the art can be used to measure the level of the protein. In some aspects, the gene expression profile is measured using an immunohistochemistry (IHC) assay. In certain aspects, the IHC is an automated IHC.
[0141] In some aspects, the expression of one or more of the genes of the pan-tumor inflammation gene panel is normalized relative to the expression of one or more housekeeping genes. In some aspects, the one or more housekeeping genes are made up of genes that have relatively consistent expression across various tumor types in various subjects.
[0142] In some aspects, raw gene expression values are normalized following standard gene expression profiling protocols. In these aspects, a gene expression profile can be calculated as the median or average of the log2 -transformed normalized and scaled expression values across all of the target genes in the signature, and presented on a linear scale. In certain aspects, profiles have positive or negative values, depending on whether gene expression is up- or down-regulated under a particular condition.
[0143] In certain aspects, increased/decreased expression is characterized by an expression level that is greater/less than the expression of the same gene or genes in a reference sample. In some aspects, the reference sample comprises a non-tumor tissue of the same subject. In some aspects, the reference sample comprises a corresponding non-tumor tissue of the same subject. In some aspects the reference sample comprises a corresponding tissue in a subject that does not have a tumor. In some aspects, the reference sample comprises more than one tumor tissue sample from more than one other subject, e.g., the increased expression is relative to an average expression level across more than one other tumor samples.
[0144] In some aspects, the increased/decreased expression is characterized by an expression level that is greater/less than a reference expression level. In some aspects, the reference expression level is an average expression level. In some aspects, the average expression level is determined by measuring the expression of the gene (or the multiple genes) present in the gene panel in tumor samples obtained from a population of subjects, and calculating the average for the population of subjects. In some aspects, each member of the population of subjects is afflicted with the same tumor as the subject being administered the 1-0 therapy, e.g., an anti-PD- 1/PD-L1 antagonist.
[0145] In some aspects, increased expression of the upregulated genes is characterized by an expression level that is at least about 5%, at least about 10%, at least about 15%, at least about
20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about
45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about
70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about
95%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 225%, at least about 250%, at least about 275%, or at least about 300% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 25% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 30% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 35% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 40% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 45% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 50% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 55% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 60% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 65% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 70% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 75% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 80% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 85% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 90% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 95% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 100% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 125% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 150% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 175% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 200% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 225% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 250% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 275% higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 300% higher than an expression level in a reference sample or than an average expression level.
[0146] In some aspects, increased expression of the upregulated genes is characterized by an expression level that is at least about 1.25-fold, at least about 1.30-fold, at least about 1.35- fold, at least about 1.40-fold, at least about 1.45-fold, at least about 1.50-fold, at least about 1.55- fold, at least about 1.60-fold, at least about 1.65-fold, at least about 1.70-fold, at least about 1.75- fold, at least about 1.80-fold, at least about 1.85-fold, at least about 1.90-fold, at least about 1.95- fold, at least about 2-fold, at least about 2.25-fold, at least about 2.50-fold, at least about 2.75- fold, at least about 3-fold, at least about 3.25-fold, at least about 3.50-fold, at least about 3.75- fold, or at least about 400-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.25-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.30-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.35-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.40-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.45- fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.50-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.55-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.60-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.65-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.70-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.75-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.80-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.85- fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.90-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 1.95-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2.25-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2.50-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 2.75-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3.25- fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3.50-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 3.75-fold higher than an expression level in a reference sample or than an average expression level. In certain aspects, increased expression is characterized by an expression level that is at least about 4-fold higher than an expression level in a reference sample or than an average expression level.
[0147] In certain aspects, decreased expression of the down-regulated genes is characterized by an expression level that is less than a reference expression level. In some aspects, decreased expression is characterized by an expression level that is at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, at least about 200%, at least about 225%, at least about 250%, at least about 275%, or at least about 300% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 25% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 30% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 35% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 40% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 45% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 50% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 55% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 60% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 65% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 70% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 75% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 80% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 85% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 90% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 95% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 100% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 125% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 150% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 175% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 200% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 225% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 250% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 275% lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 300% lower than an expression level in a reference sample or than an average expression level.
[0148] In some aspects, decreased expression of the down-regulated genes is characterized by an expression level that is at least about 1.25-fold, at least about 1.30-fold, at least about 1.35-fold, at least about 1.40-fold, at least about 1.45-fold, at least about 1.50-fold, at least about 1.55-fold, at least about 1.60-fold, at least about 1.65-fold, at least about 1.70-fold, at least about 1.75-fold, at least about 1.80-fold, at least about 1.85-fold, at least about 1.90-fold, at least about 1.95-fold, at least about 2-fold, at least about 2.25-fold, at least about 2.50-fold, at least about 2.75-fold, at least about 3-fold, at least about 3.25-fold, at least about 3.50-fold, at least about 3.75-fold, or at least about 400-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.25-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.30-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.35-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.40- fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.45-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.50-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.55-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.60-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.65-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.70-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.75-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.80- fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.85-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.90-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 1.95-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2.25-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2.50-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 2.75-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3.25- fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3.50-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 3.75-fold lower than an expression level in a reference sample or than an average expression level. In certain aspects, decreased expression is characterized by an expression level that is at least about 4-fold lower than an expression level in a reference sample or than an average expression level.
ILB. Methods of Treatment
[0149] Certain aspects of the present disclosure are directed to methods of identifying a subject suitable for a therapy and then administering the therapy to the suitable subject. The methods of identifying a suitable subject described herein can be used in advance of any immuno-oncology (I-O) therapy. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds a protein selected from PD-1, PD-L1, CTLA-4, LAG-3, TIGIT, TIM3, CSF1R, NKG2a, 0X40, ICOS, CD137, KIR, TGFp, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, MICA, MICB, and any combination thereof. [0150] In some aspects, the method of identifying a patient in need of an 1-0 therapy comprises (a) obtaining a tumor sample from the patient, and (b) analyzing the expression level of CCL4, CCL5, CD27, CD276, CD3D, CDS A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HLA- DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSFJ8, and any combination thereof, according to the methods disclosed herein. In some aspects, the gene panel consists of CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSFJ8. In some aspects, the method further comprises isolating mRNA from the tumor sample prior to analyzing the expression level of the one or more genes. In some aspects, the expression level of the one or more genes in the gene panel is analyzed by measuring an mRNA level of the one or more genes in the gene panel in the tumor sample. In some aspects, the expression level is measured a nuclease protection assay. In some aspects, the expression level is measured using next-generation sequencing. In some aspects, the expression level is measured using reverse transcriptase polymerase chain reaction (RT-PCR).
[0151] In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigenbinding fragment thereof that specifically binds CTLA-4. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds LAG-3. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds TIGIT. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds TIM3. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds GITR. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds MICA. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigenbinding fragment thereof that specifically binds MICB. In some aspects, the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds CSF1R. [0152] In some aspects, the suitable subject is to be administered and/or subsequently administered more than one antibody or antigen-binding fragment thereof disclosed herein. In some aspects, the suitable subject is to be administered and/or subsequently administered at least two antibodies or antigen-binding fragments thereof. In some aspects, the suitable subject is to be administered and/or subsequently administered at least three antibodies or antigen-binding fragments thereof. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1 and an antibody or antigen-binding fragment thereof that specifically binds CTLA-4. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1 and an antibody or antigen-binding fragment thereof that specifically binds CTLA-4. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1 and an antibody or antigen-binding fragment thereof that specifically binds CSF1R. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1 and an antibody or antigen-binding fragment thereof that specifically binds CSF1R. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-1 and an antibody or antigen-binding fragment thereof that specifically binds LAG-3. In certain aspects the suitable subject is to be administered and/or subsequently administered an antibody or antigen-binding fragment thereof that specifically binds PD-L1 and an antibody or antigenbinding fragment thereof that specifically binds LAG-3.
[0153] In certain aspects, the therapy is administered to the suitable subject after the gene expression profile has been measured. In some aspects, the measuring is in vitro. In other aspects, the measuring is in vivo. In some aspects, the therapy is administered at least about 1 day, at least about 2 days, at least about 3 days, at least about 4 days, at least about 5 days, at least about 6 days, at least about 7 days, at least about 8 days, at least about 9 days, at least about 10 days, at least about 11 days, at least about 12 days, at least about 13 days, or at least about 14 days after the gene expression profile has been measured.
[0154] In some aspects, the particular therapy to be administered and/or subsequently administered to the suitable subject is dependent on the gene expression profile. In some aspects, a first therapy is administered if a subject has a first type of gene expression profile. In some aspects, a second therapy is administered if a subject has a second type of gene expression profile. In some aspects, the first and second therapy are different. In some aspects, the first therapy, the second therapy, or both comprises an 1-0 monotherapy. In some aspects, a subject identified as having a high pan-tumor gene signature score is administered an 1-0 therapy as a first therapy and/or a second therapy. In some aspects, the first therapy, the second therapy, or both comprises a combination therapy comprising (i) an 1-0 therapy and (ii) one or more additional anti-cancer agent. In some aspects, a subject identified as having a moderate pantumor gene signature score is administered a combination therapy comprising (i) an 1-0 therapy and (ii) one or more additional anti-cancer agent as a first therapy and/or a second therapy. In some aspects, the first therapy comprises an 1-0 monotherapy and the second therapy comprises a combination therapy comprising (i) an 1-0 therapy and (ii) one or more additional anti-cancer agent.
[0155] In some aspects, the present disclosure is directed to a pharmaceutical composition comprising an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist, for use in a method of identifying a human subject suitable for the 1-0 therapy, e.g., anti-PD-l/PD-Ll antagonist, wherein the method comprises measuring expression of CCL4, CCL5, CD27, CD276, CD3D, CDS A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HIA-DRA, HLA-DRB1, I.GAI.S9, NKG7, TMEM173 (STING1), and TNFSF18, according to the methods disclosed herein. In some aspects, the subject is identified as being suitable for an 1-0 therapy when the tumor sample exhibits a high pan-tumor gene signature score. In some aspects, the subject is identified as being suitable for a combination therapy comprising (i) an 1-0 therapy and (ii) an additional anticancer agent, when the tumor sample exhibits a moderate pan-tumor gene signature score. In some aspects, the subject is identified as being suitable for an anticancer therapy when the tumor sample exhibits a low pan-tumor gene signature score, wherein the anticancer therapy does not comprise an 1-0 therapy.
[0156] The present disclosure provides, in some aspects, a pharmaceutical composition comprising an 1-0 therapy, e.g., an anti-PD-l/PD-Ll antagonist, for use in a method of treating a human subject afflicted with a tumor, wherein a tumor sample obtained from the subject exhibits a high pan-tumor gene signature score.
[0157] In other aspects, the disclosure provides a method of identifying a human subject suitable for an 1-0 therapy, e.g. an anti-PD-l/PD-Ll antagonist therapy, comprising measuring expression of CCI.4, CCL5, CD27, CD276, CD3D, CDS A, CXCI.10, CXCI.9, CXCR1, HLA- DMB, HIA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), and TNFSF18, according to the methods disclosed herein. In some aspects, the measuring is in vitro. In other aspects, the measuring is in vivo.
[0158] In certain aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an 1-0 therapy described herein. In some aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an anti-PD-l/PD-Ll antagonist. In some aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an anti-PD-1 antibody. In some aspects, the subject identified as having a high pan-tumor gene signature score is to be administered and/or subsequently administered an anti-PD-Ll antibody.
[0159] In certain aspects, the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an 1-0 therapy described herein and (ii) one or more additional anti-cancer agents. In some aspects, the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an anti- PD-1/PD-L1 antagonist and (ii) one or more additional anti-cancer agents. In some aspects, the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an anti-PD-1 antibody and (ii) one or more additional anti-cancer agents. In some aspects, the subject identified as having a moderate pan-tumor gene signature score is to be administered and/or subsequently administered a combination therapy comprising (i) an anti-PD-Ll antibody and (ii) one or more additional anti-cancer agents.
II.C. Antibodies
[0160] Certain aspects of the present disclosure are directed to methods of treating a suitable subject, as determined according to a method disclosed herein, using an 1-0 therapy. Any 1-0 therapy known in the art can be used in the methods described herein. In certain aspects, the 1-0 therapy comprises administering to the suitable subject an antibody or an antigen-binding fragment thereof the specifically binds a protein selected from PD-1, PD-L1, CTLA-4, LAG-3, TIGIT, TIM3, CSF1R, NKG2a, 0X40, ICOS, CD137, KIR, TGFp, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, MICA, MICB, and any combination thereof. [0161] In some aspects, the subject is administered a single 1-0 therapy, i.e., a monotherapy. In some aspects, the subject is administered an anti-PD-1 antibody monotherapy. In some aspects, the subject is administered a combination therapy comprising a first 1-0 therapy and a second 1-0 therapy. In some aspects, the subject is administered a combination therapy comprising administering a first 1-0 therapy and an additional anti-cancer agent. In some aspects, the additional anti-cancer agent comprises a second 1-0 therapy, a chemotherapy, a standard of care therapy, or any combination thereof.
[0162] In certain aspects, the subject is administered a combination therapy comprising an anti-PD-1 antibody and a second anti-cancer agent. In certain aspects, the subject is administered a combination therapy comprising an anti-PD-1 antibody and an anti-CTLA-4 antibody. In certain aspects, the subject is administered a combination therapy comprising an anti-PD-1 antibody and an anti-CSFIR antibody.
[0163] In certain aspects, the subject is administered a combination therapy comprising an anti-PD-Ll antibody and a second anti-cancer agent. In certain aspects, the subject is administered a combination therapy comprising an anti-PD-Ll antibody and an anti-CTLA-4 antibody. In certain aspects, the subject is administered a combination therapy comprising an anti-PD-Ll antibody and an anti-CSFIR antibody.
II.C.1. Anti-PD-1 Antibodies Useful for the Disclosure
[0164] Anti-PD-1 antibodies that are known in the art can be used in the presently described compositions and methods. Various human monoclonal antibodies that bind specifically to PD-1 with high affinity have been disclosed in U.S. Patent No. 8,008,449. Anti- PD-1 human antibodies disclosed in U.S. Patent No. 8,008,449 have been demonstrated to exhibit one or more of the following characteristics: (a) bind to human PD-1 with a KD of 1 x 10" 7 M or less, as determined by surface plasmon resonance using a Biacore biosensor system; (b) do not substantially bind to human CD28, CTLA-4 or ICOS; (c) increase T-cell proliferation in a Mixed Lymphocyte Reaction (MLR) assay; (d) increase interferon-y production in an MLR assay; (e) increase IL-2 secretion in an MLR assay; (f) bind to human PD-1 and cynomolgus monkey PD-1; (g) inhibit the binding of PD-L1 and/or PD-L2 to PD-1; (h) stimulate antigenspecific memory responses; (i) stimulate antibody responses; and (j) inhibit tumor cell growth in vivo. Anti-PD-1 antibodies usable in the present disclosure include monoclonal antibodies that bind specifically to human PD-1 and exhibit at least one, in some aspects, at least five, of the preceding characteristics. [0165] Other anti-PD-1 monoclonal antibodies have been described in, for example, U.S. Patent Nos. 6,808,710, 7,488,802, 8,168,757 and 8,354,509, US Publication No. 2016/0272708, and PCT Publication Nos. WO 2012/145493, WO 2008/156712, WO 2015/112900, WO 2012/145493, WO 2015/112800, WO 2014/206107, WO 2015/35606, WO 2015/085847, WO
2014/179664, WO 2017/020291, WO 2017/020858, WO 2016/197367, WO 2017/024515, WO
2017/025051, WO 2017/123557, WO 2016/106159, WO 2014/194302, WO 2017/040790, WO
2017/133540, WO 2017/132827, WO 2017/024465, WO 2017/025016, WO 2017/106061, WO
2017/19846, WO 2017/024465, WO 2017/025016, WO 2017/132825, and WO 2017/133540 each of which is incorporated by reference in its entirety.
[0166] In some aspects, the anti-PD-1 antibody is selected from the group consisting of nivolumab (also known as OPDIVO®, 5C4, BMS-936558, MDX-1106, and ONO-4538), pembrolizumab (Merck; also known as KEYTRUDA®, lambrolizumab, and MK-3475; see WO2008/156712), PDR001 (Novartis; see WO 2015/112900), MEDI-0680 (AstraZeneca; also known as AMP-514; see WO 2012/145493), cemiplimab (Regeneron; also known as REGN- 2810; see WO 2015/112800), JS001 (TAIZHOU JUNSHI PHARMA; also known as toripalimab; see Si-Yang Liu et al., J. Hematol. Oncol. 70: 136 (2017)), BGB-A317 (Beigene; also known as Tislelizumab; see WO 2015/35606 and US 2015/0079109), INCSHR1210 (Jiangsu Hengrui Medicine; also known as SHR-1210; see WO 2015/085847; Si-Yang Liu et al., J. Hematol. Oncol. 70: 136 (2017)), TSR-042 (Tesaro Biopharmaceutical; also known as ANB011; see WO2014/179664), GLS-010 (Wuxi/Harbin Gloria Pharmaceuticals; also known as WBP3055; see Si-Yang Liu et al., J. Hematol. Oncol. 70: 136 (2017)), AM-0001 (Armo), STI- 1110 (Sorrento Therapeutics; see WO 2014/194302), AGEN2034 (Agenus; see WO 2017/040790), MGA012 (Macrogenics, see WO 2017/19846), BCD-100 (Biocad; Kaplon et al., mAbs 70(2/ 183-203 (2018), and IBI308 (Innovent; see WO 2017/024465, WO 2017/025016, WO 2017/132825, and WO 2017/133540).
[0167] In one aspect, the anti-PD-1 antibody is nivolumab. Nivolumab is a fully human IgG4 (S228P) PD-1 immune checkpoint inhibitor antibody that selectively prevents interaction with PD-1 ligands (PD-L1 and PD-L2), thereby blocking the down-regulation of antitumor T-cell functions (U.S. Patent No. 8,008,449; Wang et al., 2014 Cancer Immunol Res. 2(9/846-56).
[0168] In another aspect, the anti-PD-1 antibody is pembrolizumab. Pembrolizumab is a humanized monoclonal IgG4 (S228P) antibody directed against human cell surface receptor PD- 1 (programmed death-1 or programmed cell death-1). Pembrolizumab is described, for example, in U.S. Patent Nos. 8,354,509 and 8,900,587. [0169] Anti-PD-1 antibodies usable in the disclosed compositions and methods also include isolated antibodies that bind specifically to human PD-1 and cross-compete for binding to human PD-1 with any anti-PD-1 antibody disclosed herein, e.g., nivolumab (see, e.g., U.S. Patent No. 8,008,449 and 8,779,105; WO 2013/173223). In some aspects, the anti-PD-1 antibody binds the same epitope as any of the anti-PD-1 antibodies described herein, e.g, nivolumab. The ability of antibodies to cross-compete for binding to an antigen indicates that these monoclonal antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other cross-competing antibodies to that particular epitope region. These cross-competing antibodies are expected to have functional properties very similar those of the reference antibody, e.g, nivolumab, by virtue of their binding to the same epitope region of PD-1. Cross-competing antibodies can be readily identified based on their ability to cross-compete with nivolumab in standard PD-1 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
[0170] In certain aspects, the antibodies that cross-compete for binding to human PD-1 with, or bind to the same epitope region of human PD-1 antibody, nivolumab, are monoclonal antibodies. For administration to human subjects, these cross-competing antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies. Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
[0171] Anti-PD-1 antibodies usable in the compositions and methods of the disclosed disclosure also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
[0172] Anti-PD-1 antibodies suitable for use in the disclosed compositions and methods are antibodies that bind to PD-1 with high specificity and affinity, block the binding of PD-L1 and or PD-L2, and inhibit the immunosuppressive effect of the PD-1 signaling pathway. In any of the compositions or methods disclosed herein, an anti-PD-1 "antibody" includes an antigenbinding portion or fragment that binds to the PD-1 receptor and exhibits the functional properties similar to those of whole antibodies in inhibiting ligand binding and up-regulating the immune system. In certain aspects, the anti-PD-1 antibody or antigen-binding portion thereof crosscompetes with nivolumab for binding to human PD-1.
[0173] In some aspects, the anti-PD-1 antibody is administered at a dose ranging from 0.1 mg/kg to 20.0 mg/kg body weight once every 2, 3, 4, 5, 6, 7, or 8 weeks, e.g., 0.1 mg/kg to 10.0 mg/kg body weight once every 2, 3, or 4 weeks. In other aspects, the anti-PD-1 antibody is administered at a dose of about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, or 10 mg/kg body weight once every 2 weeks. In other aspects, the anti-PD-1 antibody is administered at a dose of about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, or 10 mg/kg body weight once every 3 weeks. In one aspect, the anti-PD-1 antibody is administered at a dose of about 5 mg/kg body weight about once every 3 weeks. In another aspect, the anti-PD-1 antibody, e.g., nivolumab, is administered at a dose of about 3 mg/kg body weight about once every 2 weeks. In other aspects, the anti-PD-1 antibody, e.g., pembrolizumab, is administered at a dose of about 2 mg/kg body weight about once every 3 weeks.
[0174] The anti-PD-1 antibody useful for the present disclosure can be administered as a flat dose. In some aspects, the anti-PD-1 antibody is administered at a flat dose of from about 100 to about 1000 mg, from about 100 mg to about 900 mg, from about 100 mg to about 800 mg, from about 100 mg to about 700 mg, from about 100 mg to about 600 mg, from about 100 mg to about 500 mg, from about 200 mg to about 1000 mg, from about 200 mg to about 900 mg, from about 200 mg to about 800 mg, from about 200 mg to about 700 mg, from about 200 mg to about 600 mg, from about 200 mg to about 500 mg, from about 200 mg to about 480 mg, or from about 240 mg to about 480 mg, In one aspect, the anti-PD-1 antibody is administered as a flat dose of at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, at least about 400 mg, at least about 420 mg, at least about 440 mg, at least about 460 mg, at least about 480 mg, at least about 500 mg, at least about 520 mg, at least about 540 mg, at least about 550 mg, at least about 560 mg, at least about 580 mg, at least about 600 mg, at least about 620 mg, at least about 640 mg, at least about 660 mg, at least about 680 mg, at least about 700 mg, or at least about 720 mg at a dosing interval of about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 weeks. In another aspects, the anti-PD-1 antibody is administered as a flat dose of about 200 mg to about 800 mg, about 200 mg to about 700 mg, about 200 mg to about 600 mg, about 200 mg to about 500 mg, at a dosing interval of about 1, 2, 3, or 4 weeks.
[0175] In some aspects, the anti-PD-1 antibody is administered as a flat dose of about 200 mg at about once every 3 weeks. In other aspects, the anti-PD-1 antibody is administered as a flat dose of about 200 mg at about once every 2 weeks. In other aspects, the anti-PD-1 antibody is administered as a flat dose of about 240 mg at about once every 2 weeks. In certain aspects, the anti-PD-1 antibody is administered as a flat dose of about 480 mg at about once every 4 weeks. [0176] In some aspects, nivolumab is administered at a flat dose of about 240 mg once about every 2 weeks. In some aspects, nivolumab is administered at a flat dose of about 240 mg once about every 3 weeks. In some aspects, nivolumab is administered at a flat dose of about 360 mg once about every 3 weeks. In some aspects, nivolumab is administered at a flat dose of about 480 mg once about every 4 weeks.
[0177] In some aspects, pembrolizumab is administered at a flat dose of about 200 mg once about every 2 weeks. In some aspects, pembrolizumab is administered at a flat dose of about 200 mg once about every 3 weeks. In some aspects, pembrolizumab is administered at a flat dose of about 400 mg once about every 4 weeks.
[0178] In some aspects, the PD-1 inhibitor is a small molecule. In some aspects, the PD-1 inhibitor comprises a millamolecule. In some aspects, the PD-1 inhibitor comprises a macrocyclic peptide. In certain aspects, the PD-1 inhibitor comprises BMS-986189. In some aspects, the PD-1 inhibitor comprises an inhibitor disclosed in International Publication No. WO2014/151634, which is incorporated by reference herein in its entirety. In some aspects, the PD-1 inhibitor comprises INCMGA00012 (Insight Pharmaceuticals). In some aspects, the PD-1 inhibitor comprises a combination of an anti -PD-1 antibody disclosed herein and a PD-1 small molecule inhibitor.
II.C.2. Anti-PD-Ll Antibodies Useful for the Disclosure
[0179] In certain aspects, an anti-PD-Ll antibody is substituted for the anti-PD-1 antibody in any of the methods disclosed herein. Anti-PD-Ll antibodies that are known in the art can be used in the compositions and methods of the present disclosure. Examples of anti-PD-Ll antibodies useful in the compositions and methods of the present disclosure include the antibodies disclosed in US Patent No. 9,580,507. Anti-PD-Ll human monoclonal antibodies disclosed in U.S. Patent No. 9,580,507 have been demonstrated to exhibit one or more of the following characteristics: (a) bind to human PD-L1 with a KD of 1 x 10'7 M or less, as determined by surface plasmon resonance using a Biacore biosensor system; (b) increase T-cell proliferation in a Mixed Lymphocyte Reaction (MLR) assay; (c) increase interferon-y production in an MLR assay; (d) increase IL-2 secretion in an MLR assay; (e) stimulate antibody responses; and (f) reverse the effect of T regulatory cells on T cell effector cells and/or dendritic cells. Anti- PD-Ll antibodies usable in the present disclosure include monoclonal antibodies that bind specifically to human PD-L1 and exhibit at least one, in some aspects, at least five, of the preceding characteristics. [0180] In certain aspects, the anti-PD-Ll antibody is selected from the group consisting of BMS-936559 (also known as 12A4, MDX-1105; see, e.g, U.S. Patent No. 7,943,743 and WO 2013/173223), atezolizumab (Roche; also known as TECENTRIQ®; MPDL3280A, RG7446; see US 8,217,149; see, also, Herbst et al. (2013) J Clin Oncol 31(suppl):3000), durvalumab (AstraZeneca; also known as IMFINZI™, MEDI-4736; see WO 2011/066389), avelumab (Pfizer; also known as BAVENCIO®, MSB-0010718C; see WO 2013/079174), STI-1014 (Sorrento; see WO2013/181634), CX-072 (Cytomx; see WO2016/149201 ), KN035 (3D Med/Alphamab; see Zhang et al., Cell Discov. 7:3 (March 2017), LY3300054 (Eli Lilly Co.; see, e.g., WO 2017/034916), BGB-A333 (BeiGene; see Desai et al., JCO 36 (15suppl)^ 3 3 (2018)), and CK-301 (Checkpoint Therapeutics; see Gorelik et al., AACR:Abstract 4606 (Apr 2016)).
[0181] In certain aspects, the PD-L1 antibody is atezolizumab (TECENTRIQ®). Atezolizumab is a fully humanized IgGl monoclonal anti-PD-Ll antibody.
[0182] In certain aspects, the PD-L1 antibody is durvalumab (IMFINZI™). Durvalumab is a human IgGl kappa monoclonal anti-PD-Ll antibody.
[0183] In certain aspects, the PD-L1 antibody is avelumab (BAVENCIO®). Avelumab is a human IgGl lambda monoclonal anti-PD-Ll antibody.
[0184] Anti-PD-Ll antibodies usable in the disclosed compositions and methods also include isolated antibodies that bind specifically to human PD-L1 and cross-compete for binding to human PD-L1 with any anti-PD-Ll antibody disclosed herein, e.g., atezolizumab, durvalumab, and/or avelumab. In some aspects, the anti-PD-Ll antibody binds the same epitope as any of the anti-PD-Ll antibodies described herein, e.g., atezolizumab, durvalumab, and/or avelumab. The ability of antibodies to cross-compete for binding to an antigen indicates that these antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other crosscompeting antibodies to that particular epitope region. These cross-competing antibodies are expected to have functional properties very similar those of the reference antibody, e.g., atezolizumab and/or avelumab, by virtue of their binding to the same epitope region of PD-L1. Cross-competing antibodies can be readily identified based on their ability to cross-compete with atezolizumab and/or avelumab in standard PD-L1 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
[0185] In certain aspects, the antibodies that cross-compete for binding to human PD-L1 with, or bind to the same epitope region of human PD-L1 antibody as, atezolizumab, durvalumab, and/or avelumab, are monoclonal antibodies. For administration to human subjects, these cross-competing antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies. Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
[0186] Anti-PD-Ll antibodies usable in the compositions and methods of the disclosed disclosure also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
[0187] Anti-PD-Ll antibodies suitable for use in the disclosed compositions and methods are antibodies that bind to PD-L1 with high specificity and affinity, block the binding of PD-1, and inhibit the immunosuppressive effect of the PD-1 signaling pathway. In any of the compositions or methods disclosed herein, an anti-PD-Ll "antibody" includes an antigen-binding portion or fragment that binds to PD-L1 and exhibits the functional properties similar to those of whole antibodies in inhibiting receptor binding and up-regulating the immune system. In certain aspects, the anti-PD-Ll antibody or antigen-binding portion thereof cross-competes with atezolizumab, durvalumab, and/or avelumab for binding to human PD-L1.
[0188] The anti-PD-Ll antibody useful for the present disclosure can be any PD-L1 antibody that specifically binds to PD-L1, e.g., antibodies that cross-compete with durvalumab, avelumab, or atezolizumab for binding to human PD-1, e.g., an antibody that binds to the same epitope as durvalumab, avelumab, or atezolizumab. In a particular aspect, the anti-PD-Ll antibody is durvalumab. In other aspects, the anti-PD-Ll antibody is avelumab. In some aspects, the anti-PD-Ll antibody is atezolizumab.
[0189] In some aspects, the anti-PD-Ll antibody is administered at a dose ranging from about 0.1 mg/kg to about 20.0 mg/kg body weight, about 2 mg/kg, about 3 mg/kg, about 4 mg/kg, about 5 mg/kg, about 6 mg/kg, about 7 mg/kg, about 8 mg/kg, about 9 mg/kg, about 10 mg/kg, about 11 mg/kg, about 12 mg/kg, about 13 mg/kg, about 14 mg/kg, about 15 mg/kg, about 16 mg/kg, about 17 mg/kg, about 18 mg/kg, about 19 mg/kg, or about 20 mg/kg, about once every 2, 3, 4, 5, 6, 7, or 8 weeks.
[0190] In some aspects, the anti-PD-Ll antibody is administered at a dose of about 15 mg/kg body weight at about once every 3 weeks. In other aspects, the anti-PD-Ll antibody is administered at a dose of about 10 mg/kg body weight at about once every 2 weeks.
[0191] In other aspects, the anti-PD-Ll antibody useful for the present disclosure is a flat dose. In some aspects, the anti-PD-Ll antibody is administered as a flat dose of from about 200 mg to about 1600 mg, about 200 mg to about 1500 mg, about 200 mg to about 1400 mg, about 200 mg to about 1300 mg, about 200 mg to about 1200 mg, about 200 mg to about 1100 mg, about 200 mg to about 1000 mg, about 200 mg to about 900 mg, about 200 mg to about 800 mg, about 200 mg to about 700 mg, about 200 mg to about 600 mg, about 700 mg to about 1300 mg, about 800 mg to about 1200 mg, about 700 mg to about 900 mg, or about 1100 mg to about 1300 mg. In some aspects, the anti-PD-Ll antibody is administered as a flat dose of at least about 240 mg, at least about 300 mg, at least about 320 mg, at least about 400 mg, at least about 480 mg, at least about 500 mg, at least about 560 mg, at least about 600 mg, at least about 640 mg, at least about 700 mg, at least 720 mg, at least about 800 mg, at least about 840 mg, at least about 880 mg, at least about 900 mg, at least 960 mg, at least about 1000 mg, at least about 1040 mg, at least about 1100 mg, at least about 1120 mg, at least about 1200 mg, at least about 1280 mg, at least about 1300 mg, at least about 1360 mg, or at least about 1400 mg, at a dosing interval of about 1, 2, 3, or 4 weeks. In some aspects, the anti-PD-Ll antibody is administered as a flat dose of about 1200 mg at about once every 3 weeks. In other aspects, the anti-PD-Ll antibody is administered as a flat dose of about 800 mg at about once every 2 weeks. In other aspects, the anti-PD-Ll antibody is administered as a flat dose of about 840 mg at about once every 2 weeks. [0192] In some aspects, atezolizumab is administered as a flat dose of about 1200 mg once about every 3 weeks. In some aspects, atezolizumab is administered as a flat dose of about 800 mg once about every 2 weeks. In some aspects, atezolizumab is administered as a flat dose of about 840 mg once about every 2 weeks.
[0193] In some aspects, avelumab is administered as a flat dose of about 800 mg once about every 2 weeks.
[0194] In some aspects, durvalumab is administered at a dose of about 10 mg/kg once about every 2 weeks. In some aspects, durvalumab is administered as a flat dose of about 800 mg/kg once about every 2 weeks. In some aspects, durvalumab is administered as a flat dose of about 1200 mg/kg once about every 3 weeks.
[0195] In some aspects, the PD-L1 inhibitor is a small molecule. In some aspects, the PD- L1 inhibitor comprises a millamolecule. In some aspects, the PD-L1 inhibitor comprises a macrocyclic peptide. In certain aspects, the PD-L1 inhibitor comprises BMS-986189.
[0196] In some aspects, the PD-L1 inhibitor comprises a millamolecule having a formula set forth in formula (II):
Figure imgf000063_0001
wherein Rx-R13 are amino acid side chains, Ra-Rn are hydrogen, methyl, or form a ring with a vicinal R group, and R14 is -C(O)NHR15, wherein R15 is hydrogen, or a glycine residue optionally substituted with additional glycine residues and/or tails which can improve pharmacokinetic properties. In some aspects, the PD-L1 inhibitor comprises a compound disclosed in International Publication No. WO2014/151634, which is incorporated by reference herein in its entirety. In some aspects, the PD-L1 inhibitor comprises a compound disclosed in International Publication No. WO2016/039749, WO2016/149351, WO2016/077518, W02016/100285, WO2016/100608, WO2016/126646, WO2016/057624, W02017/151830, WO20 17/176608, W02018/085750, WO2018/237153, or WO2019/070643, each of which is incorporated by reference herein in its entirety.
[0198] In certain aspects the PD-L1 inhibitor comprises a small molecule PD-L1 inhibitor disclosed in International Publication No. WO2015/034820, WO2015/160641, WO2018/044963, WO20 17/066227, W02018/009505, WO2018/183171, WO2018/118848, WO2019/147662, or WO2019/169123, each of which is incorporated by reference herein in its entirety.
[0199] In some aspects, the PD-L1 inhibitor comprises a combination of an anti-PD-Ll antibody disclosed herein and a PD-L1 small molecule inhibitor disclosed herein.
II.C.3. Anti-CTLA-4 Antibodies
[0200] Anti-CTLA-4 antibodies that are known in the art can be used in the compositions and methods of the present disclosure. Anti-CTLA-4 antibodies of the instant disclosure bind to human CTLA-4 so as to disrupt the interaction of CTLA-4 with a human B7 receptor. Because the interaction of CTLA-4 with B7 transduces a signal leading to inactivation of T-cells bearing the CTLA-4 receptor, disruption of the interaction effectively induces, enhances or prolongs the activation of such T cells, thereby inducing, enhancing or prolonging an immune response.
[0201] Human monoclonal antibodies that bind specifically to CTLA-4 with high affinity have been disclosed in U.S. Patent Nos. 6,984,720. Other anti-CTLA-4 monoclonal antibodies have been described in, for example, U.S. Patent Nos. 5,977,318, 6,051,227, 6,682,736, and 7,034,121 and International Publication Nos. WO 2012/122444, WO 2007/113648, WO 2016/196237, and WO 2000/037504, each of which is incorporated by reference herein in its entirety. The anti-CTLA-4 human monoclonal antibodies disclosed in U.S. Patent No. Nos. 6,984,720 have been demonstrated to exhibit one or more of the following characteristics: (a) binds specifically to human CTLA-4 with a binding affinity reflected by an equilibrium association constant (Ka) of at least about 107 M'1, or about 109 M'1, or about 1010 M'1 to 1011 M'1 or higher, as determined by Biacore analysis; (b) a kinetic association constant (U of at least about 103, about 104, or about 105 m'1 s'1; (c) a kinetic disassociation constant (k< ) of at least about 103, about 104, or about 105 m'1 s'1; and (d) inhibits the binding of CTLA-4 to B7-1 (CD80) and B7-2 (CD86). Anti-CTLA-4 antibodies useful for the present disclosure include monoclonal antibodies that bind specifically to human CTLA-4 and exhibit at least one, at least two, or at least three of the preceding characteristics.
[0202] In certain aspects, the CTLA-4 antibody is selected from the group consisting of ipilimumab (also known as YERVOY®, MDX-010, 10D1; see U.S. Patent No. 6,984,720), MK- 1308 (Merck), AGEN-1884 (Agenus Inc.; see WO 2016/196237), and tremelimumab (AstraZeneca; also known as ticilimumab, CP-675,206; see WO 2000/037504 and Ribas, Update Cancer Ther. 2(3): 133-39 (2007)). In particular aspects, the anti-CTLA-4 antibody is ipilimumab.
[0203] In particular aspects, the CTLA-4 antibody is ipilimumab for use in the compositions and methods disclosed herein. Ipilimumab is a fully human, IgGl monoclonal antibody that blocks the binding of CTLA-4 to its B7 ligands, thereby stimulating T cell activation and improving overall survival (OS) in patients with advanced melanoma.
[0204] In particular aspects, the CTLA-4 antibody is tremelimumab.
[0205] In particular aspects, the CTLA-4 antibody is MK-1308.
[0206] In particular aspects, the CTLA-4 antibody is AGEN-1884.
[0207] Anti-CTLA-4 antibodies usable in the disclosed compositions and methods also include isolated antibodies that bind specifically to human CTLA-4 and cross-compete for binding to human CTLA-4 with any anti-CTLA-4 antibody disclosed herein, e.g., ipilimumab and/or tremelimumab. In some aspects, the anti-CTLA-4 antibody binds the same epitope as any of the anti-CTLA-4 antibodies described herein, e.g., ipilimumab and/or tremelimumab. The ability of antibodies to cross-compete for binding to an antigen indicates that these antibodies bind to the same epitope region of the antigen and sterically hinder the binding of other crosscompeting antibodies to that particular epitope region. These cross-competing antibodies are expected to have functional properties very similar those of the reference antibody, e.g., ipilimumab and/or tremelimumab, by virtue of their binding to the same epitope region of CTLA-4. Cross-competing antibodies can be readily identified based on their ability to crosscompete with ipilimumab and/or tremelimumab in standard CTLA-4 binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
[0208] In certain aspects, the antibodies that cross-compete for binding to human CTLA- 4 with, or bind to the same epitope region of human CTLA-4 antibody as, ipilimumab and/or tremelimumab, are monoclonal antibodies. For administration to human subjects, these crosscompeting antibodies are chimeric antibodies, engineered antibodies, or humanized or human antibodies. Such chimeric, engineered, humanized or human monoclonal antibodies can be prepared and isolated by methods well known in the art.
[0209] Anti-CTLA-4 antibodies usable in the compositions and methods of the disclosed disclosure also include antigen-binding portions of the above antibodies. It has been amply demonstrated that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody.
[0210] Anti-CTLA-4 antibodies suitable for use in the disclosed methods or compositions are antibodies that bind to CTLA-4 with high specificity and affinity, block the activity of CTLA-4, and disrupt the interaction of CTLA-4 with a human B7 receptor. In any of the compositions or methods disclosed herein, an anti-CTLA-4 "antibody" includes an antigenbinding portion or fragment that binds to CTLA-4 and exhibits the functional properties similar to those of whole antibodies in inhibiting the interaction of CTLA-4 with a human B7 receptor and up-regulating the immune system. In certain aspects, the anti-CTLA-4 antibody or antigenbinding portion thereof cross-competes with ipilimumab and/or tremelimumab for binding to human CTLA-4.
[0211] In some aspects, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered at a dose ranging from 0.1 mg/kg to 10.0 mg/kg body weight once every 2, 3, 4, 5, 6, 7, or 8 weeks. In some aspects, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered at a dose of 1 mg/kg or 3 mg/kg body weight once every 3, 4, 5, or 6 weeks. In one aspect, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered at a dose of 3 mg/kg body weight once every 2 weeks. In another aspect, the anti-PD-1 antibody or antigenbinding portion thereof is administered at a dose of 1 mg/kg body weight once every 6 weeks.
[0212] In some aspects, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered as a flat dose. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose of from about 10 to about 1000 mg, from about 10 mg to about 900 mg, from about 10 mg to about 800 mg, from about 10 mg to about 700 mg, from about 10 mg to about 600 mg, from about 10 mg to about 500 mg, from about 100 mg to about 1000 mg, from about 100 mg to about 900 mg, from about 100 mg to about 800 mg, from about 100 mg to about 700 mg, from about 100 mg to about 100 mg, from about 100 mg to about 500 mg, from about 100 mg to about 480 mg, or from about 240 mg to about 480 mg. In one aspect, the anti-CTLA-4 antibody or antigenbinding portion thereof is administered as a flat dose of at least about 60 mg, at least about 80 mg, at least about 100 mg, at least about 120 mg, at least about 140 mg, at least about 160 mg, at least about 180 mg, at least about 200 mg, at least about 220 mg, at least about 240 mg, at least about 260 mg, at least about 280 mg, at least about 300 mg, at least about 320 mg, at least about 340 mg, at least about 360 mg, at least about 380 mg, at least about 400 mg, at least about 420 mg, at least about 440 mg, at least about 460 mg, at least about 480 mg, at least about 500 mg, at least about 520 mg at least about 540 mg, at least about 550 mg, at least about 560 mg, at least about 580 mg, at least about 600 mg, at least about 620 mg, at least about 640 mg, at least about 660 mg, at least about 680 mg, at least about 700 mg, or at least about 720 mg. In another aspect, the anti-CTLA-4 antibody or antigen-binding portion thereof is administered as a flat dose about once every 1, 2, 3, 4, 5, 6, 7, or 8 weeks.
[0213] In some aspects, ipilimumab is administered at a dose of about 3 mg/kg once about every 3 weeks. In some aspects, ipilimumab is administered at a dose of about 10 mg/kg once about every 3 weeks. In some aspects, ipilimumab is administered at a dose of about 10 mg/kg once about every 12 weeks. In some aspects, the ipilimumab is administered for four doses.
II.C.4. Anti-LAG-3 Antibodies
[0214] Anti-LAG-3 antibodies of the instant disclosure bind to human LAG-3. Antibodies that bind to LAG-3 have been disclosed in Int'l Publ. No. WO/2015/042246 and U.S. Publ. Nos. 2014/0093511 and 2011/0150892, each of which is incorporated by reference herein in its entirety. [0215] An exemplary LAG-3 antibody useful in the present disclosure is 25F7 (described in U.S. Publ. No. 2011/0150892). An additional exemplary LAG-3 antibody useful in the present disclosure is BMS-986016. In one aspect, an anti-LAG-3 antibody useful for the composition cross-competes with 25F7 or BMS-986016. In another aspect, an anti -LAG-3 antibody useful for the composition binds to the same epitope as 25F7 or BMS-986016. In other aspects, an anti- LAG-3 antibody comprises six CDRs of 25F7 or BMS-986016. In another aspect, the anti-LAG- 3 antibody is IMP731 (H5L7BW), MK-4280 (28G-10), REGN3767, humanized BAP050, IMP-
701 (LAG-5250), TSR-033, BI754111, MGD013, or FS-118. These and other anti-LAG-3 antibodies useful in the claimed invention can be found in, for example: WO2016/028672, W02017/106129, WO2017/062888, W02009/044273, WO2018/069500, WO2016/126858, WO2014/179664, WO2016/200782, WO2015/200119, WO2017/019846, WO2017/198741, WO2017/220555, WO2017/220569, WO2018/071500, W02017/015560, WO2017/025498, WO2017/087589, WO2017/087901, W02018/083087, WO2017/149143, WO2017/219995,
US2017/0260271, WO2017/086367, WO2017/086419, WO2018/034227, and W02014/140180, each of which is incorporated by reference herein in its entirety.
II.C.5. Anti-CD137 Antibodies
[0216] Anti-CD137 antibodies specifically bind to and activate CD137-expressing immune cells, stimulating an immune response, in particular a cytotoxic T cell response, against tumor cells. Antibodies that bind to CD137 have been disclosed in U.S. Publ. No. 2005/0095244 and U.S. Pat. Nos. 7,288,638, 6,887,673, 7,214,493, 6,303,121, 6,569,997, 6,905,685, 6,355,476, 6,362,325, 6,974,863, and 6,210,669, each of which is incorporated by reference herein in its entirety.
[0217] In some aspects, the anti-CD137 antibody is urelumab (BMS-663513), described in U.S. Pat. No. 7,288,638 (20H4.9-IgG4 [10C7 or BMS-663513]). In some aspects, the anti- CD137 antibody is BMS-663031 (20H4.9-IgGl), described in U.S. Pat. No. 7,288,638. In some aspects, the anti-CD137 antibody is 4E9 or BMS-554271, described in U.S. Pat. No. 6,887,673. In some aspects, the anti-CD137 antibody is an antibody disclosed in U.S. Pat. Nos. 7,214,493; 6,303,121; 6,569,997; 6,905,685; or 6,355,476. In some aspects, the anti-CD137 antibody is 1D8 or BMS-469492; 3H3 or BMS-469497; or 3E1, described in U.S. Pat. No. 6,362,325. In some aspects, the anti-CD137 antibody is an antibody disclosed in issued U.S. Pat. No. 6,974,863 (such as 53A2). In some aspects, the anti-CD137 antibody is an antibody disclosed in issued U.S. Pat. No. 6,210,669 (such as 1D8, 3B8, or 3E1). In some aspects, the antibody is Pfizer's PF- 05082566 (PF-2566). In other aspects, an anti-CD137 antibody useful for the methods disclosed herein cross-competes with the anti-CD137 antibodies disclosed herein. In some aspects, an anti- CD137 antibody binds to the same epitope as the anti-CD137 antibody disclosed herein. In other aspects, an anti-CD137 antibody useful in the disclosure comprises six CDRs of the anti-CD137 antibodies disclosed herein.
II.C.6. Anti-KIR Antibodies
[0218] Antibodies that bind specifically to KIR block the interaction between Killer-cell immunoglobulin-like receptors (KIR) on NK cells with their ligands. Blocking these receptors facilitates activation of NK cells and, potentially, destruction of tumor cells by the latter. Examples of anti-KIR antibodies have been disclosed in Int'l Publ. Nos. WO/2014/055648, WO 2005/003168, WO 2005/009465, WO 2006/072625, WO 2006/072626, WO 2007/042573, WO 2008/084106, WO 2010/065939, WO 2012/071411 and WO/2012/160448, each of which is incorporated by reference herein in its entirety.
[0219] One anti-KIR antibody useful in the present disclosure is lirilumab (also referred to as BMS-986015, IPH2102, or the S241P variant of 1-7F9), first described in Int'l Publ. No. WO 2008/084106. An additional anti-KIR antibody useful in the present disclosure is 1-7F9 (also referred to as IPH2101), described in Int'l Publ. No. WO 2006/003179. In one aspect, an anti- KIR antibody for the present composition cross competes for binding to KIR with lirilumab or I- 7F9. In another aspect, an anti-KIR antibody binds to the same epitope as lirilumab or I-7F9. In other aspects, an anti-KIR antibody comprises six CDRs of lirilumab or I-7F9.
II.C.7. Anti-GITR antibodies
[0220] Anti-GITR antibodies useful in the methods disclosed herein include any anti- GITR antibody that binds specifically to human GITR target and activates the glucocorticoid- induced tumor necrosis factor receptor (GITR). GITR is a member of the TNF receptor superfamily that is expressed on the surface of multiple types of immune cells, including regulatory T cells, effector T cells, B cells, natural killer (NK) cells, and activated dendritic cells ("anti-GITR agonist antibodies"). Specifically, GITR activation increases the proliferation and function of effector T cells, as well as abrogating the suppression induced by activated T regulatory cells. In addition, GITR stimulation promotes anti-tumor immunity by increasing the activity of other immune cells such as NK cells, antigen presenting cells, and B cells. Examples of anti-GITR antibodies have been disclosed in Int'l Publ. Nos. WO/2015/031667, WO2015/184,099, WO2015/026,684, WO11/028683 and WO/2006/105021, U.S. Pat. Nos. 7,812,135 and 8,388,967 and U.S. Publ. Nos. 2009/0136494, 2014/0220002, 2013/0183321 and 2014/0348841, each of which is incorporated by reference herein in its entirety. [0221] In one aspect, an anti-GITR antibody useful in the present disclosure is TRX518 (described in, for example, Schaer et al. Curr Opin Immunol. (2012) Apr; 24(2): 217-224, and WO/2006/105021). In another aspect, the anti-GITR antibody is selected from MK4166, MK1248, and antibodies described in WO11/028683 and U.S. 8,709,424, and comprising, e.g., a VH chain comprising SEQ ID NO: 104 and a VL chain comprising SEQ ID NO: 105 (wherein the SEQ ID NOs are from WO11/028683 or U.S. 8,709,424). In certain aspects, an anti-GITR antibody is an anti-GITR antibody that is disclosed in WO2015/031667, e.g., an antibody comprising VH CDRs 1-3 comprising SEQ ID NOs: 31, 71 and 63 of WO2015/031667, respectively, and VL CDRs 1-3 comprising SEQ ID NOs: 5, 14 and 30 of WO2015/031667. In certain aspects, an anti-GITR antibody is an anti-GITR antibody that is disclosed in WO2015/184099, e.g., antibody Hum231#l or Hum231#2, or the CDRs thereof, or a derivative thereof (e.g., pabl967, pabl975 or pabl979). In certain aspects, an anti-GITR antibody is an anti- GITR antibody that is disclosed in JP2008278814, W009/009116, WO2013/039954, US20140072566, US20140072565, US20140065152, or WO2015/026684, or is INBRX-110 (INHIBRx), LKZ-145 (Novartis), or MEDI-1873 (Medlmmune). In certain aspects, an anti-GITR antibody is an anti-GITR antibody that is described in PCT/US2015/033991 (e.g., an antibody comprising the variable regions of 28F3, 18E10 or 19D3).
[0222] In certain aspects, the anti-GITR antibody cross-competes with an anti-GITR antibody described herein, e.g., TRX518, MK4166 or an antibody comprising a VH domain and a VL domain amino acid sequence described herein. In some aspects, the anti-GITR antibody binds the same epitope as that of an anti-GITR antibody described herein, e.g., TRX518 or MK4166. In certain aspects, the anti-GITR antibody comprises the six CDRs of TRX518 or MK4166.
II.C.8. Anti-TIM3 antibodies
[0223] Any anti-TIM3 antibody or antigen binding fragment thereof known in the art can be used in the methods described herein. In some aspects, the anti-TIM3 antibody is be selected from the anti-TIM3 antibodies disclosed in Int'l Publ. Nos.W02018013818, WO/2015/117002 (e.g., MGB453, Novartis), WO/2016/161270 (e.g., TSR-022, Tesaro/AnaptysBio),
WO201 1155607, WO2016/144803 (e.g., STI-600, Sorrento Therapeutics), WO2016/071448, WO17055399; W017055404, WO17178493, WO18036561, W018039020 (e.g., Ly-3221367, Eli Lilly), WO2017205721, WO17079112; WO17079115; WO17079116, WO11159877, W013006490, W02016068802 W02016068803, WO2016/111947, and WO/2017/031242, each of which is incorporated by reference herein in its entirety. II.C.9. Anti-OX40 antibodies
[0224] Any antibody or antigen-binding fragment thereof that specifically binds 0X40 (also known as CD 134, TNFRSF4, ACT35 and/or TXGP1L) can be used in the methods disclosed herein. In some aspects, the anti-OX40 antibody is BMS-986178 (Bristol-Myers Squibb Company), described in Int'l Publ. No. WO20160196228. In some aspects, the anti- 0X40 antibody is selected from the anti-OX40 antibodies described in Int'l Publ. Nos. WO95012673, WO199942585, WO14148895, WO15153513, WO15153514, WO13038191, WO16057667, WO03106498, WO12027328, WO13028231, W016200836, WO 17063162, WO17134292, WO 17096179, WO 17096281, and WO 17096182, each of which is incorporated by reference herein in its entirety.
II.C.10. Anti-NKG2A Antibodies
[0225] Any antibody or antigen-binding fragment thereof that specifically binds NKG2A can be used in the methods disclosed herein. NKG2A is a member of the C-type lectin receptor family that is expressed on natural killer (NK) cells and a subset of T lymphocytes. Specifically, NKG2A primarily expressed on tumor infiltrating innate immune effector NK cells, as well as on some CD8+ T cells. Its natural ligand human leukocyte antigen E (HLA-E) is expressed on solid and hematologic tumors. NKG2A is an inhibitory receptor that blinds HLA-E.
[0226] In some aspects, the anti-NKG2A antibody may be BMS-986315, a human monoclonal antibody that blocks the interaction of NKG2A to its ligand HLA-E, thus allowing activation of an anti-tumor immune response. In some aspects, the anti-NKG2A antibody is a checkpoint inhibitor that activates T cells, NK cells, and/or tumor-infiltrating immune cells. In some aspects, the anti-NKG2A antibody is selected from the anti-NKG2A antibodies described in, for example, WO 2006/070286 (Innate Pharma S.A.; University of Genova); U.S. Patent No. 8,993,319 (Innate Pharma S.A.; University of Genova); WO 2007/042573 (Innate Pharma S/A; Novo Nordisk A/S; University of Genova); U.S. Patent No. 9,447,185 (Innate Pharma S/A; Novo Nordisk A/S; University of Genova); WO 2008/009545 (Novo Nordisk A/S); US. Patent Nos. 8,206,709; 8,901,283; 9,683,041 (Novo Nordisk A/S); WO 2009/092805 (Novo Nordisk A/S); U.S. Patent Nos. 8,796,427 and 9,422,368 (Novo Nordisk A/S); WO 2016/134371 (Ohio State Innovation Foundation); WO 2016/032334 (Janssen); WO 2016/041947 (Innate); WO 2016/041945 (Academisch Ziekenhuis Leiden H.O.D.N. LUMC); WO 2016/041947 (Innate Pharma); and WO 2016/041945 (Innate Pharma), each of which is incorporated by reference herein in its entirety.
II.C.11. Anti-ICOS Antibodies [0227] Any antibody or antigen-binding fragment thereof that specifically binds ICOS can be used in the methods disclosed herein. ICOS is an immune checkpoint protein that is a member of the CD28-superfamily. ICOS is a 55-60 kDa type I transmembrane protein that is expressed on T cells after T cell activation and co-stimulates T-cell activation after binding its ligand, ICOS-L (B7H2). ICOS is also known as inducible T-cell co-stimulator, CVID1, AILIM, inducible costimulator, CD278, activation-inducible lymphocyte immunomediatory molecule, and CD278 antigen.
[0228] In some aspects, the anti-ICOS antibody is BMS-986226, a humanized IgG monoclonal antibody that binds to and stimulates human ICOS. In some aspects, the anti-ICOS antibody is selected from anti-ICOS antibodies described in, for example, WO 2016/154177 (Jounce Therapeutics, Inc.), WO 2008/137915 (Medlmmune), WO 2012/131004 (INSERM, French National Institute of Health and Medical Research), EP3147297 (INSERM, French National Institute of Health and Medical Research), WO 2011/041613 (Memorial Sloan Kettering Cancer Center), EP 2482849 (Memorial Sloan Kettering Cancer Center), WO 1999/15553 (Robert Koch Institute), U.S. Patent Nos. 7,259,247 and 7,722,872 (Robert Kotch Institute); WO 1998/038216 (Japan Tobacco Inc.), US. Patents. Nos. 7,045,615; 7,112,655, and 8,389,690 (Japan Tobacco Inc.), U.S. Patent Nos. 9,738,718 and 9,771,424 (GlaxoSmithKline), and WO 2017/220988 (Kymab Limited), each of which is incorporated by reference herein in its entirety.
II.C.12. Anti-TIGIT Antibodies
[0229] Any antibody or antigen-binding fragment thereof that specifically binds TIGIT can be used in the methods disclosed herein. In some aspects, the anti-TIGIT antibody is BMS- 986207. In some aspects, the anti-TIGIT antibody is clone 22G2, as described in WO 2016/106302. In some aspects, the anti-TIGIT antibody is MTIG7192A/RG6058/RO7092284, or clone 4.1D3, as described in WO 2017/053748. In some aspects, the anti-TIGIT antibody is selected from the anti-TIGIT antibodies described in, for example, WO 2016/106302 (Bristol- Myers Squibb Company) and WO 2017/053748 (Genentech).
II.C.13. Anti-CSFIR Antibodies
[0230] Any antibody or antigen-binding fragment thereof that specifically binds CSF1R can be used in the methods disclosed herein. In some aspects, the anti-CSFIR antibody is an antibody species disclosed in any of international publications WO2013/132044, W02009/026303, WO2011/140249, or W02009/112245, such as cabiralizumab, RG7155 (emactuzumab), AMG820, SNDX 6352 (UCB 6352), CXIIG6, IMC-CS4, JNJ-40346527, MCS1 10, or the anti-CSFIR antibody in the methods is replaced with an anti-CSFIR inhibitor or anti-CSFl inhibitor such as BLZ-945, pexidartinib (PLX3397, PLX108-01), AC-708, PLX-5622, PLX7486, ARRY-382, or PLX-73086.
II.D. Additional Anti-Cancer Therapies
[0231] In some aspects of the present disclosure, the methods disclosed herein comprise administering an 1-0 therapy, e.g., an anti-PD-1 antibody or an anti-PD-Ll antibody, and one or more additional anti-cancer therapies. In certain aspects, the method comprising administering (i) a first 1-0 therapy, e.g., an anti-PD-1 antibody or an anti-PD-Ll antibody), (ii) a second 1-0 therapy, e.g., an anti-CTLA-4 antibody or an anti-CSFIR antibody, and (iii) one or more additional anti-cancer therapies. In some aspects, the methods dislosed herein comprise administering an anti-cancer therapy to a subject identified as having a low pan-tumor gene signature score, wherein the anti-cancer agent does not comprise an 1-0 therapy.
[0232] The anti-cancer therapy can comprise any therapy known in the art for the treatment of a tumor in a subject and/or any standard-of-care therapy, as disclosed herein. In some aspects, the anti-cancer therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof. In some aspects, the anti-cancer therapy comprises a chemotherapy, including any chemotherapy disclosed herein.
[0233] Any chemotherapy known in the art can be used in the methods disclosed herein. In some aspects, the chemotherapy is a platinum based-chemotherapy. Platinum-based chemotherapies are coordination complexes of platinum. In some aspects, the platinum-based chemotherapy is a platinum-doublet chemotherapy. In some aspects, the chemotherapy is administered at the approved dose for the particular indication. In other aspects, the chemotherapy is administered at any dose disclosed herein. In some aspects, the platinum-based chemotherapy is cisplatin, carboplatin, oxaliplatin, satraplatin, picoplatin, Nedaplatin, Triplatin, Lipoplatin, or combinations thereof. In certain aspects, the platinum-based chemotherapy is any other platinum-based chemotherapy known in the art. In some aspects, the chemotherapy is the nucleotide analog gemcitabine. In an aspect, the chemotherapy is a folate antimetabolite. In an aspect, the folate antimetabolite is pemetrexed. In certain aspects the chemotherapy is a taxane. In other aspects, the taxane is paclitaxel. In some aspects, the chemotherapy is any other chemotherapy known in the art. In certain aspects, at least one, at least two or more chemotherapeutic agents are administered in combination with the 1-0 therapy. In some aspects, the 1-0 therapy is administered in combination with gemcitabine and cisplatin. In some aspects, the 1-0 therapy is administered in combination with pemetrexed and cisplatin. In certain aspects, the 1-0 therapy is administered in combination with gemcitabine and pemetrexed. In one aspect, the 1-0 therapy is administered in combination with paclitaxel and carboplatin. In an aspect, an I- O therapy is additionally administered.
[0234] In some aspects, the additional anti-cancer therapy, e.g., administered to a subject having a moderate pan-tumor gene signature score, comprises an immunotherapy. In some aspects, the additional anti-cancer therapy comprises administration of an antibody or antigenbinding portion thereof that specifically binds LAG-3, TIGIT, TIM3, NKG2a, CSF1R, 0X40, ICOS, MICA, MICB, CD137, KIR, TGFp, IL-10, IL-8, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, or any combination thereof.
ILE. Tumors
[0235] In some aspects, the tumor is derived from a cancer selected from the group consisting of hepatocellular cancer, gastroesophageal cancer, melanoma, bladder cancer, lung cancer (e.g., non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC)), kidney cancer, renal cell carcinoma, head and neck cancer (e.g., squamous cell carcinoma of the head and neck), pancreatic cancer, gastric cancer, ovarian cancer, prostate cancer, colon cancer, colorectal cancer, urothelial carcinoma, and any combination thereof. In certain aspects, the tumor is derived from a hepatocellular cancer, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a gastroesophageal cancer, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a melanoma, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a bladder cancer, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a lung cancer, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a kidney cancer, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a head and neck cancer, wherein the tumor has a high inflammatory signature score. In certain aspects, the tumor is derived from a colon cancer, wherein the tumor has a high inflammatory signature score.
[0236] In certain aspects, the subject has received one, two, three, four, five or more prior cancer treatments. In other aspects, the subject is treatment-naive. In some aspects, the subject has progressed on other cancer treatments. In certain aspects, the prior cancer treatment comprised an immunotherapy. In other aspects, the prior cancer treatment comprised a chemotherapy. In some aspects, the tumor has reoccurred. In some aspects, the tumor is metastatic. In other aspects, the tumor is not metastatic. In some aspects, the tumor is locally advanced.
[0237] In some aspects, the subject has received a prior therapy to treat the tumor and the tumor is relapsed or refractory. In certain aspects, the at least one prior therapy comprises a standard-of-care therapy. In some aspects, the at least one prior therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof. In some aspects, the at least one prior therapy comprises a chemotherapy. In some aspects, the subject has received a prior immuno-oncology (I-O) therapy to treat the tumor and the tumor is relapsed or refractory. In some aspects, the subject has received more than one prior therapy to treat the tumor and the subject is relapsed or refractory. In other aspects, the subject has received either an anti-PD-1 or anti-PD-Ll antibody therapy.
[0238] In some aspects, the previous line of therapy comprises a chemotherapy. In some aspects, the chemotherapy comprises a platinum-based therapy. In some aspects, the platinumbased therapy comprises a platinum-based antineoplastic selected from the group consisting of cisplatin, carboplatin, oxaliplatin, nedaplatin, triplatin tetranitrate, phenanthriplatin, picoplatin, satraplatin, and any combination thereof. In certain aspects, the platinum-based therapy comprises cisplatin. In one particular aspect, the platinum-based therapy comprises carboplatin.
[0239] In some aspects, the at least one prior therapy is selected from a therapy comprising administration of an anti-cancer agent selected from the group consisting of a platinum agent (e.g., cisplatin, carboplatin), a taxanes agent (e.g., paclitaxel, albumin-bound paclitaxel, docetaxel), vinorelbine, vinblastine, etoposide, pemetrexed, gemcitabine, bevacizumab (AVASTIN®), erlotinib (TARCEVA®), crizotinib (XALKORI®), cetuximab (ERBITUX®), and any combination thereof. In certain aspects, the at least one prior therapy comprises a platinum-based doublet chemotherapy.
[0240] In some aspects, the subject has experienced disease progression after the at least one prior therapy. In certain aspects, the subject has received at least two prior therapies, at least three prior therapies, at least four prior therapies, or at least five prior therapies. In certain aspects, the subject has received at least two prior therapies. In one aspect, the subject has experienced disease progression after the at least two prior therapies. In certain aspects, the at least two prior therapies comprises a first prior therapy and a second prior therapy, wherein the subject has experienced disease progression after the first prior therapy and/or the second prior therapy, and wherein the first prior therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof; and wherein the second prior therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof. In some aspects, the first prior therapy comprises a platinum-based doublet chemotherapy, and the second prior therapy comprises a single-agent chemotherapy. In certain aspects, the single-agent chemotherapy comprises docetaxel.
ILF. Pharmaceutical Compositions and Dosages
[0241] Therapeutic agents of the present disclosure can be constituted in a composition, e.g., a pharmaceutical composition containing an antibody and/or a cytokine and a pharmaceutically acceptable carrier. As used herein, a "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier for a composition containing an antibody is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration (e.g., by injection or infusion), whereas the carrier for a composition containing an antibody and/or a cytokine is suitable for non-parenteral, e.g., oral, administration. In some aspects, the subcutaneous injection is based on Halozyme Therapeutics’ ENHANZE® drug-delivery technology (see U.S. Patent No. 7,767,429, which is incorporated by reference herein in its entirety). ENHANZE® uses a coformulation of an antibody with recombinant human hyaluronidase enzyme (rHuPH20), which removes traditional limitations on the volume of biologies and drugs that can be delivered subcutaneously due to the extracellular matrix (see U.S. Patent No. 7,767,429). A pharmaceutical composition of the disclosure can include one or more pharmaceutically acceptable salts, antioxidant, aqueous and non-aqueous carriers, and/or adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Therefore, in some aspects, the pharmaceutical composition for the present disclosure can further comprise recombinant human hyaluronidase enzyme, e.g., rHuPH20.
[0242] Although higher nivolumab monotherapy dosing up to 10 mg/kg every two weeks has been achieved without reaching the maximum tolerated does (MTD), the significant toxi cities reported in other trials of checkpoint inhibitors plus anti-angiogenic therapy (see, e.g., Johnson el al., 2013; Rini et al., 2011) support the selection of a nivolumab dose lower than 10 mg/kg.
[0243] Treatment is continued as long as clinical benefit is observed or until unacceptable toxicity or disease progression occurs. Nevertheless, in certain aspects, the antibodies disclosed herein are administered at doses that are significantly lower than the approved dosage, i.e., a subtherapeutic dosage, of the agent. The antibody can be administered at the dosage that has been shown to produce the highest efficacy as monotherapy in clinical trials, e.g., about 3 mg/kg of nivolumab administered once every three weeks (Topalian et al., 2012a; Topalian et al., 2012), or at a significantly lower dose, i.e., at a subtherapeutic dose.
[0244] Dosage and frequency vary depending on the half-life of the antibody in the subject. In general, human antibodies show the longest half-life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies. The dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is typically administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives. In therapeutic applications, a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
[0245] Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present disclosure can be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being unduly toxic to the patient. The selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present disclosure employed, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A composition of the present disclosure can be administered via one or more routes of administration using one or more of a variety of methods well known in the art. As will be appreciated by the skilled artisan, the route and/or mode of administration will vary depending upon the desired results.
III. Kits
[0246] Also within the scope of the present disclosure are kits comprising (a) an anti-PD- 1 antibody or an anti-PD-Ll antibody for therapeutic uses. Kits typically include a label indicating the intended use of the contents of the kit and instructions for use. The term label includes any writing, or recorded material supplied on or with the kit, or which otherwise accompanies the kit. Accordingly, this disclosure provides a kit for treating a subject afflicted with a tumor, the kit comprising: (a) a dosage ranging from 0.1 to 10 mg/kg body weight of an anti-PD-1 antibody or a dosage ranging from 0.1 to 20 mg/kg body weight of an anti-PD-Ll antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-Ll antibody in the methods disclosed herein. This disclosure further provides a kit for treating a subject afflicted with a tumor, the kit comprising: (a) a dosage ranging from about 4 mg to about 500 mg of an anti-PD-1 antibody or a dosage ranging from about 4 mg to about 2000 mg of an anti-PD-Ll antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-Ll antibody in the methods disclosed herein. In some aspects, this disclosure provides a kit for treating a subject afflicted with a tumor, the kit comprising: (a) a dosage ranging from 200 mg to 800 mg of an anti-PD-1 antibody or a dosage ranging from 200 mg to 1800 mg of an anti-PD-Ll antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-Ll antibody in the methods disclosed herein.
[0247] In certain aspects for treating human patients, the kit comprises an anti-human PD-1 antibody disclosed herein, e.g., nivolumab or pembrolizumab. In certain aspects for treating human patients, the kit comprises an anti-human PD-L1 antibody disclosed herein, e.g., atezolizumab, durvalumab, or avelumab.
[0248] In some aspects, the kit further comprises an anti-CTLA-4 antibody. In certain aspects for treating human patients, the kit comprises an anti-human CTLA-4 antibody disclosed herein, e.g., ipilimumab, tremelimumab, MK-1308, or AGEN-1884.
[0249] In some aspects, the kit further includes a gene panel assay disclosed herein. In some aspects, the kit further includes instructions to administer the anti-PD-1 antibody or the anti-PD-Ll antibody to a suitable subject according to the methods disclosed herein.
[0250] All of the references cited above, as well as all references cited herein, are incorporated herein by reference in their entireties.
[0251] The following examples are offered by way of illustration and not by way of limitation.
EXAMPLES
Example 1
[0252] Across multiple tumor types, an inflammatory phenotype in the tumor microenvironment (TME) has been associated with improved clinical efficacy in patients treated with immuno-oncology (1-0) therapies (see Darvin P, et al. Exp Mol Med 2018;50: 165). Infiltration of CD8+ T cells can be used as a surrogate marker for inflammation and is often assessed using immunohistochemistry (IHC) (see, e.g., Barnes et al, Br J Cancer 2017; 117:451- 460; and Stoll et al. Oncotarget 2015;6: 11894-11909). Interrogation of multiple biomarkers by IHC has its limitations. Simultaneous analysis of multiple transcripts in the TME using gene expression profiling (GEP) may provide a robust characterization of inflammation.
[0253] A number of GEP platforms are available, including RNA sequencing (RNA-seq) and GEP panels targeting select sets of genes or pathways. However, cross-platform consistency remains to be determined.
[0254] Gene expression signatures indicative of inflammation in the TME have been derived using GEP from tumor samples in a number of studies (see, e.g., Szabo et al. J Clin Oncol 2019;37(suppl) Abstract 2593; Szabo et al. J Immunother Cancer 2019;7(suppl 1) Abstract P109; Danaher et al. J Immunother Cancer 2017;5(18); Danaher et al. J Immunother Cancer 2018;6(63); Cristescu et al. Science 2018;362:eaar3593).
[0255] A 16-gene expression signature (CD8 signature) has been developed as an investigational use only assay that correlated with pathology-assisted digital scoring of CD8 expression by IHC (CD8 IHC) across multiple tumor types (Szabo et al. J Clin Oncol 2019;37(suppl) Abstract 2593). The CD8 signature was associated with clinical efficacy in post- hoc analyses of samples from patients with urothelial carcinoma treated with nivolumab in a clinical trial for the treatment of urothelial carcinoma (Szabo et al. J Immunother Cancer 2019;7(suppl 1) Abstract Pl 09).
[0256] Other GEP panels include a 2-gene T-cell abundance (TCA) signature, which can be used as a measure of CD8+ T-cell infiltration in the TME (see Danaher et al. J Immunother Cancer 2017;5(l 8)) and an 18-gene tumor inflammation signature (TIS), which was associated with inflammatory responses and clinical efficacy of 1-0 therapy in post-hoc analyses across multiple tumor types (see Danaher et al. J Immunother Cancer 2018;6(63); Cristescu et al. Science 2018;362:eaar3593).
[0257] The present example presents data from a study designed to assess correlations between gene expression signatures and other predictive biomarkers of response to immune checkpoint inhibitors. In addition, this study compared CD8 IHC with three different gene expression signatures to assess their suitability as surrogate biomarkers of inflammation; investigated the relationship between gene expression signatures and programmed death ligand 1 (PD-L1) expression on tumor cells, assessed using IHC; and evaluated the consistency of the CD8 signature derived from tumor samples using RNA-seq and four different gene expression panels.
[0258] Methods
[0259] Formalin-fixed, paraffin-embedded (FFPE) tumor samples (melanoma [n = 97] and squamous cell carcinoma of the head and neck [n = 101]) were commercially procured. Tissue quality and diagnosis of tumor type were verified on hematoxylin and eosin-stained sections by a pathologist prior to GEP.
[0260] CD8 signature scores were derived using data from RNA-seq and the four GEP panels. RNA-seq was used as a reference to derive Pearson’s correlation coefficients (r) for cross-platform comparisons. The TCA (Danaher P, et al. J Immunother Cancer 2017;5(l 8)) and TIS (Danaher P, et al. J Immunother Cancer 2018;6(63)) scores were derived using data from the IO 360 panel.
[0261] CD8 IHC was performed using a monoclonal CD8 antibody. CD8+ T cells were quantified using pathologist-supervised, digital scoring-based analysis of 20* whole slide images. Scores were calculated from CD8+ T cells as a percentage of total cells in the tumor parenchyma, tumor stroma, and tumor margin. Melanoma (MEL) samples were labelled with an alkaline phosphatase (AP)-linked secondary antibody (PowerVision Poly-AP Anti-Mouse IgG; Leica Biosystems, Buffalo Grove, IL) and visualized using Fast Red chromogen (Leica). All other tumor types were labeled with Envision FLEX+ Mouse LINKER antibody and horseradish peroxidase polymer (Agilent) and visualized with 3, 3 '-diaminobenzidine (DAB).
[0262] Tissues stained using Fast Red and DAB were evaluated using a Nuclear v9 Mosaic Red-Melanin MTM UT220-vl and Nuclear v9 MTM UT 220 algorithms, respectively (both Aperio). For comparisons between CD8 signature scores and CD8 IHC, CD8 IHC scores were square-root transformed to meet linear model distribution assumptions. PD-L1 expression on tumor cells was assessed using the Dako PD-L1 IHC 28-8 pharmDx assay (Agilent Technologies, Santa Clara, CA). PD-Ll-positive (PD-L1+) and PD-L1 -negative (PD-L1-) status were determined around a cutoff of PD-L1 expression on 1% of tumor cells. Pearson’s correlation coefficients (r) were derived from comparisons between gene signature scores and CD8 IHC or PD-L1 IHC.
[0263] For macrodissection, prior to GEP, a region of interest (ROI) was defined by a pathologist to include tumor parenchyma and contiguous tumor-associated stroma within 500 pm of the edge of the tumor. Sample characteristics (including tumor grade, biopsy type, tumor content, percent necrosis) were recorded for both the entire section and the ROI (data available on request). Tissue outside the ROI was then removed, and remaining samples were scraped into sterile 1.7 mL nuclease-free microcentrifuge tubes using a sterile razor blade.
[0264] GEP Platforms
[0265] Inflammation gene signatures were determined using RNA-seq and 3 alternative GEP panel assays (Tumor Immunology Panel [TIP], PanCancer IO 360™ Panel, and Oncomine™ Immune Response Research Assay) (Table XX).
Table XX: GEP Platform Summary.
Figure imgf000080_0001
GEP, gene expression profiling; NGS, next-generation sequencing; RNA-seq, RNA sequencing.
[0266] RNA-seq
[0267] RNA was extracted and pooled from fifteen unstained sections from commercially procured FFPE samples using the AllPrep RNA/DNA FFPE Kit (Qiagen, Germantown, MD). RNA-seq was performed using the TruSeq™ RNA Exome (targeting 21,415 genes; Illumina, San Diego, CA). Paired-end sequence reads in FASTQ format were aligned to the GRCh37 human reference genome (version 75) and External RNA Control Consortium spike-in control sequences using STAR v2.6.0c in two-pass mode with default parameters. RSEM vl.3.0 was used for abundance estimation for transcriptome assembly. Pre- and post-alignment, QC metrics were assessed using Picard vl.140. The Trimmed Mean of M-values (TMM) normalization of transcript counts was implemented with the EdgeR package (v5.2.645.3826) in R, and transformed to log2 TPM.
[0268] TIP
[0269] The CD8 signature was developed and analytically validated as an IUO assay using the custom TIP. This EdgeSeq (HTG Molecular Diagnostics, Tucson, AZ) panel comprises 95 probes, including genes related to tumor inflammation, other 1-0 genes of interest, housekeeping genes, and control probes. mRNA expression levels of all 95 probes on the panel were determined using an automated, extraction-free, nuclease protection assay, combined with NGS.
[0270] A minimum sample area of 7.2 mm2 macrodissected tissue from an unstained section was required for GEP using the TIP (tissue from more than one section was used in some cases). Tissues were lysed in lysis buffer (HTG Molecular Diagnostics; 50 pL per 7.2 mm2 tissue) and incubated for 15-20 minutes at 95°C. Proteinase K was added to a final concentration of 1 mg/mL, and samples were incubated for 3 hours at 50°C with shaking. Each sample was then added to a well of a 96-well plate. Nuclease protection probes were added to the lysed samples and the nuclease protection step was performed using an HTG EdgeSeq Processor with controller software version v6.1.653.2205. SI nuclease was then added to degrade nonhybridized probes and nontargeted RNA, followed by heat inactivation (85°C, 20 minutes, pH 9). Samples were then individually barcoded using a 16-cycle PCR to add adapters and molecular barcodes. Barcoded samples were individually purified using Agencourt AMPure XP beads (Beckman Coulter, Indianapolis, IN; catalog number A63880) and quantitated using a KAPA Library Quantification kit (Kapa Biosciences, Wilmington, MA; catalog number KK4873). PCR- enriched libraries were sequenced using a MiSeqDx with controller software version v2.5.05 (Illumina).
[0271] NGS data were processed using HTG EdgeSeq Host System Software (HTG Molecular Diagnostics). Demultiplexed FASTQ files were parsed and aligned to probe FASTQ files and raw counts were transformed to log2 counts per million (CPM). No additional normalization was applied. Normalized data were filtered based on quality control metrics examined in the following order: input RNA quality (samples with reads for positive control probes < 21% of total reads passed this filter), sequencing read depth (cutoff varied by number of samples in the assay plate), and samples with minimum expression variability (samples passed if the relative standard deviation of log2 CPM among 77 probes was > 0.19).
[0272] PanCancer IO 360™ Panel
[0273] The 18-gene Tumor Inflammation Signature (TIS) and 2-gene CD8+ T-cell abundance (TCA) signature were previously developed using the NanoString gene expression platform (NanoString Technologies, Seattle, WA). In this study, GEP was performed by Expression Analysis, Inc. (Durham, NC) on the same pool of extracted RNA as for RNA-seq, using the NanoString PanCancer IO 360™ Gene Expression Panel, targeting 770 genes. Data resulting from the NanoString nCounter® system were expressed as number of molecules (counts), per standardized sample input. Background correction was performed, and counts were normalized to housekeeping genes using nSolver™ software (v4.0.70; NanoString Technologies). Data were subsequently transformed to log2 scale.
[0274] Oncomine™ Immune Response Research Assay
[0275] GEP was performed on the same pool of extracted RNA as for RNA-seq, using the Oncomine™ Immune Response Research Assay (Thermo Fisher Scientific, Waltham, MA), targeting 395 genes according to manufacturer’s instructions. Absolute digital gene expression counts were generated using Torrent Suite software (v5.0.2) with the immuneResponseRNA plugin (Thermo Fisher Scientific). TMM normalization was applied using the EdgeSeq Host System Software (HTG Molecular Diagnostics), and normalized data were transformed to log2 CPM.
[0276] Derivation of the CD8 Signature
[0277] A machine-learning method utilized a multivariate regression model to develop a classifier with 63 probes on the TIP as the predictors and CD8 IHC as the outcome variable. The combination of genes that gave the best correlation with CD8 IHC using MEL (n = 335) and squamous cell carcinoma of the head and neck (SCCHN, n = 392) samples was determined (data for 70 samples failed quality control filters). Tumor-specific signatures were derived independently for MEL and SCCHN samples. A similar approach was then applied to pooled MEL and SCCHN samples to derive a combined MEL/SCCHN signature. The least absolute shrinkage and selection operator (LASSO) technique, with optimal shrinkage parameters for lambda.min and lambda. Ise, was applied to variable selection to derive a multitumor 16-gene expression model (CD8 signature) for predicting CD8 IHC across 10 additional tumor types. The CD8 signature was then validated using data from 1633 (92%) samples that passed quality control steps. This set included data from patients with colorectal cancer (n = 96), gastric cancer (GC, n = 90), hepatocellular carcinoma (HCC, n = 95), MEL (n = 335), non-small cell lung cancer (NSCLC, n = 91), ovarian cancer (OVC, n = 97), pancreatic cancer (PANC, n = 78), prostate cancer (PC, n = 86), renal cell carcinoma (n = 91), SCCHN (n = 392), small cell lung cancer (SCLC, n = 87), and urothelial carcinoma (n = 95) (FIGs. 1 A-1C).
[0278] Calculation of Signature Scores
[0279] Scores for the CD8 signature, TIS, and TCA signature were derived from the gene expression of each of the genes listed in Table XX. Each gene in the CD8 signature was assigned a weighting coefficient, and the score was calculated as the sum of each weighted gene expression (FIG. ID). TIS and TCA signature scores were calculated as previously described, using automated nSolver analysis of data from the PanCancer IO 360™ Panel, and no additional processing was applied.
Supplementary Table XX: Gene Signatures Assessed in This Study
Figure imgf000083_0001
TCA, T-cell abundance; TIS, tumor inflammation signature.
[0280] Statistical Analyses
[0281] All data analyses were performed with R 3.4.1 for Linux and graphical representations were generated using ggplot v3.2.1. For comparisons between CD8 signature scores and CD8 IHC, the 95% confidence interval (CI) for prediction of CD8 IHC was computed from the multiple linear regression model obtained using MEL and SCCHN samples. The model was considered accurate if ~ 95% of samples were contained within this CI.
[0282] Relationships between CD8 IHC and gene expression signature scores were investigated by Pearson’s correlation (r) for each tumor type. To compare inflammation prevalence determined by GEP and IHC, a predictor model was developed to illustrate inflammation prevalence when CD8 signature and CD8 IHC scores were expressed on a continuous scale. Samples were classified as inflammation-positive or inflammation-negative above or below a cutoff applied to the CD8 signature score. Reference inflammation status (positive or negative) was derived by applying the same cutoff to the observed CD8 IHC score. Associations between PD-L1 status and gene expression signature scores were determined using the Wilcoxon-Mann-Whitney test.
[0283] Assay reproducibility and precision across plates, operators, processors, lots, and days were assessed using the TIP to derive CD8 signature scores from 20 replicates per sample across 8 samples from each of 12 tumor types. Intraclass correlation coefficients (ICC) were derived using a mixed model approach. Assay precision was further assessed in 1405 samples (15 replicates of the same sample across 8 samples from each of 12 tumor types; 35 samples failed quality control). Sample replicates were randomized across plate, processor, reagent lot, and processing day. CD8 signature scores for each sample replicate were used to construct a precision profile that defined the relationship between the CD8 signature score and the percent coefficient of variation (% CV).
[0284] Kaplan-Meier plots were used to illustrate associations between CD8 signature scores and survival outcomes from CheckMate 275 (progression-free survival [PFS] and overall survival [OS]), based on retrospective categorization of patients with CD8 signature scores in the lower (scores < 7.9), middle (7.9-12.6), or upper (> 12.6) tertiles.
[0285] Results
[0286] Analytical Validation of the CD8 Signature to Predict CD8+ T-cell Abundance in Multiple Tumor Types
[0287] From an initial set of 1778 samples, CD8+ T-cell abundance was assessed by GEP and IHC in parallel using 1633 matched samples from a total of 12 tumor types. Reasons for sample attrition included insufficient RNA, insufficient read depth, or low variation in gene expression. A machine-learning approach was used to derive gene expression signatures that correlated with CD8+ T-cell abundance in the TME by IHC in MEL (n = 335) and SCCHN (n = 392) samples (FIGs. 1A-1B). Based on these initial analyses, a 16-gene CD8 signature was derived, and its ability to predict CD8 IHC was tested in 10 additional tumor types. The range of CD8 expression, via CD8 IHC and the GEP-derived CD8 signature score, was consistent across most of the tumor types investigated (FIG. 2). MEL, SCCHN, NSCLC, and GC samples showed higher median CD8 scores and wider ranges by both IHC and GEP. Lower ranges and median scores by GEP were observed in tumor types with lower CD8 expression by IHC (SCLC, OVC, and PC) (FIG. 2). [0288] CD8 signature scores correlated with CD8 IHC scores across multiple tumor types (FIGs. 3A-3L). Pearson’s r values, reflecting variance in CD8 IHC associated with CD8 signature score, were > 0.82 (P < 0.0001) for all tumor types except PANC (r = 0.79) and PC (r = 0.57). Approximately 95% of CD8 IHC/CD8 signature scores fell within the 95% CI derived from the MEL/SCCHN model, except for SCLC (for SCLC samples, 77% of scores fell within the 95% CI), demonstrating that the CD8 signature accurately predicted CD8 IHC across multiple tumor types. Inflammation prevalence, characterized by the presence of CD8+ T cells in the TME, was determined across the 12 tumor types using CD8 IHC and the CD8 signature as surrogate assays for inflammation. A continuous model was applied to facilitate the prediction of inflammation prevalence around clinically relevant cutoffs that may vary across tumor types. FIGs. 4A-4L shows the proportion of samples with CD8 IHC scores or CD8 signature scores above cutoff values that varied from 0-40. Across multiple tumor types, prevalence was similar between assay methods and depended on the cutoff used to define “CD8-positive”. In SCLC and PC samples, however, prevalence according to the different assay methods was offset, reflecting the differences in the range of CD8 IHC and CD8 signature scores seen in these samples (FIG. 2).
[0289] Assay Reproducibility and Precision
[0290] A study was designed to investigate the reproducibility and precision of the CD8 signature assay across plates, operators, processors, lots, and days. Overall precision across MEL and SCCHN replicate samples was high (ICC = 0.97; n = 315). Across 5 plates, intra-plate precision ranged from 0.98-1.00. ICC for inter-operator/processor and intra-lot/day was 0.98. High ICCs were maintained in further precision studies across multiple tumor types (data not shown).
[0291] To further demonstrate assay precision, CD8 signature scores were assessed in 15 replicates from 8 samples per tumor type. Samples that were representative of the dynamic range of inflammation seen in each of the 12 tumor types were selected. Across all tumor types, % CV decreased as a function of increasing CD8 signature score, and variation was reduced to < 25 in samples with CD8 signature scores > 5 (FIG. 5).
[0292] Comparison of the CD8 Signature Scores Derived Using Different GEP Platforms
[0293] The consistency of the CD8 signature was evaluated across 4 different GEP platforms using replicate samples from a subset of patients with MEL (n = 97) and SCCHN (n = 101). FIGs. 6A-6Q show correlations between gene expression data for the 16 genes in the CD8 signature, derived using RNA-seq vs one of 3 different GEP panels. Median Pearson’s r and interquartile ranges for individual gene expression were comparable across the 3 GEP panels [0.87 (0.78-0.94), 0.82 (0.66-0.86), and 0.71 (0.44-0.83) for the PanCancer IO 360™ Panel, Oncomine™ Immune Response Research Assay, and TIP, respectively]. Whereas high correlations with RNA-seq were observed across different GEP platforms for genes with a wide range of expression across samples, low or variable correlations were seen for genes where probe performance by RNA-seq or GEP panel was inconsistent (eg, CXCR1, TNFSF18, LGALS9). Panel-derived CD8 signature scores, calculated using the weighted expression of all 16 genes in the signature, correlated with those derived using RNA-seq for all panels (r > 0.89; FIGs. 7A- 7C), consistent with the expression of highly correlating genes being heavily weighted in the CD8 signature score calculation (FIG. ID).
[0294] Comparison of the CD8 Signature and Other Inflammation Signatures in Predicting CD8+ T-cell Infiltration and PD-L1 Expression on Tumor Cells
[0295] We then explored the interchangeability of different gene expression signatures to predict CD8+ T-cell infiltration in the TME. Across the replicate MEL and SCCHN samples, each of the inflammation signatures tested correlated with CD8 IHC (r > 0.69), with the CD8 signature showing the highest correlation with CD8 IHC (FIGs. 8A-8C). In contrast, neither the CD8 signature, TCA signature, nor TIS correlated with tumor PD-L1 expression by IHC (r < 0.23; FIGs. 8D-8F), with high and low signature scores observed in both PD-L1 -negative and PD-L1 -positive samples (PD-L1 expression on < 1% and > 1% of tumor cells, respectively; FIGs. 8G-8I) Across all twelve tumor types, neither CD8 IHC nor CD8 signature scores correlated with PD-L1 expression on tumor cells (FIGs. 9A-9X).
TMEM173 (STING1)
[0296] Clinical Validation in Urothelial Carcinoma Samples
[0297] In order to explore the clinical utility of the CD8 signature in predicting inflammatory responses in the TME and clinical response to ICIs, the CD8 signature was determined in pretreatment platinum-resistant metastatic UC samples from patients subsequently treated with nivolumab (3 mg/kg intravenously every 2 weeks). Of 270 samples from patients enrolled in CheckMate 275 (NCT02387996), 263 (97%) were evaluable for CD8 IHC, 205 (76%) were evaluable for the CD8 signature by GEP, and 203 (75%) were evaluable for both CD8 IHC and the CD8 signature. Reasons for sample attrition included insufficient tumor biopsy available for GEP or insufficient RNA quality. Baseline characteristics were similar between all treated patients and the GEP-evaluable cohort (Table XX). Median PFS was 1.94 months (95% CI, 1.87-2.33) for all treated patients and 1.90 months (95% CI, 1.87-2.56) in the GEP-evaluable cohort. Median OS was 8.57 (95% CI, 6.04-11.27) for all treated patients and 9.49 (95% CI, 6.31-11.40) for the GEP-evaluable cohort. In the 203 GEP-and IHC evaluable samples, a strong correlation between CD8 signature and CD8 IHC seen with clinical samples (r = 0.77; FIG. 10) was similar to that observed in commercially procured UC samples (r = 0.87; FIG. 3C).
Table XX. Baseline Characteristics in Patients From CheckMate 275.
Figure imgf000087_0001
*One patient had ECOG performance status of 3. ECOG, Eastern Cooperative Oncology Group. [0298] In retrospective analyses, associations with survival were examined by grouping GEP-evaluable patients (n = 205) by CD8 signature score tertiles defined as low (< 7.9), medium (7.9-12.6), or high (> 12.6). Median PFS was higher for patients with CD8 signature scores in the upper tertile (3.65 months, 95% CI, 1.97-5.49) than for patients with CD8 signature scores in the lower two tertiles (1.81 months, 95% CI, 1.68-3.15, and 1.87 months, 95% CI, 1.68-2.04, for patients with scores in the middle and lower tertiles, respectively; FIG. 11 A). Prolonged OS was also observed in patients with CD8 signature scores in the upper tertile (22.14 months, 95% CI, 9.99-30.72) compared with patients with CD8 signature scores in the lower two tertiles (5.72 months, 95% CI, 4.37-11.43, and 6.24 months, 95% CI, 4.53-9.00, for patients with scores in the middle and lower tertiles, respectively; FIG. 1 IB).
[0299] Discussion
[0300] The level of multiplexing possible by IHC is limited, whereas transcriptomic analysis allows the simultaneous interrogation of multiple biomarkers in the TME and may provide a robust characterization of inflammation. This study reports the development of an NGS-based IUO assay for a 16-gene expression signature, including markers of tumorinfiltrating lymphocytes, cytokine signaling, and other inflammatory responses, to be used as a surrogate assessment of inflammation in tumor samples.
[0301] Infiltration of CD8+ T cells, often assessed by IHC, can be used as a robust marker of inflammation that, for some tumor types, is associated with prognosis and/or response to treatment. The CD8 signature was developed to predict CD8+ T-cell abundance across multiple tumor types and analytically validated across 1633 commercial samples. Inflammation status determined by the CD8 signature score correlated with that derived by CD8 IHC (r > 0.82) in all tumor types tested except pancreatic and prostate cancer, indicating that this gene signature may be utilized as an accurate surrogate measure of inflammation across a range of tumor types. For most tumor types, prevalence of CD8-positivity was similar for GEP -based and IHC -based methods, suggesting that a CD8 signature assay has the potential to be utilized prospectively in a clinical trial setting to determine inflammation-positive patients. For SCLC, CD8+ T-cell abundance was lower than observed in other tumor types, and a high correlation between CD8 signature and CD8 IHC scores was observed. However, a linear increase in CD8 signature scores resulted in fewer samples falling within the 95% CI for CD8 signature vs CD8 IHC correlations than seen for other tumor types and in increased variability in the assessment of inflammation prevalence. For patients with prostate cancer, associations have been observed between intratumoral T-cell abundance and favorable clinical benefit with ICIs, yet the full clinical relevance of CD8+ T-cell infiltration is still unclear for patients with this tumor type. The ranges of both CD8 IHC and CD8 signature scores were lower in prostate cancer samples than for other tumor types, and the modest linear correlation between CD8 IHC and CD8 signature was consistent with the low prevalence of inflammation observed in this set of prostate cancer samples. These results warrant further investigation to determine the suitability of this assay for profiling patients with tumor types such as prostate cancer, and to validate whether CD8 is an appropriate biomarker of response to ICI therapy in tumor types where the prevalence of CD8+ T-cell infiltration is low.
[0302] The sample processing, pathological evaluation, and data processing steps employed in this IUO assay have enabled reliable GEP -based surrogate assessment of CD8+ T- cell abundance in the TME. The IUO CD8 signature assay demonstrated high reproducibility when using the TIP to determine CD8 signature scores across 12 tumor types. Assay precision (CV < 25%) was demonstrated for samples with CD8 signature scores > 5. CD8 signature scores correlated with CD8 IHC scores on all platforms tested, demonstrating platform-independent consistency for the CD8 signature as a surrogate measure of CD8+ T-cell abundance. Similar results were observed for select alternative GEP-derived tumor inflammation signatures, demonstrating the robust nature of inflammation gene expression signatures and potential feasibility of utilizing multiple GEP platforms for their assessment. The TIP assay combines extraction-free RNase protection with NGS in an automated EdgeSeq system. This system can generate high-quality sequencing data from small amounts of FFPE samples (as little as 7.2 mm2 of a 4-5 pm section), overcoming some of the limitations experienced with alternative GEP methods, including sample acquisition and assay standardization.
[0303] Across all tumor types tested, CD8 signature scores did not correlate with PD-L1 expression on tumor cells, although a trend for increased CD8 signature score was observed in PD-L1 -positive patients. Additional studies are warranted to assess the relationship between CD8 signature scores and PD-L1 expression on tumor cells, immune cells, or both. The lack of correlation between potential biomarkers of response to ICIs suggests that some or all of these may reflect distinct, nonoverlapping, biological aspects of inflammation in the TME and provides a rationale for additional studies to evaluate the potential of multiple biomarkers in combination. Indeed, retrospective analysis of GEP data from CheckMate 275 showed improvements in response rates, median PFS, and median OS for patients with a combination of high CD8 IHC and low EMT/stroma-related signature scores. Retrospective analyses across multiple tumor types have also demonstrated improved response rates and PFS with ICIs in patients with high TMB combined with high TIS scores, compared with patients with low scores for either these biomarkers. Recent evidence therefore suggests that gene expression signatures may complement existing methods of biomarker assessment, and combined biomarker assessment may lead to more effective precision medicine for patients with cancer.
[0304] This study demonstrated that an analytically validated multitumor gene expression panel assay for the 16-gene CD8 signature can identify an inflammatory phenotype in the TME and may identify biomarker-selected patients that are distinct from PD-L1 -positive populations. Comparisons suggest potential to validate CD8 signature assays on additional platforms. In retrospective, post-hoc analyses of patients with UC, the CD8 signature was associated with the clinical efficacy of nivolumab. Further investigation is ongoing to determine the predictive power of the CD8 signature in selecting patients most likely to respond to ICIs and to assess the clinical utility of gene expression signatures to guide precision medicine in patients with cancer.

Claims

What is claimed is:
1. A pharmaceutical composition comprising an immune-oncology (I-O) therapy for use in a method of treating a tumor in a subject in need thereof, wherein the subject has been identified as having a high pan-tumor gene signature score; wherein the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and wherein the pan-tumor gene signature score is determined according to the following formula: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein:
(a) = any real number between 0.01 and 0.05;
(b) = any real number between 0.10 and 0.15;
(c) = any real number between 0.01 and 0.05;
(d) = any real number between -0.10 and -0.15;
(e) = any real number between 0.03 and 0.09;
(f) = any real number between 0.35 and 0.45;
(g) = any real number between 0.07 and 0.13;
(h) = any real number between 0.02 and 0.08;
(i) = any real number between -0.01 and -0.05;
(j) = any real number between -0.0001 and -0.0010;
(k) = any real number between 0.01 and 0.06;
(l) = any real number between 0.02 and 0.08;
(m)= any real number between -0.008 and -0.040;
(n) = any real number between 0.01 and 0.050;
89 (o) = any real number between -0.03 and -0.080; and
(p) = any real number between -0.005 and -0.010. The pharmaceutical composition for use of claim 1, wherein the high pan-tumor gene signature score is characterized by a score of greater than about 12.6. A pharmaceutical composition comprising an anticancer agent for use in a method of treating a tumor in a subject in need thereof, wherein the anticancer agent does not comprise an 1-0 therapy, wherein the subject has been identified as having a low pantumor gene signature score; wherein the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and wherein the pan-tumor gene signature score is determined according to the following formula: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein:
(a) = any real number between 0.01 and 0.05;
(b) = any real number between 0.10 and 0.15;
(c) = any real number between 0.01 and 0.05;
(d) = any real number between -0.10 and -0.15;
(e) = any real number between 0.03 and 0.09;
(f) = any real number between 0.35 and 0.45;
(g) = any real number between 0.07 and 0.13;
(h) = any real number between 0.02 and 0.08;
(i) = any real number between -0.01 and -0.05;
(j) = any real number between -0.0001 and -0.0010;
90 (k) = any real number between 0.01 and 0.06;
(l) = any real number between 0.02 and 0.08;
(m)= any real number between -0.008 and -0.040;
(n) = any real number between 0.01 and 0.050;
(o) = any real number between -0.03 and -0.080; and
(p) = any real number between -0.005 and -0.010. The pharmaceutical composition for use of claim 3, wherein the low pan-tumor gene signature score is characterized by a score of less than about 7.9. A pharmaceutical composition comprising an 1-0 therapy for use in a method of treating a tumor in a subject in need thereof, wherein the 1-0 therapy is to be administered in combination with an additional anticancer agent to a subject that has been identified as having a moderate pan-tumor gene signature score; wherein the pan-tumor gene signature score is obtained by measuring the expression of a panel of genes in a tumor sample obtained from the subject, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and wherein the pan-tumor gene signature score is determined according to the following formula: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein:
(a) = any real number between 0.01 and 0.05;
(b) = any real number between 0.10 and 0.15;
(c) = any real number between 0.01 and 0.05;
(d) = any real number between -0.10 and -0.15;
(e) = any real number between 0.03 and 0.09;
(f) = any real number between 0.35 and 0.45;
91 (g) = any real number between 0.07 and 0.13;
(h) = any real number between 0.02 and 0.08;
(i) = any real number between -0.01 and -0.05;
(j) = any real number between -0.0001 and -0.0010;
(k) = any real number between 0.01 and 0.06;
(l) = any real number between 0.02 and 0.08;
(m)= any real number between -0.008 and -0.040;
(n) = any real number between 0.01 and 0.050;
(o) = any real number between -0.03 and -0.080; and
(p) = any real number between -0.005 and -0.010. The pharmaceutical composition for use of claim 3, wherein the moderate pan-tumor gene signature score is characterized by a score of from about 7.9 to about 12.6. A pan-tumor gene panel for use in a method of identifying a subject suitable for an 1-0 therapy comprising calculating a pan-tumor gene signature score for a tumor sample obtained from the subject, wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the pan-tumor gene panel in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying the following formula to the measured expression values: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein:
(a) = any real number between 0.01 and 0.05;
(b) = any real number between 0.10 and 0.15;
(c) = any real number between 0.01 and 0.05;
(d) = any real number between -0.10 and -0.15;
(e) = any real number between 0.03 and 0.09;
92 (f) = any real number between 0.35 and 0.45;
(g) = any real number between 0.07 and 0.13;
(h) = any real number between 0.02 and 0.08;
(i) = any real number between -0.01 and -0.05;
(j) = any real number between -0.0001 and -0.0010;
(k) = any real number between 0.01 and 0.06;
(l) = any real number between 0.02 and 0.08;
(m)= any real number between -0.008 and -0.040;
(n) = any real number between 0.01 and 0.050;
(o) = any real number between -0.03 and -0.080; and
(p) = any real number between -0.005 and -0.010. The pan-tumor gene panel for use of claim 7, wherein the subject is identified as being suitable for an 1-0 therapy if the subject has a high pan-tumor gene signature score. A pan-tumor gene panel for use in a method of stratifying a subject in need of an 1-0 therapy, comprising determining a pan-tumor gene signature score for a tumor sample obtained from the subject, wherein the pan-tumor gene signature score is calculated by measuring the expression of a panel of genes in the sample, wherein the panel of genes comprises CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCL10, CXCL9, CXCR1, HLA-DMB, HLA-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173, and TNFSF18; and applying the following formula to the measured expression values: pan-tumor signature score = [CCL4] * (a) + [CCL5] * (b) + [CD27] * (c) + [CD276] * (d) + [CD3D] * (e) + [CD8A] * (f) + [CXCL10] * (g) + [CXCL9] * (h) + [CXCR1] * (i) + [HLA-DMB] * (j) + [HLA-DRA] * (k) + [HLA-DRB1] * (1) + [LGALS9] * (m) + [NKG7] * (n) + [TMEM173] * (o) + [TNFSF18] * (p); wherein:
(a) = any real number between 0.01 and 0.05;
(b) = any real number between 0.10 and 0.15;
(c) = any real number between 0.01 and 0.05;
(d) = any real number between -0.10 and -0.15;
93 (e) = any real number between 0.03 and 0.09;
(f) = any real number between 0.35 and 0.45;
(g) = any real number between 0.07 and 0.13;
(h) = any real number between 0.02 and 0.08;
(i) = any real number between -0.01 and -0.05;
(j) = any real number between -0.0001 and -0.0010;
(k) = any real number between 0.01 and 0.06;
(l) = any real number between 0.02 and 0.08;
(m)= any real number between -0.008 and -0.040;
(n) = any real number between 0.01 and 0.050;
(o) = any real number between -0.03 and -0.080; and
(p) = any real number between -0.005 and -0.010; and wherein the subject is identified as having a high pan-tumor gene signature score if the score greater than about 12.6; wherein the subject is identified as having a low pan-tumor gene signature score if the score is less than about 7.9; and wherein the subject is identified as having a moderate pan-tumor gene signature score if the score is from about 7.9 to about 12.6. The pan-tumor gene panel for use of claim 9, wherein the subject identified as having a high gene signature score is to be administered an 1-0 therapy. The pan-tumor gene panel for use of claim 9, wherein the subject identified as having a low gene signature score is to be administered an anticancer agent other than an 1-0 therapy. The pan-tumor gene panel for use of claim 9, wherein the subject identified as having a moderate gene signature score is to be administered an 1-0 therapy in combination with an additional anticancer agent. The pharmaceutical composition for use of any one of claims 1 to 6 or the pan-tumor gene panel for use of any one of claims 7 to 12, wherein (a) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.05.
94 The pharmaceutical composition for use of any one of claims 1 to 6 and 13 or the pantumor gene panel for use of any one of claims 7 to 13, wherein (a) = about 0.03 to about 0.04. The pharmaceutical composition for use of any one of claims 1 to 6, 13, and 14 or the pan-tumor gene panel for use of any one of claims 7 to 14, wherein (a) = about 0.03447. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 15 or the pan-tumor gene panel for use of any one of claims 7 to 15, wherein (b) = about 0.1, about 0.105, about 0.11, about 0.115, about 0.12, about 0.125, about 0.13, about 0.135, about 0.14, about 0.145, or about 0.15. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 16 or the pan-tumor gene panel for use of any one of claims 7 to 16, wherein (b) = about 0.13 to about 0.14. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 17 or the pan-tumor gene panel for use of any one of claims 7 to 17, wherein (b) = about 0.13825. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 18 or the pan-tumor gene panel for use of any one of claims 7 to 18, wherein (c) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.05. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 19 or the pan-tumor gene panel for use of any one of claims 7 to 19, wherein (c) = about 0.02 to about 0.03. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 20 or the pan-tumor gene panel for use of any one of claims 7 to 20, wherein (c) = about 0.02756. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 21 or the pan-tumor gene panel for use of any one of claims 7 to 21, wherein (d) = about -0.1, about -0.105, about -0.11, about -0.115, about -0.12, about -0.125, about -0.13, about - 0.135, about -0.14, about -0.145, or about -0.15.
95 The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 22 or the pan-tumor gene panel for use of any one of claims 7 to 22, wherein (d) = about -0.10 to about -0.15. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 23 or the pan-tumor gene panel for use of any one of claims 7 to 23, wherein (d) = about -0.13089. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 24 or the pan-tumor gene panel for use of any one of claims 7 to 24, wherein (e) = about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, about 0.08, about 0.085, or about 0.09. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 25 or the pan-tumor gene panel for use of any one of claims 7 to 25, wherein (e) = about 0.06 to about 0.07. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 26 or the pan-tumor gene panel for use of any one of claims 7 to 26, wherein (e) = about 0.06368. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 27 or the pan-tumor gene panel for use of any one of claims 7 to 27, wherein (f) = about 0.35, about 0.36, about 0.37, about 0.38, about 0.39, about 0.40, about 0.41, about 0.42, about 0.43, about 0.44, or about 0.45. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 28 or the pan-tumor gene panel for use of any one of claims 7 to 28, wherein (f) = about 0.39 to about 0.40. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 29 or the pan-tumor gene panel for use of any one of claims 7 to 29, wherein (f) = about 0.39185. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 30 or the pan-tumor gene panel for use of any one of claims 7 to 30, wherein (g) = about 0.07, about 0.075, about 0.08, about 0.085, about 0.09, about 0.095, about 0.10, about 0.105, about 0.11, about 0.115, about 0.12, about 0.125, or about 0.13.
96 The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 31 or the pan-tumor gene panel for use of any one of claims 7 to 31, wherein (g) = about 0.10 to about 0.11. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 32 or the pan-tumor gene panel for use of any one of claims 7 to 32, wherein (g) = about 0.10976. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 33 or the pan-tumor gene panel for use of any one of claims 7 to 33, wherein (h) = about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.08. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 34 or the pan-tumor gene panel for use of any one of claims 7 to 34, wherein (h) = about 0.04 to about 0.05. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 35 or the pan-tumor gene panel for use of any one of claims 7 to 35, wherein (h) = about 0.04913. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 36 or the pan-tumor gene panel for use of any one of claims 7 to 36, wherein (i) = about -0.01, about -0.015, about -0.02, about -0.025, about -0.03, about -0.035, about -0.04, about - 0.045, or about -0.05. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 37 or the pan-tumor gene panel for use of any one of claims 7 to 37, wherein (i) = about -0.02 to about -0.03. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 38 or the pan-tumor gene panel for use of any one of claims 7 to 38, wherein (i) = about -0.02537. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 39 or the pan-tumor gene panel for use of any one of claims 7 to 39, wherein (j) = about -0.0005, about -0.00055, about -0.0006, about -0.00065, about -0.0007, about -0.00075, about - 0.0008, about -0.00085, about -0.0009, about -0.00095, or about -0.0010. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 40 or the pan-tumor gene panel for use of any one of claims 7 to 40, wherein (j) = about -0.0008 to about -0.0009. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 41 or the pan-tumor gene panel for use of any one of claims 7 to 41, wherein (j) = about -0.00081. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 42 or the pan-tumor gene panel for use of any one of claims 7 to 42, wherein (k) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, or about 0.06. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 43 or the pan-tumor gene panel for use of any one of claims 7 to 43, wherein (k) = about 0.03 to about 0.04. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 44 or the pan-tumor gene panel for use of any one of claims 7 to 44, wherein (k) = about 0.03654. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 45 or the pan-tumor gene panel for use of any one of claims 7 to 45, wherein (1) = about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, about 0.05, about 0.055, about 0.06, about 0.065, about 0.07, about 0.075, or about 0.8. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 46 or the pan-tumor gene panel for use of any one of claims 7 to 46, wherein (1) = about 0.04 to about 0.05. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 47 or the pan-tumor gene panel for use of any one of claims 7 to 47, wherein (1) = about 0.04797. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 48 or the pan-tumor gene panel for use of any one of claims 7 to 48, wherein (m) = about -0.008, about -0.010, about -0.015, about -0.020, about -0.025, about -0.030, about -0.035, or about -0.0400. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 49 or the pan-tumor gene panel for use of any one of claims 7 to 49, wherein (m) = about -0.01 to about -0.02. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 50 or the pan-tumor gene panel for use of any one of claims 7 to 50, wherein (m) = about -0.01751. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 51 or the pan-tumor gene panel for use of any one of claims 7 to 51, wherein (n) = about 0.01, about 0.015, about 0.02, about 0.025, about 0.03, about 0.035, about 0.04, about 0.045, or about 0.050. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 52 or the pan-tumor gene panel for use of any one of claims 7 to 52, wherein (n) = about 0.02 to about 0.03. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 53 or the pan-tumor gene panel for use of any one of claims 7 to 53, wherein (n) = about 0.02254. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 54 or the pan-tumor gene panel for use of any one of claims 7 to 54, wherein (o) = about -0.03, about -0.035, about -0.04, about -0.045, about -0.05, about -0.055, about -0.06, about - 0.065, about -0.07, about -0.075, or about -0.08. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 55 or the pan-tumor gene panel for use of any one of claims 7 to 55, wherein (o) = about -0.05 to about -0.06. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 56 or the pan-tumor gene panel for use of any one of claims 7 to 56, wherein (o) = about -0.05317. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 57 or the pan-tumor gene panel for use of any one of claims 7 to 57, wherein (p) = about -0.005, about -0.0055, about -0.006, about -0.0065, about -0.007, about -0.0075, about -0.008, about -0.0085, about -0.009, about -0.0095, or about -0.010.
99 The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 58 or the pan-tumor gene panel for use of any one of claims 7 to 58, wherein (p) = about 0.007 to about 0.008. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 59 or the pan-tumor gene panel for use of any one of claims 7 to 59, wherein (p) = about -0.00710. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 60 or the pan-tumor gene panel for use of any one of claims 7 to 60, wherein:
(a) = about 0.03447;
(b) = about 0.13825;
(c) = about 0.02756;
(d) = about -0.13089;
(e) = about 0.06368;
(f) = about 0.39185;
(g) = about 0.10976;
(h) = about 0.04913;
(i) = about -0.02537;
(j) = about -0.00081;
(k) = about 0.03654;
(l) = about 0.04797;
(m)= about -0.01751;
(n) = about 0.02254;
(o) = about -0.05317; and
(p) = about -0.00710. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 61 or the pan-tumor gene panel for use of any one of claims 7 to 61, wherein the gene panel further comprises one or more housekeeping genes. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 62 or the pan-tumor gene panel for use of any one of claims 7 to 62, which further comprises one or more housekeeping genes selected from the group consisting of ACTB, ATP5F1,
100 DDX5, EEF1G, GAPDH, NCL, 0AZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ. and any combination thereof. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 63 or the pan-tumor gene panel for use of any one of claims 7 to 63, which further comprises one or more control genes. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 64 or the pan-tumor gene panel for use of any one of claims 7 to 64, which further comprises one or more control genes selected from the group consisting of ANTI, ANT2, ANT3, ANT4, PCL-1, PCL-10, PCL-2, PCL-3, PCL-4, PCL-5, PCL-6, PCL-7, PCL-8, PCL-9, POS1, POS2, POS3, POS4, and any combination thereof. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 65 or the pan-tumor gene panel for use of any one of claims 7 to 65, wherein the 1-0 therapy comprises an immune checkpoint inhibitor. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 66 or the pan-tumor gene panel for use of any one of claims 7 to 66, wherein the 1-0 therapy comprises an anti-PDl/PD-L2 antagonist. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 66 or the pan-tumor gene panel for use of any one of claims 7 to 66, wherein the 1-0 therapy comprises an antibody that specifically binds a protein selected from PD-1, PD-L1, LAG- 3, TIGIT, TIM3, NKG2a, CSF1R, 0X40, ICOS, MICA, MICB,CD137, KIR, TGFp, IL- 10, IL-8, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, or any combination thereof. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 68 or the pan-tumor gene panel for use of any one of claims 7 to 68, wherein the 1-0 comprises an anti -PD-1 antibody. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 68 or the pan-tumor gene panel for use of any one of claims 7 to 68, wherein the 1-0 therapy comprises an anti-PD-Ll antibody.
101 The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 70 or the pan-tumor gene panel for use of any one of claims 7 to 70, wherein the tumor sample is not subjected to CD8 immunohistochemistry. A method of identifying a patient in need of an 1-0 therapy,
(a) obtaining a tumor sample from the patient;
(b) analyzing the expression level of one or more genes in a gene panel selected from C(71.4, CCL5, CD27, CD276, CD3D, CD8A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HI.A-DRA, HLA-DRB1, I.GAI.S9, NKG7, TMEM173 (STING1), TNFSF18, and any combination thereof; and
(c) determining a pan-tumor gene signature score according to any of claims 1 to 71. The method of claim 72, further comprising isolating mRNA from the tumor sample prior to analyzing the expression level of the one or more genes. A method of treating a subject in need thereof, comprising:
(a) obtaining a tumor sample from the subject;
(b) isolating mRNA from the tumor sample;
(c) analyzing the expression level of one or more genes in a gene panel selected from CCL4, CCL5, CD27, CD276, CD3D, CD8A, CXCI.10, CXCI.9, CXCR1, HI.A-DMB, HI.A-DRA, HLA-DRB1, LGALS9, NKG7, TMEM173 (STING1), TNFSF18, and any combination thereof;
(d) determining a pan-tumor gene signature score according to any of claims 1 to 73; and
(e) administering to the subject an 1-0 therapy The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 71, the pantumor gene panel for use of any one of claims 7 to 71, or the method of any one of claims 72 to 74, wherein the expression level of the one or more genes in the gene panel is analyzed by measuring an mRNA level of the one or more genes in the gene panel in the tumor sample.
102 The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 71, the pantumor gene panel for use of any one of claims 7 to 71, or the method of any one of claims 72 to 75, wherein the expression level is measured using a nuclease protection assay. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 71, the pantumor gene panel for use of any one of claims 7 to 71, or the method of any one of claims 72 to 75, wherein the expression level is measured using next-generation sequencing. The pharmaceutical composition for use of any one of claims 1 to 6 and 13 to 71, the pantumor gene panel for use of any one of claims 7 to 71, or the method of any one of claims 72 to 75, wherein the expression level is measured using reverse transcriptase polymerase chain reaction (RT-PCR).
103
PCT/US2021/061823 2020-12-03 2021-12-03 Multi-tumor gene signatures and uses thereof WO2022120179A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063121137P 2020-12-03 2020-12-03
US63/121,137 2020-12-03

Publications (1)

Publication Number Publication Date
WO2022120179A1 true WO2022120179A1 (en) 2022-06-09

Family

ID=79093013

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/061823 WO2022120179A1 (en) 2020-12-03 2021-12-03 Multi-tumor gene signatures and uses thereof

Country Status (1)

Country Link
WO (1) WO2022120179A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220292661A1 (en) * 2021-03-10 2022-09-15 Adlink Technology Inc. Automated inspection system and operating method thereof

Citations (179)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995012673A1 (en) 1993-11-03 1995-05-11 The Board Of Trustees Of The Leland Stanford Junior University Receptor on the surface of activated t-cells: acts-4
WO1998038216A1 (en) 1997-02-27 1998-09-03 Japan Tobacco Inc. Cell surface molecule mediating cell adhesion and signal transmission
WO1999015553A2 (en) 1997-09-23 1999-04-01 Bundesrepublik Deutschland Letztvertreten Durch Den Direktor Des Robert-Koch-Instituts Costimulating t-cell polypeptide, monoclonal antibodies, their preparation and use
WO1999042585A1 (en) 1998-02-24 1999-08-26 Sisters Of Providence In Oregon Compositions containing an ox-40 receptor binding agent or a nucleic acid encoding the same and methods for enhancing antigen-specific immune response
US5977318A (en) 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
WO2000037504A2 (en) 1998-12-23 2000-06-29 Pfizer Inc. Human monoclonal antibodies to ctla-4
US6210669B1 (en) 1996-10-11 2001-04-03 Bristol-Myers Squibb Co. Methods and compositions for immunomodulation
US6303121B1 (en) 1992-07-30 2001-10-16 Advanced Research And Technology Method of using human receptor protein 4-1BB
US6355476B1 (en) 1988-11-07 2002-03-12 Advanced Research And Technologyinc Nucleic acid encoding MIP-1α Lymphokine
US6362325B1 (en) 1988-11-07 2002-03-26 Advanced Research And Technology Institute, Inc. Murine 4-1BB gene
WO2003106498A2 (en) 2002-06-13 2003-12-24 Crucell Holland, B.V. Agonistic binding molecules to the human ox40 receptor
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US6808710B1 (en) 1999-08-23 2004-10-26 Genetics Institute, Inc. Downmodulating an immune response with multivalent antibodies to PD-1
WO2005003168A2 (en) 2003-07-02 2005-01-13 Novo Nordisk A/S Methods for the production and cytotoxicity evaluation of kir2dl nk-receptor antibodies
WO2005009465A1 (en) 2003-07-24 2005-02-03 Innate Pharma Methods and compositions for increasing the efficiency of therapeutic antibodies using nk cell potentiating compounds
US6887673B2 (en) 2002-07-30 2005-05-03 Bristol-Myers Squibb Company Humanized antibodies against human 4-1BB
US20050095244A1 (en) 2003-10-10 2005-05-05 Maria Jure-Kunkel Fully human antibodies against human 4-1BB
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
WO2006003179A2 (en) 2004-07-01 2006-01-12 Novo Nordisk A/S Antibodies binding to receptors kir2dl1, -2, 3 but not kir2ds4 and their therapeutic use
US7034121B2 (en) 2000-01-27 2006-04-25 Genetics Institue, Llc Antibodies against CTLA4
US7045615B2 (en) 1997-02-27 2006-05-16 Japan Tobacco, Inc. Nucleic acids encoding JTT-1 protein
WO2006070286A2 (en) 2004-12-28 2006-07-06 Innate Pharma S.A. Monoclonal antibodies against nkg2a
WO2006072626A1 (en) 2005-01-06 2006-07-13 Novo Nordisk A/S Kir-binding agents and methods of use thereof
WO2006072625A2 (en) 2005-01-06 2006-07-13 Novo Nordisk A/S Anti-kir combination treatments and methods
WO2006105021A2 (en) 2005-03-25 2006-10-05 Tolerrx, Inc. Gitr binding molecules and uses therefor
WO2007042573A2 (en) 2005-10-14 2007-04-19 Innate Pharma Compositions and methods for treating proliferative disorders
WO2007113648A2 (en) 2006-04-05 2007-10-11 Pfizer Products Inc. Ctla4 antibody combination therapy
WO2008009545A1 (en) 2006-06-30 2008-01-24 Novo Nordisk A/S Anti-nkg2a antibodies and uses thereof
WO2008084106A1 (en) 2007-01-11 2008-07-17 Novo Nordisk A/S Anti-kir antibodies, formulations, and uses thereof
WO2008137915A2 (en) 2007-05-07 2008-11-13 Medimmune, Llc Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
JP2008278814A (en) 2007-05-11 2008-11-20 Igaku Seibutsugaku Kenkyusho:Kk Release of immunoregulation by agonistic anti-human gitr antibody, and application thereof
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
WO2009009116A2 (en) 2007-07-12 2009-01-15 Tolerx, Inc. Combination therapies employing gitr binding molecules
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2009026303A1 (en) 2007-08-21 2009-02-26 Amgen Inc. Human c-fms antigen binding proteins
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2009092805A1 (en) 2008-01-24 2009-07-30 Novo Nordisk A/S Humanized anti-human nkg2a monoclonal antibody
WO2009112245A1 (en) 2008-03-14 2009-09-17 Transgene S.A. Antibody against the csf-1 r
US7722872B2 (en) 1997-09-23 2010-05-25 Bundersrepublik Deutschland Letztvertreten Durch Den Direktor Des Robert-Koch-Institutes Treatment of cancer with antibodies to costimulating polypeptide of T cells
WO2010065939A1 (en) 2008-12-05 2010-06-10 Indiana University Research & Technology Corporation Combination therapy to enhace nk cell mediated cytotoxicty
US7767429B2 (en) 2003-03-05 2010-08-03 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
WO2011028683A1 (en) 2009-09-03 2011-03-10 Schering Corporation Anti-gitr antibodies
WO2011041613A2 (en) 2009-09-30 2011-04-07 Memorial Sloan-Kettering Cancer Center Combination immunotherapy for the treatment of cancer
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2011066389A1 (en) 2009-11-24 2011-06-03 Medimmmune, Limited Targeted binding agents against b7-h1
US20110150892A1 (en) 2008-08-11 2011-06-23 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
WO2011155607A1 (en) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anti-tim-3 antibody
WO2011159877A2 (en) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions
WO2012027328A2 (en) 2010-08-23 2012-03-01 Board Of Regents, The University Of Texas System Anti-ox40 antibodies and methods of using the same
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
WO2012071411A2 (en) 2010-11-22 2012-05-31 Innate Pharma Sa Nk cell modulating treatments and methods for treatment of hematological malignancies
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2012122444A1 (en) 2011-03-10 2012-09-13 Provectus Pharmaceuticals, Inc. Combination of local and systemic immunomodulative therapies for enhanced treatment of cancer
WO2012131004A2 (en) 2011-03-31 2012-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies directed against icos and uses thereof
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2012160448A2 (en) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
WO2013006490A2 (en) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Antibodies that specifically bind to tim3
WO2013028231A1 (en) 2011-08-23 2013-02-28 Board Of Regents, The University Of Texas System Anti-ox40 antibodies and methods of using the same
WO2013039954A1 (en) 2011-09-14 2013-03-21 Sanofi Anti-gitr antibodies
WO2013038191A2 (en) 2011-09-16 2013-03-21 Bioceros B.V. Anti-cd134 (ox40) antibodies and uses thereof
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013132044A1 (en) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2013173223A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2013181634A2 (en) 2012-05-31 2013-12-05 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
US20140065152A1 (en) 2012-06-08 2014-03-06 National Cancer Center Novel epitope for switching to th1 cell and use thereof
US20140093511A1 (en) 2012-07-02 2014-04-03 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014055648A1 (en) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combination of anti-kir antibodies and anti-pd-1 antibodies to treat cancer
WO2014140180A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
WO2014151634A1 (en) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Macrocyclic inhibitors of the pd-1/pd-l1 and cd80(b7-1)/pd-l1 protein/protein interactions
WO2014148895A1 (en) 2013-03-18 2014-09-25 Biocerox Products B.V. Humanized anti-cd134 (ox40) antibodies and uses thereof
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2014194302A2 (en) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Antigen binding proteins that bind pd-1
WO2014206107A1 (en) 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 Anti-pd-1 antibody and use thereof
WO2015026684A1 (en) 2013-08-20 2015-02-26 Merck Sharp & Dohme Corp. Modulation of tumor immunity
WO2015031667A2 (en) 2013-08-30 2015-03-05 Amgen Inc. Gitr antigen binding proteins
WO2015034820A1 (en) 2013-09-04 2015-03-12 Bristol-Myers Squibb Company Compounds useful as immunomodulators
US20150079109A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
WO2015042246A1 (en) 2013-09-20 2015-03-26 Bristol-Myers Squibb Company Combination of anti-lag-3 antibodies and anti-pd-1 antibodies to treat tumors
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015117002A1 (en) 2014-01-31 2015-08-06 Novartis Ag Antibody molecules to tim-3 and uses thereof
WO2015153514A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
WO2015153513A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Anti-ox40 antibodies and methods of use
WO2015160641A2 (en) 2014-04-14 2015-10-22 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2015184099A1 (en) 2014-05-28 2015-12-03 4-Antibody Ag Anti-gitr antibodies and methods of use thereof
WO2015200119A1 (en) 2014-06-26 2015-12-30 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
WO2016028672A1 (en) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anti-lag3 antibodies and antigen-binding fragments
WO2016032334A1 (en) 2014-08-28 2016-03-03 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Cd94/nkg2a and/or cd94/nkg2b antibody, vaccine combinations
WO2016039749A1 (en) 2014-09-11 2016-03-17 Bristol-Myers Squibb Company Macrocyclic inhibitors of the pd-1/pd-l1 and cd80 (b7-1)/pd-li protein/protein interactions
WO2016041945A1 (en) 2014-09-16 2016-03-24 Innate Pharma Neutralization of inhibitory pathways in lymphocytes
WO2016041947A1 (en) 2014-09-16 2016-03-24 Innate Pharma Treatment regimens using anti-nkg2a antibodies
WO2016057624A1 (en) 2014-10-10 2016-04-14 Bristol-Myers Squibb Company Immunomodulators
WO2016057667A1 (en) 2014-10-10 2016-04-14 Medimmune, Llc Humanized anti-ox40 antibodies and uses thereof
WO2016068802A1 (en) 2014-10-27 2016-05-06 Agency For Science, Technology And Research Anti-tim-3 antibodies
WO2016068803A1 (en) 2014-10-27 2016-05-06 Agency For Science, Technology And Research Anti-tim-3 antibodies
WO2016071448A1 (en) 2014-11-06 2016-05-12 F. Hoffmann-La Roche Ag Anti-tim3 antibodies and methods of use
WO2016077518A1 (en) 2014-11-14 2016-05-19 Bristol-Myers Squibb Company Macrocyclic peptides useful as immunomodulators
WO2016100608A1 (en) 2014-12-19 2016-06-23 Bristol-Myers Squibb Company Immunomodulators
WO2016100285A1 (en) 2014-12-18 2016-06-23 Bristol-Myers Squibb Company Immunomodulators
WO2016106302A1 (en) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Antibodies to tigit
WO2016106159A1 (en) 2014-12-22 2016-06-30 Enumeral Biomedical Holding, Inc. Anti-pd-1 antibodies
WO2016111947A2 (en) 2015-01-05 2016-07-14 Jounce Therapeutics, Inc. Antibodies that inhibit tim-3:lilrb2 interactions and uses thereof
WO2016126646A1 (en) 2015-02-04 2016-08-11 Bristol-Myers Squibb Company Immunomodulators
WO2016126858A2 (en) 2015-02-03 2016-08-11 Anaptysbio, Inc. Antibodies directed against lymphocyte activation gene 3 (lag-3)
WO2016134371A2 (en) 2015-02-20 2016-08-25 Ohio State Innovation Foundation Bivalent antibody directed against nkg2d and tumor associated antigens
WO2016144803A2 (en) 2015-03-06 2016-09-15 Sorrento Therapeutics, Inc. Antibody therapeutics that bind tim3
WO2016149201A2 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
WO2016149351A1 (en) 2015-03-18 2016-09-22 Bristol-Myers Squibb Company Immunomodulators
WO2016154177A2 (en) 2015-03-23 2016-09-29 Jounce Therapeutics, Inc. Antibodies to icos
WO2016161270A1 (en) 2015-04-01 2016-10-06 Anaptysbio, Inc. Antibodies directed against t cell immunoglobulin and mucin protein 3 (tim-3)
WO2016196228A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
WO2016196237A1 (en) 2015-05-29 2016-12-08 Agenus Inc. Anti-ctla-4 antibodies and methods of use thereof
WO2016197367A1 (en) 2015-06-11 2016-12-15 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-l1 antibodies
WO2016200836A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies
WO2016200782A1 (en) 2015-06-08 2016-12-15 Macrogenics, Inc. Lag-3-binding molecules and methods of use thereof
WO2017015560A2 (en) 2015-07-22 2017-01-26 Sorrento Therapeutics, Inc. Antibody therapeutics that bind lag3
WO2017019846A1 (en) 2015-07-30 2017-02-02 Macrogenics, Inc. Pd-1-binding molecules and methods use thereof
WO2017020858A1 (en) 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-l1 antibodies
WO2017024515A1 (en) 2015-08-11 2017-02-16 Wuxi Biologics (Cayman) Inc. Novel anti-pd-1 antibodies
WO2017025051A1 (en) 2015-08-11 2017-02-16 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-1 antibodies
WO2017025016A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017025498A1 (en) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Novel fusion polypeptide specific for lag-3 and pd-1
WO2017031242A1 (en) 2015-08-20 2017-02-23 Sutro Biopharma, Inc. Anti-tim-3 antibodies, compositions comprising anti-tim-3 antibodies and methods of making and using anti-tim-3 antibodies
WO2017034916A1 (en) 2015-08-24 2017-03-02 Eli Lilly And Company Pd-l1 ("programmed death-ligand 1") antibodies
WO2017040790A1 (en) 2015-09-01 2017-03-09 Agenus Inc. Anti-pd-1 antibodies and methods of use thereof
WO2017053748A2 (en) 2015-09-25 2017-03-30 Genentech, Inc. Anti-tigit antibodies and methods of use
WO2017055404A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific antibodies specific for pd1 and tim3
WO2017055399A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Cellular based fret assay for the determination of simultaneous binding
WO2017062888A1 (en) 2015-10-09 2017-04-13 Regeneron Pharmaceuticals, Inc. Anti-lag3 antibodies and uses thereof
WO2017066227A1 (en) 2015-10-15 2017-04-20 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2017063162A1 (en) 2015-10-15 2017-04-20 苏州丁孚靶点生物技术有限公司 Anti-ox40 antibody and application thereof
WO2017079112A1 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and their uses
WO2017087589A2 (en) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Pd1 and/or lag3 binders
WO2017086367A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Combination therapy using t cell redirection antigen binding molecule against cell having immunosuppressing function
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
WO2017087901A2 (en) 2015-11-19 2017-05-26 Sutro Biopharma, Inc. Anti-lag3 antibodies, compositions comprising anti-lag3 antibodies and methods of making and using anti-lag3 antibodies
WO2017096182A1 (en) 2015-12-03 2017-06-08 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
WO2017096281A1 (en) 2015-12-02 2017-06-08 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
WO2017096179A1 (en) 2015-12-02 2017-06-08 Agenus Inc. Antibodies and methods of use thereof
WO2017106061A1 (en) 2015-12-14 2017-06-22 Macrogenics, Inc. Bispecific molecules having immunoreactivity with pd-1 and ctla-4, and methods of use thereof
WO2017106129A1 (en) 2015-12-16 2017-06-22 Merck Sharp & Dohme Corp. Anti-lag3 antibodies and antigen-binding fragments
WO2017123557A1 (en) 2016-01-11 2017-07-20 Armo Biosciences, Inc. Interleukin-10 in production of antigen-specific cd8+ t cells and methods of use of same
WO2017134292A1 (en) 2016-02-04 2017-08-10 Glenmark Pharmaceuticals S.A. Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis
WO2017132825A1 (en) 2016-02-02 2017-08-10 华为技术有限公司 Emission power verification method, user equipment, and base station
WO2017133540A1 (en) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
US9738718B2 (en) 2015-01-28 2017-08-22 Glaxosmithkline Intellectual Property Development Limited ICOS binding proteins
WO2017149143A1 (en) 2016-03-04 2017-09-08 Agency For Science, Technology And Research Anti-lag-3 antibodies
WO2017151830A1 (en) 2016-03-04 2017-09-08 Bristol-Myers Squibb Company Immunomodulators
US20170260271A1 (en) 2014-05-13 2017-09-14 Chugai Seiyaku Kabushiki Kaisha T Cell-Redirected Antigen-Binding Molecule For Cells Having Immunosuppression Function
WO2017176608A1 (en) 2016-04-05 2017-10-12 Bristol-Myers Squibb Company Macrocyclic inhibitors of the pd-1/pd-l1 and cd80/pd-l1 protein/protein interactions
WO2017178493A1 (en) 2016-04-12 2017-10-19 Symphogen A/S Anti-tim-3 antibodies and compositions
WO2017198741A1 (en) 2016-05-18 2017-11-23 Boehringer Ingelheim International Gmbh Anti pd-1 and anti-lag3 antibodies for cancer treatment
WO2017205721A1 (en) 2016-05-27 2017-11-30 Agenus Inc. Anti-tim-3 antibodies and methods of use thereof
WO2017219995A1 (en) 2016-06-23 2017-12-28 江苏恒瑞医药股份有限公司 Lag-3 antibody, antigen-binding fragment thereof, and pharmaceutical application thereof
WO2017220555A1 (en) 2016-06-20 2017-12-28 F-Star Beta Limited Lag -3 binding members
WO2017220988A1 (en) 2016-06-20 2017-12-28 Kymab Limited Multispecific antibodies for immuno-oncology
WO2017220569A1 (en) 2016-06-20 2017-12-28 F-Star Delta Limited Binding molecules binding pd-l1 and lag-3
WO2018009505A1 (en) 2016-07-08 2018-01-11 Bristol-Myers Squibb Company 1,3-dihydroxy-phenyl derivatives useful as immunomodulators
WO2018013818A2 (en) 2016-07-14 2018-01-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
WO2018034227A1 (en) 2016-08-15 2018-02-22 国立大学法人北海道大学 Anti-lag-3 antibody
WO2018036561A1 (en) 2016-08-26 2018-03-01 Beigene, Ltd. Anti-tim-3 antibodies and use thereof
WO2018039020A1 (en) 2016-08-25 2018-03-01 Eli Lilly And Company Anti-tim-3 antibodies
WO2018044963A1 (en) 2016-09-01 2018-03-08 Bristol-Myers Squibb Company Biaryl compounds useful as immunomodulators
WO2018069500A2 (en) 2016-10-13 2018-04-19 Symphogen A/S Anti-lag-3 antibodies and compositions
WO2018071500A1 (en) 2016-10-11 2018-04-19 Agenus Inc. Anti-lag-3 antibodies and methods of use thereof
WO2018083087A2 (en) 2016-11-02 2018-05-11 Glaxosmithkline Intellectual Property (No.2) Limited Binding proteins
WO2018085750A2 (en) 2016-11-07 2018-05-11 Bristol-Myers Squibb Company Immunomodulators
WO2018118848A1 (en) 2016-12-20 2018-06-28 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2018183171A1 (en) 2017-03-27 2018-10-04 Bristol-Myers Squibb Company Substituted isoquionline derivatives as immunomudulators
WO2018237153A1 (en) 2017-06-23 2018-12-27 Bristol-Myers Squibb Company Immunomodulators acting as antagonists of pd-1
WO2019070643A1 (en) 2017-10-03 2019-04-11 Bristol-Myers Squibb Company Immunomodulators
WO2019147662A1 (en) 2018-01-23 2019-08-01 Bristol-Myers Squibb Company 2,8-diacyl-2,8-diazaspiro[5.5]undecane compounds useful as immunomodulators
WO2019169123A1 (en) 2018-03-01 2019-09-06 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2020243563A1 (en) * 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Multi-tumor gene signatures for suitability to immuno-oncology therapy
WO2021092220A1 (en) * 2019-11-06 2021-05-14 Bristol-Myers Squibb Company Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy

Patent Citations (217)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6905685B2 (en) 1988-11-07 2005-06-14 Byoung S. Kwon Methods of using antibodies to human receptor protein 4-1BB
US6974863B2 (en) 1988-11-07 2005-12-13 Indiana University Research And Technology Corporation Antibody for 4-1BB
US6362325B1 (en) 1988-11-07 2002-03-26 Advanced Research And Technology Institute, Inc. Murine 4-1BB gene
US6355476B1 (en) 1988-11-07 2002-03-12 Advanced Research And Technologyinc Nucleic acid encoding MIP-1α Lymphokine
US5977318A (en) 1991-06-27 1999-11-02 Bristol Myers Squibb Company CTLA4 receptor and uses thereof
US6303121B1 (en) 1992-07-30 2001-10-16 Advanced Research And Technology Method of using human receptor protein 4-1BB
WO1995012673A1 (en) 1993-11-03 1995-05-11 The Board Of Trustees Of The Leland Stanford Junior University Receptor on the surface of activated t-cells: acts-4
US6569997B1 (en) 1995-03-23 2003-05-27 Advanced Research And Technology Institute, Inc. Antibody specific for H4-1BB
US6051227A (en) 1995-07-25 2000-04-18 The Regents Of The University Of California, Office Of Technology Transfer Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
US6210669B1 (en) 1996-10-11 2001-04-03 Bristol-Myers Squibb Co. Methods and compositions for immunomodulation
US8389690B2 (en) 1997-02-27 2013-03-05 Japan Tobacco Inc. Antibodies to JTT-1 protein and cells secreting such antibodies
US7112655B1 (en) 1997-02-27 2006-09-26 Japan Tobacco, Inc. JTT-1 protein and methods of inhibiting lymphocyte activation
US7045615B2 (en) 1997-02-27 2006-05-16 Japan Tobacco, Inc. Nucleic acids encoding JTT-1 protein
WO1998038216A1 (en) 1997-02-27 1998-09-03 Japan Tobacco Inc. Cell surface molecule mediating cell adhesion and signal transmission
US7259247B1 (en) 1997-09-23 2007-08-21 Bundersrespublik Deutschaland Letztvertreten Durch Den Direktor Des Robert-Koch-Institutes Anti-human T-cell costimulating polypeptide monoclonal antibodies
WO1999015553A2 (en) 1997-09-23 1999-04-01 Bundesrepublik Deutschland Letztvertreten Durch Den Direktor Des Robert-Koch-Instituts Costimulating t-cell polypeptide, monoclonal antibodies, their preparation and use
US7722872B2 (en) 1997-09-23 2010-05-25 Bundersrepublik Deutschland Letztvertreten Durch Den Direktor Des Robert-Koch-Institutes Treatment of cancer with antibodies to costimulating polypeptide of T cells
WO1999042585A1 (en) 1998-02-24 1999-08-26 Sisters Of Providence In Oregon Compositions containing an ox-40 receptor binding agent or a nucleic acid encoding the same and methods for enhancing antigen-specific immune response
WO2000037504A2 (en) 1998-12-23 2000-06-29 Pfizer Inc. Human monoclonal antibodies to ctla-4
US6682736B1 (en) 1998-12-23 2004-01-27 Abgenix, Inc. Human monoclonal antibodies to CTLA-4
US6808710B1 (en) 1999-08-23 2004-10-26 Genetics Institute, Inc. Downmodulating an immune response with multivalent antibodies to PD-1
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
US7034121B2 (en) 2000-01-27 2006-04-25 Genetics Institue, Llc Antibodies against CTLA4
WO2003106498A2 (en) 2002-06-13 2003-12-24 Crucell Holland, B.V. Agonistic binding molecules to the human ox40 receptor
US6887673B2 (en) 2002-07-30 2005-05-03 Bristol-Myers Squibb Company Humanized antibodies against human 4-1BB
US7214493B2 (en) 2002-07-30 2007-05-08 Bristol-Myers Squibb Company Polynucleotides encoding humanized antibodies against human 4-1BB
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
US7767429B2 (en) 2003-03-05 2010-08-03 Halozyme, Inc. Soluble hyaluronidase glycoprotein (sHASEGP), process for preparing the same, uses and pharmaceutical compositions comprising thereof
WO2005003168A2 (en) 2003-07-02 2005-01-13 Novo Nordisk A/S Methods for the production and cytotoxicity evaluation of kir2dl nk-receptor antibodies
WO2005009465A1 (en) 2003-07-24 2005-02-03 Innate Pharma Methods and compositions for increasing the efficiency of therapeutic antibodies using nk cell potentiating compounds
US20050095244A1 (en) 2003-10-10 2005-05-05 Maria Jure-Kunkel Fully human antibodies against human 4-1BB
US7288638B2 (en) 2003-10-10 2007-10-30 Bristol-Myers Squibb Company Fully human antibodies against human 4-1BB
WO2006003179A2 (en) 2004-07-01 2006-01-12 Novo Nordisk A/S Antibodies binding to receptors kir2dl1, -2, 3 but not kir2ds4 and their therapeutic use
US8993319B2 (en) 2004-12-28 2015-03-31 Innate Pharma S.A. Monoclonal antibodies against NKG2A
WO2006070286A2 (en) 2004-12-28 2006-07-06 Innate Pharma S.A. Monoclonal antibodies against nkg2a
WO2006072625A2 (en) 2005-01-06 2006-07-13 Novo Nordisk A/S Anti-kir combination treatments and methods
WO2006072626A1 (en) 2005-01-06 2006-07-13 Novo Nordisk A/S Kir-binding agents and methods of use thereof
US7812135B2 (en) 2005-03-25 2010-10-12 Tolerrx, Inc. GITR-binding antibodies
WO2006105021A2 (en) 2005-03-25 2006-10-05 Tolerrx, Inc. Gitr binding molecules and uses therefor
US8388967B2 (en) 2005-03-25 2013-03-05 Gitr, Inc. Methods for inducing or enhancing an immune response by administering agonistic GITR-binding antibodies
US20130183321A1 (en) 2005-03-25 2013-07-18 Gitr, Inc. Gitr binding molecules and uses therefor
US8779105B2 (en) 2005-05-09 2014-07-15 Medarex, L.L.C. Monoclonal antibodies to programmed death 1 (PD-1)
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US9580507B2 (en) 2005-07-01 2017-02-28 E.R. Squibb & Sons, L. L. C. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US9447185B2 (en) 2005-10-14 2016-09-20 Innate Pharma, S.A. Compositions and methods for treating proliferative disorders
WO2007042573A2 (en) 2005-10-14 2007-04-19 Innate Pharma Compositions and methods for treating proliferative disorders
WO2007113648A2 (en) 2006-04-05 2007-10-11 Pfizer Products Inc. Ctla4 antibody combination therapy
US8901283B2 (en) 2006-06-30 2014-12-02 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
US8206709B2 (en) 2006-06-30 2012-06-26 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
WO2008009545A1 (en) 2006-06-30 2008-01-24 Novo Nordisk A/S Anti-nkg2a antibodies and uses thereof
US9683041B2 (en) 2006-06-30 2017-06-20 Novo Nordisk A/S Anti-NKG2A antibodies and uses thereof
WO2008084106A1 (en) 2007-01-11 2008-07-17 Novo Nordisk A/S Anti-kir antibodies, formulations, and uses thereof
WO2008137915A2 (en) 2007-05-07 2008-11-13 Medimmune, Llc Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
JP2008278814A (en) 2007-05-11 2008-11-20 Igaku Seibutsugaku Kenkyusho:Kk Release of immunoregulation by agonistic anti-human gitr antibody, and application thereof
US8900587B2 (en) 2007-06-18 2014-12-02 Merck Sharp & Dohme Corp. Antibodies to human programmed death receptor PD-1
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
US20090136494A1 (en) 2007-07-12 2009-05-28 Tolerx, Inc. Combination therapies employing GITR binding molecules
WO2009009116A2 (en) 2007-07-12 2009-01-15 Tolerx, Inc. Combination therapies employing gitr binding molecules
US20140220002A1 (en) 2007-07-12 2014-08-07 Gitr, Inc. Combination therapies employing gitr binding molecules
WO2009026303A1 (en) 2007-08-21 2009-02-26 Amgen Inc. Human c-fms antigen binding proteins
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
WO2009092805A1 (en) 2008-01-24 2009-07-30 Novo Nordisk A/S Humanized anti-human nkg2a monoclonal antibody
US8796427B2 (en) 2008-01-24 2014-08-05 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
US9422368B2 (en) 2008-01-24 2016-08-23 Novo Nordisk A/S Humanized anti-human NKG2A monoclonal antibody
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
WO2009112245A1 (en) 2008-03-14 2009-09-17 Transgene S.A. Antibody against the csf-1 r
US20110150892A1 (en) 2008-08-11 2011-06-23 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
WO2010065939A1 (en) 2008-12-05 2010-06-10 Indiana University Research & Technology Corporation Combination therapy to enhace nk cell mediated cytotoxicty
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US20140348841A1 (en) 2009-09-03 2014-11-27 Merck Sharp & Dohme Corp. Anti-gitr antibodies
US8709424B2 (en) 2009-09-03 2014-04-29 Merck Sharp & Dohme Corp. Anti-GITR antibodies
WO2011028683A1 (en) 2009-09-03 2011-03-10 Schering Corporation Anti-gitr antibodies
WO2011041613A2 (en) 2009-09-30 2011-04-07 Memorial Sloan-Kettering Cancer Center Combination immunotherapy for the treatment of cancer
EP2482849A2 (en) 2009-09-30 2012-08-08 Memorial Sloan-Kettering Cancer Center Combination immunotherapy for the treatment of cancer
WO2011066389A1 (en) 2009-11-24 2011-06-03 Medimmmune, Limited Targeted binding agents against b7-h1
WO2011140249A2 (en) 2010-05-04 2011-11-10 Five Prime Therapeutics, Inc. Antibodies that bind csf1r
WO2011155607A1 (en) 2010-06-11 2011-12-15 協和発酵キリン株式会社 Anti-tim-3 antibody
WO2011159877A2 (en) 2010-06-18 2011-12-22 The Brigham And Women's Hospital, Inc. Bi-specific antibodies against tim-3 and pd-1 for immunotherapy in chronic immune conditions
WO2012027328A2 (en) 2010-08-23 2012-03-01 Board Of Regents, The University Of Texas System Anti-ox40 antibodies and methods of using the same
WO2012071411A2 (en) 2010-11-22 2012-05-31 Innate Pharma Sa Nk cell modulating treatments and methods for treatment of hematological malignancies
WO2012122444A1 (en) 2011-03-10 2012-09-13 Provectus Pharmaceuticals, Inc. Combination of local and systemic immunomodulative therapies for enhanced treatment of cancer
EP3147297A1 (en) 2011-03-31 2017-03-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies directed against icos and uses thereof
WO2012131004A2 (en) 2011-03-31 2012-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies directed against icos and uses thereof
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2012160448A2 (en) 2011-05-25 2012-11-29 Innate Pharma, S.A. Anti-kir antibodies for the treatment of inflammatory disorders
WO2013006490A2 (en) 2011-07-01 2013-01-10 Cellerant Therapeutics, Inc. Antibodies that specifically bind to tim3
WO2013028231A1 (en) 2011-08-23 2013-02-28 Board Of Regents, The University Of Texas System Anti-ox40 antibodies and methods of using the same
WO2013039954A1 (en) 2011-09-14 2013-03-21 Sanofi Anti-gitr antibodies
WO2013038191A2 (en) 2011-09-16 2013-03-21 Bioceros B.V. Anti-cd134 (ox40) antibodies and uses thereof
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies and uses thereof
WO2013132044A1 (en) 2012-03-08 2013-09-12 F. Hoffmann-La Roche Ag Combination therapy of antibodies against human csf-1r and uses thereof
WO2013173223A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2013181634A2 (en) 2012-05-31 2013-12-05 Sorrento Therapeutics Inc. Antigen binding proteins that bind pd-l1
US20140072565A1 (en) 2012-06-08 2014-03-13 National Cancer Center Novel epitope for switching to th17 cell and use thereof
US20140072566A1 (en) 2012-06-08 2014-03-13 National Cancer Center Novel epitope for switching to th2 cell and use thereof
US20140065152A1 (en) 2012-06-08 2014-03-06 National Cancer Center Novel epitope for switching to th1 cell and use thereof
US20140093511A1 (en) 2012-07-02 2014-04-03 Bristol-Myers Squibb Company Optimization of antibodies that bind lymphocyte activation gene-3 (lag-3), and uses thereof
WO2014055648A1 (en) 2012-10-02 2014-04-10 Bristol-Myers Squibb Company Combination of anti-kir antibodies and anti-pd-1 antibodies to treat cancer
WO2014140180A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
WO2014151634A1 (en) 2013-03-15 2014-09-25 Bristol-Myers Squibb Company Macrocyclic inhibitors of the pd-1/pd-l1 and cd80(b7-1)/pd-l1 protein/protein interactions
WO2014148895A1 (en) 2013-03-18 2014-09-25 Biocerox Products B.V. Humanized anti-cd134 (ox40) antibodies and uses thereof
WO2014179664A2 (en) 2013-05-02 2014-11-06 Anaptysbio, Inc. Antibodies directed against programmed death-1 (pd-1)
WO2014194302A2 (en) 2013-05-31 2014-12-04 Sorrento Therapeutics, Inc. Antigen binding proteins that bind pd-1
US20160272708A1 (en) 2013-06-26 2016-09-22 Shanghai Junshi Biosciences Inc. Anti-pd-1 antibody and use thereof
WO2014206107A1 (en) 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 Anti-pd-1 antibody and use thereof
WO2015026684A1 (en) 2013-08-20 2015-02-26 Merck Sharp & Dohme Corp. Modulation of tumor immunity
WO2015031667A2 (en) 2013-08-30 2015-03-05 Amgen Inc. Gitr antigen binding proteins
WO2015034820A1 (en) 2013-09-04 2015-03-12 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2015035606A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-pd1 antibodies and their use as therapeutics and diagnostics
US20150079109A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
WO2015042246A1 (en) 2013-09-20 2015-03-26 Bristol-Myers Squibb Company Combination of anti-lag-3 antibodies and anti-pd-1 antibodies to treat tumors
WO2015085847A1 (en) 2013-12-12 2015-06-18 上海恒瑞医药有限公司 Pd-1 antibody, antigen-binding fragment thereof, and medical application thereof
WO2015112800A1 (en) 2014-01-23 2015-07-30 Regeneron Pharmaceuticals, Inc. Human antibodies to pd-1
WO2015112900A1 (en) 2014-01-24 2015-07-30 Dana-Farber Cancer Institue, Inc. Antibody molecules to pd-1 and uses thereof
WO2015117002A1 (en) 2014-01-31 2015-08-06 Novartis Ag Antibody molecules to tim-3 and uses thereof
WO2015153513A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Anti-ox40 antibodies and methods of use
WO2015153514A1 (en) 2014-03-31 2015-10-08 Genentech, Inc. Combination therapy comprising anti-angiogenesis agents and ox40 binding agonists
WO2015160641A2 (en) 2014-04-14 2015-10-22 Bristol-Myers Squibb Company Compounds useful as immunomodulators
US20170260271A1 (en) 2014-05-13 2017-09-14 Chugai Seiyaku Kabushiki Kaisha T Cell-Redirected Antigen-Binding Molecule For Cells Having Immunosuppression Function
WO2015184099A1 (en) 2014-05-28 2015-12-03 4-Antibody Ag Anti-gitr antibodies and methods of use thereof
WO2015200119A1 (en) 2014-06-26 2015-12-30 Macrogenics, Inc. Covalently bonded diabodies having immunoreactivity with pd-1 and lag-3, and methods of use thereof
WO2016028672A1 (en) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anti-lag3 antibodies and antigen-binding fragments
WO2016032334A1 (en) 2014-08-28 2016-03-03 Academisch Ziekenhuis Leiden H.O.D.N. Lumc Cd94/nkg2a and/or cd94/nkg2b antibody, vaccine combinations
WO2016039749A1 (en) 2014-09-11 2016-03-17 Bristol-Myers Squibb Company Macrocyclic inhibitors of the pd-1/pd-l1 and cd80 (b7-1)/pd-li protein/protein interactions
WO2016041945A1 (en) 2014-09-16 2016-03-24 Innate Pharma Neutralization of inhibitory pathways in lymphocytes
WO2016041947A1 (en) 2014-09-16 2016-03-24 Innate Pharma Treatment regimens using anti-nkg2a antibodies
WO2016057667A1 (en) 2014-10-10 2016-04-14 Medimmune, Llc Humanized anti-ox40 antibodies and uses thereof
WO2016057624A1 (en) 2014-10-10 2016-04-14 Bristol-Myers Squibb Company Immunomodulators
WO2016068803A1 (en) 2014-10-27 2016-05-06 Agency For Science, Technology And Research Anti-tim-3 antibodies
WO2016068802A1 (en) 2014-10-27 2016-05-06 Agency For Science, Technology And Research Anti-tim-3 antibodies
WO2016071448A1 (en) 2014-11-06 2016-05-12 F. Hoffmann-La Roche Ag Anti-tim3 antibodies and methods of use
WO2016077518A1 (en) 2014-11-14 2016-05-19 Bristol-Myers Squibb Company Macrocyclic peptides useful as immunomodulators
WO2016100285A1 (en) 2014-12-18 2016-06-23 Bristol-Myers Squibb Company Immunomodulators
WO2016100608A1 (en) 2014-12-19 2016-06-23 Bristol-Myers Squibb Company Immunomodulators
WO2016106159A1 (en) 2014-12-22 2016-06-30 Enumeral Biomedical Holding, Inc. Anti-pd-1 antibodies
WO2016106302A1 (en) 2014-12-23 2016-06-30 Bristol-Myers Squibb Company Antibodies to tigit
WO2016111947A2 (en) 2015-01-05 2016-07-14 Jounce Therapeutics, Inc. Antibodies that inhibit tim-3:lilrb2 interactions and uses thereof
US9771424B2 (en) 2015-01-28 2017-09-26 Glaxosmithkline Intellectual Property Development Limited ICOS binding proteins
US9738718B2 (en) 2015-01-28 2017-08-22 Glaxosmithkline Intellectual Property Development Limited ICOS binding proteins
WO2016126858A2 (en) 2015-02-03 2016-08-11 Anaptysbio, Inc. Antibodies directed against lymphocyte activation gene 3 (lag-3)
WO2016126646A1 (en) 2015-02-04 2016-08-11 Bristol-Myers Squibb Company Immunomodulators
WO2016134371A2 (en) 2015-02-20 2016-08-25 Ohio State Innovation Foundation Bivalent antibody directed against nkg2d and tumor associated antigens
WO2016144803A2 (en) 2015-03-06 2016-09-15 Sorrento Therapeutics, Inc. Antibody therapeutics that bind tim3
WO2016149201A2 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
WO2016149351A1 (en) 2015-03-18 2016-09-22 Bristol-Myers Squibb Company Immunomodulators
WO2016154177A2 (en) 2015-03-23 2016-09-29 Jounce Therapeutics, Inc. Antibodies to icos
WO2016161270A1 (en) 2015-04-01 2016-10-06 Anaptysbio, Inc. Antibodies directed against t cell immunoglobulin and mucin protein 3 (tim-3)
WO2016196237A1 (en) 2015-05-29 2016-12-08 Agenus Inc. Anti-ctla-4 antibodies and methods of use thereof
WO2016196228A1 (en) 2015-05-29 2016-12-08 Bristol-Myers Squibb Company Antibodies against ox40 and uses thereof
WO2016200782A1 (en) 2015-06-08 2016-12-15 Macrogenics, Inc. Lag-3-binding molecules and methods of use thereof
WO2016200836A1 (en) 2015-06-08 2016-12-15 Genentech, Inc. Methods of treating cancer using anti-ox40 antibodies
WO2016197367A1 (en) 2015-06-11 2016-12-15 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-l1 antibodies
WO2017015560A2 (en) 2015-07-22 2017-01-26 Sorrento Therapeutics, Inc. Antibody therapeutics that bind lag3
WO2017019846A1 (en) 2015-07-30 2017-02-02 Macrogenics, Inc. Pd-1-binding molecules and methods use thereof
WO2017020291A1 (en) 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-l1 antibodies
WO2017020858A1 (en) 2015-08-06 2017-02-09 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-l1 antibodies
WO2017025498A1 (en) 2015-08-07 2017-02-16 Pieris Pharmaceuticals Gmbh Novel fusion polypeptide specific for lag-3 and pd-1
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017025016A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017025051A1 (en) 2015-08-11 2017-02-16 Wuxi Biologics (Shanghai) Co. Ltd. Novel anti-pd-1 antibodies
WO2017024515A1 (en) 2015-08-11 2017-02-16 Wuxi Biologics (Cayman) Inc. Novel anti-pd-1 antibodies
WO2017031242A1 (en) 2015-08-20 2017-02-23 Sutro Biopharma, Inc. Anti-tim-3 antibodies, compositions comprising anti-tim-3 antibodies and methods of making and using anti-tim-3 antibodies
WO2017034916A1 (en) 2015-08-24 2017-03-02 Eli Lilly And Company Pd-l1 ("programmed death-ligand 1") antibodies
WO2017040790A1 (en) 2015-09-01 2017-03-09 Agenus Inc. Anti-pd-1 antibodies and methods of use thereof
WO2017053748A2 (en) 2015-09-25 2017-03-30 Genentech, Inc. Anti-tigit antibodies and methods of use
WO2017055399A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Cellular based fret assay for the determination of simultaneous binding
WO2017055404A1 (en) 2015-10-02 2017-04-06 F. Hoffmann-La Roche Ag Bispecific antibodies specific for pd1 and tim3
WO2017062888A1 (en) 2015-10-09 2017-04-13 Regeneron Pharmaceuticals, Inc. Anti-lag3 antibodies and uses thereof
WO2017066227A1 (en) 2015-10-15 2017-04-20 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2017063162A1 (en) 2015-10-15 2017-04-20 苏州丁孚靶点生物技术有限公司 Anti-ox40 antibody and application thereof
WO2017079112A1 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and their uses
WO2017079116A2 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding pd-1 and tim-3 and their uses
WO2017079115A1 (en) 2015-11-03 2017-05-11 Janssen Biotech, Inc. Antibodies specifically binding tim-3 and their uses
WO2017087589A2 (en) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Pd1 and/or lag3 binders
WO2017086367A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Combination therapy using t cell redirection antigen binding molecule against cell having immunosuppressing function
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
WO2017087901A2 (en) 2015-11-19 2017-05-26 Sutro Biopharma, Inc. Anti-lag3 antibodies, compositions comprising anti-lag3 antibodies and methods of making and using anti-lag3 antibodies
WO2017096281A1 (en) 2015-12-02 2017-06-08 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
WO2017096179A1 (en) 2015-12-02 2017-06-08 Agenus Inc. Antibodies and methods of use thereof
WO2017096182A1 (en) 2015-12-03 2017-06-08 Agenus Inc. Anti-ox40 antibodies and methods of use thereof
WO2017106061A1 (en) 2015-12-14 2017-06-22 Macrogenics, Inc. Bispecific molecules having immunoreactivity with pd-1 and ctla-4, and methods of use thereof
WO2017106129A1 (en) 2015-12-16 2017-06-22 Merck Sharp & Dohme Corp. Anti-lag3 antibodies and antigen-binding fragments
WO2017123557A1 (en) 2016-01-11 2017-07-20 Armo Biosciences, Inc. Interleukin-10 in production of antigen-specific cd8+ t cells and methods of use of same
WO2017132827A1 (en) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017133540A1 (en) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017132825A1 (en) 2016-02-02 2017-08-10 华为技术有限公司 Emission power verification method, user equipment, and base station
WO2017134292A1 (en) 2016-02-04 2017-08-10 Glenmark Pharmaceuticals S.A. Anti-ox40 antagonistic antibodies for the treatment of atopic dermatitis
WO2017149143A1 (en) 2016-03-04 2017-09-08 Agency For Science, Technology And Research Anti-lag-3 antibodies
WO2017151830A1 (en) 2016-03-04 2017-09-08 Bristol-Myers Squibb Company Immunomodulators
WO2017176608A1 (en) 2016-04-05 2017-10-12 Bristol-Myers Squibb Company Macrocyclic inhibitors of the pd-1/pd-l1 and cd80/pd-l1 protein/protein interactions
WO2017178493A1 (en) 2016-04-12 2017-10-19 Symphogen A/S Anti-tim-3 antibodies and compositions
WO2017198741A1 (en) 2016-05-18 2017-11-23 Boehringer Ingelheim International Gmbh Anti pd-1 and anti-lag3 antibodies for cancer treatment
WO2017205721A1 (en) 2016-05-27 2017-11-30 Agenus Inc. Anti-tim-3 antibodies and methods of use thereof
WO2017220555A1 (en) 2016-06-20 2017-12-28 F-Star Beta Limited Lag -3 binding members
WO2017220988A1 (en) 2016-06-20 2017-12-28 Kymab Limited Multispecific antibodies for immuno-oncology
WO2017220569A1 (en) 2016-06-20 2017-12-28 F-Star Delta Limited Binding molecules binding pd-l1 and lag-3
WO2017219995A1 (en) 2016-06-23 2017-12-28 江苏恒瑞医药股份有限公司 Lag-3 antibody, antigen-binding fragment thereof, and pharmaceutical application thereof
WO2018009505A1 (en) 2016-07-08 2018-01-11 Bristol-Myers Squibb Company 1,3-dihydroxy-phenyl derivatives useful as immunomodulators
WO2018013818A2 (en) 2016-07-14 2018-01-18 Bristol-Myers Squibb Company Antibodies against tim3 and uses thereof
WO2018034227A1 (en) 2016-08-15 2018-02-22 国立大学法人北海道大学 Anti-lag-3 antibody
WO2018039020A1 (en) 2016-08-25 2018-03-01 Eli Lilly And Company Anti-tim-3 antibodies
WO2018036561A1 (en) 2016-08-26 2018-03-01 Beigene, Ltd. Anti-tim-3 antibodies and use thereof
WO2018044963A1 (en) 2016-09-01 2018-03-08 Bristol-Myers Squibb Company Biaryl compounds useful as immunomodulators
WO2018071500A1 (en) 2016-10-11 2018-04-19 Agenus Inc. Anti-lag-3 antibodies and methods of use thereof
WO2018069500A2 (en) 2016-10-13 2018-04-19 Symphogen A/S Anti-lag-3 antibodies and compositions
WO2018083087A2 (en) 2016-11-02 2018-05-11 Glaxosmithkline Intellectual Property (No.2) Limited Binding proteins
WO2018085750A2 (en) 2016-11-07 2018-05-11 Bristol-Myers Squibb Company Immunomodulators
WO2018118848A1 (en) 2016-12-20 2018-06-28 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2018183171A1 (en) 2017-03-27 2018-10-04 Bristol-Myers Squibb Company Substituted isoquionline derivatives as immunomudulators
WO2018237153A1 (en) 2017-06-23 2018-12-27 Bristol-Myers Squibb Company Immunomodulators acting as antagonists of pd-1
WO2019070643A1 (en) 2017-10-03 2019-04-11 Bristol-Myers Squibb Company Immunomodulators
WO2019147662A1 (en) 2018-01-23 2019-08-01 Bristol-Myers Squibb Company 2,8-diacyl-2,8-diazaspiro[5.5]undecane compounds useful as immunomodulators
WO2019169123A1 (en) 2018-03-01 2019-09-06 Bristol-Myers Squibb Company Compounds useful as immunomodulators
WO2020243563A1 (en) * 2019-05-30 2020-12-03 Bristol-Myers Squibb Company Multi-tumor gene signatures for suitability to immuno-oncology therapy
WO2021092220A1 (en) * 2019-11-06 2021-05-14 Bristol-Myers Squibb Company Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy

Non-Patent Citations (31)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. AAB59385
"NCBI", Database accession no. 29126
"Oxford Dictionary Of Biochemistry And Molecular Biology, Revised", 2000, OXFORD UNIVERSITY PRESS
"The Dictionary of Cell and Molecular Biology", 1999, ACADEMIC PRESS
ANGUIANO ESPERANZA ET AL: "Comparison of gene expression profiling platforms: translatability of tumor inflammation gene signatures Scientific Content on Demand Acknowledgments", AMERICAN ASSOCIATION FOR CANCER RESEARCH (AACR) VIRTUAL ANNUAL MEETING II, JUNE 22-24, 2020, 1 January 2014 (2014-01-01), pages 1 - 1, XP055905451, Retrieved from the Internet <URL:https://www.htgmolecular.com/assets/htg/publications/Anguiano_E._Comparison_of_gene_expression_profiling_platforms_translatability_of_tumor_inflammation_gene_singatures_BMS_Poster_.pdf> [retrieved on 20220325] *
BARNES ET AL., BR J CANCER, vol. 117, 2017, pages 451 - 460
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
CRISTESCU ET AL., SCIENCE, vol. 362, 2018, pages 3593
DANAHER P ET AL., J IMMUNOTHER CANCER, vol. 5, no. 18, 2017
DANAHER P ET AL., J IMMUNOTHER CANCER, vol. 6, no. 63, 2018
DARVIN P ET AL., EXP MOL MED, vol. 50, 2018, pages 165
DESAI ET AL., JCO, vol. 36, 2018
GORELIK ET AL., AACR:ABSTRACT, vol. 4606, April 2016 (2016-04-01)
HERBST ET AL., J CLIN ONCOL, vol. 31, 2013, pages 3000
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
JUO, PEI-SHOW: "Concise Dictionary of Biomedicine and Molecular Biology", 2002, CRC PRESS
KAPLON ET AL., MABS, vol. 10, no. 2, 2018, pages 183 - 203
RIBAS, UPDATE CANCER THER., vol. 2, no. 3, 2007, pages 133 - 39
SCHAER ET AL., CURR OPIN IMMUNOL., vol. 24, no. 2, 2012, pages 217 - 224
SI-YANG LIU ET AL., J. HEMATOL. ONCOL., vol. 10, 2017, pages 136
SJOBLOM ET AL., SCIENCE, vol. 314, no. 5797, 2006, pages 268 - 274
STOLL ET AL., ONCOTARGET, vol. 6, 2015, pages 11894 - 11909
SZABO ET AL., J CLIN ONCOL, vol. 37, 2019
SZABO ET AL., J IMMUNOTHER CANCER, vol. 7, 2019
SZABO PETER A ET AL: "Single-cell transcriptomics of human T cells reveals tissue and activation signatures in health and disease", NATURE COMMUNICATIONS, vol. 10, no. 1, 17 October 2019 (2019-10-17), pages 4706 - 16, XP055846416, ISSN: 2041-1723, Retrieved from the Internet <URL:https://www.nature.com/articles/s41467-019-12464-3.pdf> DOI: 10.1038/s41467-019-12464-3 *
SZABO PETER N ET AL: "P109 Predictive performance of a CD8-derived signature by gene expression profiling in patients with urothelial carcinoma from CheckMate 275, 34th Annual Meeting & Pre-Conference Programs of the Society for Immunotherapy of Cancer (SITC 2019): part 1 : National Harbor, MD, USA. 6-10 November 2019", JOURNAL FOR IMMUNOTHERAPY OF CANCER, 6 November 2019 (2019-11-06), England, pages 282 - 282, XP055905127, Retrieved from the Internet <URL:https://jitc.bmj.com/content/jitc/7/Suppl_1/282.full.pdf> [retrieved on 20220324], DOI: 10.1186/s40425-019-0763-1 *
SZABO PETER: "Poster 2593 Association of an Inflammatory Gene Signature With CD8 Expression by Immunohistochemistry in Multiple Tumor Types", PRESENTED AT THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY ANNUAL MEETING, MAY 31-JUNE 4, 2019, CHICAGO, IL, 13 December 2018 (2018-12-13), United States, pages 1 - 1, XP055905160, Retrieved from the Internet <URL:https://www.htgmolecular.com/assets/htg/publications/SZABO_ASCO2019_Association_poster_(002).pdf> [retrieved on 20220324], DOI: 10.1038/s12276-018-0191-1 *
WANG BEN ET AL: "Analysis of genetic signatures of tumor microenvironment yields insight into mechanisms of resistance to immunotherapy", 20 March 2020 (2020-03-20), XP055905079, Retrieved from the Internet <URL:https://www.biorxiv.org/content/10.1101/2020.03.17.980789v1.full.pdf> [retrieved on 20220324], DOI: 10.1101/2020.03.17.980789 *
WANG ET AL., CANCER IMMUNOL RES., vol. 2, no. 9, 2014, pages 846 - 56
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
ZHANG ET AL., CELL DISCOV., vol. 7, March 2017 (2017-03-01), pages 3

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20220292661A1 (en) * 2021-03-10 2022-09-15 Adlink Technology Inc. Automated inspection system and operating method thereof

Similar Documents

Publication Publication Date Title
US20220002413A1 (en) Treatment of renal cell carcinoma
US20210301023A1 (en) Diagnostic methods and compositions for cancer immunotherapy
JP2023113613A (en) Predictive peripheral blood biomarker for checkpoint inhibitors
US20220363760A1 (en) Multi-tumor gene signature for suitability to immuno-oncology therapy
CN113396230A (en) Methods of diagnosis and treatment of cancer
WO2021092220A1 (en) Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy
WO2022120179A1 (en) Multi-tumor gene signatures and uses thereof
US20230303700A1 (en) Cell localization signature and immunotherapy
US20220041733A1 (en) Methods of treating tumor
EP3976832A1 (en) Methods of identifying a subject suitable for an immuno-oncology (i-o) therapy
US20220259669A1 (en) Methods of identifying a subject suitable for an immuno-oncology (i-o) therapy
US20220233691A1 (en) Cell localization signature and combination therapy
US20220339249A1 (en) Composite biomarker for cancer therapy
WO2023168404A1 (en) Methods of treating a tumor

Legal Events

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

Ref document number: 21831434

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21831434

Country of ref document: EP

Kind code of ref document: A1