WO2021092221A1 - Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy - Google Patents

Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy Download PDF

Info

Publication number
WO2021092221A1
WO2021092221A1 PCT/US2020/059174 US2020059174W WO2021092221A1 WO 2021092221 A1 WO2021092221 A1 WO 2021092221A1 US 2020059174 W US2020059174 W US 2020059174W WO 2021092221 A1 WO2021092221 A1 WO 2021092221A1
Authority
WO
WIPO (PCT)
Prior art keywords
genes
hla
antibody
pcl
tumor
Prior art date
Application number
PCT/US2020/059174
Other languages
French (fr)
Inventor
Neeraj ADYA
Patrik VITAZKA
David FELTQUATE
Bruce David CAR
Steven Gary NADLER
Joseph Daniel SZUSTAKOWSKI
Peter M. SZABO
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 WO2021092221A1 publication Critical patent/WO2021092221A1/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
    • C12Q2600/156Polymorphic or mutational markers
    • 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 a checkpoint inhibitor, e.g, an immunotherapy.
  • a checkpoint inhibitor e.g, an immunotherapy.
  • cancer immunotherapy had focused substantial effort on approaches that enhance anti-tumor immune responses by adoptive-transfer of activated effector cells, immunization against relevant antigens, or providing non-specific immune-stimulatory agents such as cytokines.
  • nivolumab and pembrolizumab (formerly lambrolizumab; US AN Council Statement, 2013) that bind specifically to the Programmed Death -1 (PD-1) receptor and block the inhibitory PD-l/PD-1 ligand pathway (Topalian et al., 2012a, b; Topalian et al., 2014; Hamid et al., 2013; Hamid and Carvajal, 2013; McDermott and Atkins, 2013).
  • 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.
  • Certain aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.
  • TMB tumor mutation burden
  • Certain aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising: (a) (a) determining whether the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD- L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined; and (b) administering an effective amount of a checkpoint inhibitor to the subject.
  • TMB tumor mutation burden
  • Certain aspects of the present disclosure are directed to a method of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy, comprising any two or more of the following: (i) determining whether the subject has a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) detecting a level of PD-L1 expression in tumor cells of the tumor; and (iii) measuring a TMB status of a biological sample obtained from the subject.
  • the method further comprises administering to the subject a checkpoint inhibitor if the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression, and (iii) the subject exhibits a TMB status of at least about 10 mutations per megabase of genes examined.
  • Certain aspects of the present disclosure are directed to a checkpoint inhibitor for treating a tumor in a human subject in need thereof, wherein the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.
  • TMB tumor mutation burden
  • Certain aspects of the present disclosure are directed to a combination of assays for identifying a tumor in a human subject in need thereof, wherein the combination of assays comprises at least two of the following: (i) the subject is to be determined for having a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject [0012] (ii) a level of PD-L1 expression in tumor cells of the tumor is to be detected; and (iii) the subject is to be measured for a TMB status of a biological sample obtained from the subject.
  • the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA-4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR,
  • the panel of genes comprises at least about 5 additional genes, at least about 10 additional genes, at least about 15 additional genes, at least about 20 additional genes, at least about 25 additional genes, at least about 30 additional genes, at least about 35 additional genes, at least about 40 additional genes, at least about 45 additional genes, at least about 50 additional genes, at least about 55 additional genes, at least about 60 additional genes, at least about 65 additional genes, at least about 70 additional genes, at least about 75 additional genes, at least about 80 additional genes, at least about 85 additional genes, at least about 90 additional genes, at least about 95 additional genes, at least about 100 additional genes, at least about 110 additional genes, at least about 120 additional genes, at least about 130 additional genes, at least about 140 additional genes, at least about 150 additional genes, at least about 160 additional genes, at least about 170 additional genes, at least about 180 additional genes, at least about 190 additional genes, or at least about 200 additional genes.
  • At least one of the additional genes is selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD
  • the panel of genes comprises at least about 63 genes. [0015] In some aspects, the panel of genes comprises at least one housekeeping gene. In some aspects, the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 housekeeping genes.
  • the housekeeping genes are 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 panel of genes comprises at least one control gene.
  • the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 control genes.
  • control genes are selected from the group consisting of ANT1, 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 panel of genes comprises at least about 95 genes.
  • the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10,
  • the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1,
  • the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR- Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2,
  • the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PR
  • a parenchymal inflammatory phenotype is characterized by a low signature score, wherein the signature score is determined by measuring the composite expression of the genes in the gene panel in a tumor sample form the subject, and comparing the composite expression of the genes in the gene panel to the average composite expression of the panel of genes in tumor samples obtained from a population of subjects afflicted with the tumor.
  • the average composite signature score is determined by measuring the composite expression of the panel of genes in tumor samples obtained from the population of subjects.
  • the low signature score is characterized by a signature score of less than about 50, less than about 45, less than about 40, less than about 35, less than about 30, less than about 25, less than about 20, less than about 15, less than about 10, or less than about 5; wherein a signature score of 100 indicates that the subject has no parenchymal inflammatory phenotype; and wherein a signature score of 1 indicates that a subject has a high parenchymal phenotype.
  • the TMB status is determined by sequencing nucleic acids in the tumor and identifying a genomic alteration in the sequenced nucleic acids.
  • alteration comprises one or more somatic mutations.
  • the genomic alteration comprises one or more nonsynonymous mutations.
  • the genomic alteration comprises one or more missense mutations. In some aspects, the genomic alteration comprises one or more alterations selected from the group consisting of a base pair substitution, a base pair insertion, a base pair deletion, a copy number alteration (CNAs), a gene rearrangement, and any combination thereof.
  • CNAs copy number alteration
  • the TMB status of the tumor comprises at least 10 mutations, at least about 11 mutations, at least about 12 mutations, at least about 13 mutations, at least about 14 mutations, at least about 15 mutations, at least about 16 mutations, at least about 17 mutations, at least about 18 mutations, at least about 19 mutations, at least about 20 mutations, at least about 21 mutations, at least about 22 mutations, at least about 23 mutations, at least about 24 mutations, at least about 25 mutations, at least about 26 mutations, at least about 27 mutations, at least about 28 mutations, at least about 29 mutations, or at least about 30 mutations per megabase of genome examined as measured by a FOUNDATIONONE® CDXTM assay.
  • PD-L1 expression is determined using an IHC assay.
  • the IHC assay is an automated IHC assay.
  • the tumor sample is a tumor tissue biopsy.
  • the tumor sample is a formalin-fixed, paraffin-embedded tumor tissue or a fresh-frozen tumor tissue.
  • the expression of the genes in the panel of genes is determined by measuring the level of mRNA, the level of a protein encoded by the gene, or both.
  • the level of mRNA is determined using reverse transcriptase PCR.
  • the level of the protein encoded by the gene is determined using an IHC assay.
  • the IHC assay is an automated IHC assay.
  • the checkpoint inhibitor comprises a polypeptide, a small molecule, a polynucleotide, or any combination thereof. In some aspects, the checkpoint inhibitor comprises an antibody or an antigen-binding fragment thereof.
  • the antibody or antigen-binding fragment thereof specifically binds a target selected from PD-1 ("anti-PD-1 antibody”), PD-L1 ("anti-PD-L1 antibody”), CTLA-4, LAG3, TIGIT, TIM3, NKG2a, OX40, ICOS, CD137, KIR, TGF ⁇ , IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, and any combination thereof.
  • the antibody is a chimeric, humanized or human monoclonal antibody or a portion thereof.
  • the anti-PD-1 antibody cross-competes with nivolumab for binding to human PD-1. In some aspects, the anti-PD-1 antibody binds to the same epitope as nivolumab. In some aspects, the anti-PD-1 antibody comprises a heavy chain constant region which is of a human IgG1 or IgG4 isotype. In some aspects, the anti-PD-1 antibody is nivolumab. In some aspects, the anti-PD-1 antibody is pembrolizumab. [0028] In some aspects, the anti-PD-1 antibody is administered at a dose ranging from at least about 0.1 mg/kg to at least about 10.0 mg/kg body weight once about every 1, 2 or 3 weeks.
  • the anti-PD-1 antibody is administered at a dose of at least about 3 mg/kg body weight once about every 2 weeks. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose. In some aspects, the anti-PD-1 antibody or antigen- binding portion thereof is administered at a flat dose of at least about 200, at least about 220, at least about 240, at least about 260, at least about 280, at least about 300, at least about 320, at least about 340, at least about 360, at least about 380, at least about 400, at least about 420, at least about 440, at least about 460, at least about 480, at least about 500 or at least about 550 mg.
  • the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 240 mg. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 480 mg. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose about once every 1, 2, 3 or 4 weeks. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose or about 240 mg once about every two weeks. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 480 mg once about every four weeks.
  • the anti-PD-1 antibody is administered for as long as clinical benefit is observed or until unmanageable toxicity or disease progression occurs.
  • the antibody is formulated for intravenous administration. In some aspects, the antibody is administered at a subtherapeutic dose.
  • the methods disclosed herein further comprise administering an antibody or an antigen binding fragment thereof that binds specifically to CTLA-4 ("an anti- CTLA-4 antibody").
  • the anti-CTLA-4 antibody cross-competes with ipilimumab or tremelimumab for binding to human CTLA-4.
  • the anti-CTLA-4 antibody binds to the same epitope as ipilimumab or tremelimumab.
  • the anti-CTLA-4 antibody is ipilimumab. In some aspects, the anti-CTLA-4 antibody is tremelimumab. In some aspects, the anti-CTLA-4 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. In some aspects, the anti-CTLA-4 antibody is administered at a dose of 1 mg/kg body weight once every 6 weeks. In some aspects, the anti-CTLA-4 antibody is administered at a dose of 1 mg/kg body weight once every 4 weeks. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose.
  • the anti-CTLA-4 antibody is administered at a flat dose of at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least about 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg,at least about 180 mg, at least about 190 mg, or at least about 200 mg.
  • the anti-CTLA-4 antibody is administered as a flat dose about once every 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the tumor is derived from a cancer selected from the group consisting of hepatocellular cancer, gastroesophageal cancer, melanoma, bladder cancer, lung cancer, kidney cancer, head and neck cancer, colon cancer, and any combination thereof.
  • the tumor is derived from a melanoma.
  • the tumor is derived from a small cell carcinoma of the head and neck.
  • the tumor is relapsed.
  • the tumor is refractory.
  • the tumor is refractory following at least one prior therapy comprising administration of at least one anticancer agent.
  • the at least one anticancer agent comprises a standard of care therapy.
  • the at least one anticancer agent comprises an immunotherapy.
  • the tumor is locally advanced.
  • the tumor is metastatic.
  • the administering treats the tumor. [0033]
  • the administering reduces the size of the tumor. In some aspects, the size of the tumor is reduced by at least about 10%, about 20%, about 30%, about 40%, or about 50% compared to the tumor size prior to the administration.
  • the subject exhibits progression-free survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about eighteen months, at least about two years, at least about three years, at least about four years, or at least about five years after the initial administration.
  • the subject exhibits stable disease after the administration.
  • the subject exhibits a partial response after the administration.
  • the subject exhibits a complete response after the administration.
  • Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, (b) a PD-L1 expression pattern in a biological sample, and (c) a tumor mutation burden (TMB) status of a biological sample.
  • a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, (b) a PD-L1 expression pattern in a biological sample, and (c) a tumor mutation burden (TMB) status of a biological sample.
  • TMB tumor mutation burden
  • the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA-4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galect
  • the means for detecting a level of expression of a panel of genes in the biological sample comprises a first biological probe, wherein the first biological probe specifically interacts with a transcript of at least one gene in the panel of genes.
  • the device further comprises a means for detecting a level of expression of the transcript.
  • the transcript comprises an mRNA or a cDNA.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe, a third biological probe, a fourth biological probe, and a fifth biological probe.
  • the first biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a first gene of the at least 5 genes; wherein the second biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a second gene of the at least 5 genes; wherein the third biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a third gene of the at least 5 genes; wherein the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes; and wherein the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes.
  • the transcript of the first gene, the transcript of the second gene, the transcript of the third gene, and/or the transcript of the fourth gene comprises an mRNA or a cDNA.
  • the biological sample comprises a tumor sample from a human subject.
  • the tumor sample is a tumor tissue biopsy.
  • the tumor sample is a formalin-fixed, paraffin-embedded tumor tissue or a fresh-frozen tumor tissue.
  • the panel of genes comprises 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 110, at least about 120, at least about 130, at least about 140, at least about 150, at least about 160, at least about 170, at least about 180, at least about 190, or at least about 110 additional genes.
  • At least one of the additional genes are selected from CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF
  • the panel of genes comprises at least about 63 genes. [0038] In some aspects, the panel of genes comprises at least one housekeeping gene. In some aspects, the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 housekeeping genes.
  • the housekeeping genes are 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 panel of genes comprises at least one control gene.
  • the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 control genes.
  • control genes are selected from the group consisting of ANT1, 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 panel of genes comprises at least about 95 genes.
  • the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10,
  • the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1,
  • the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR- Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2,
  • the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PR
  • the means for detecting a PD-L1 expression pattern in a biological sample comprises means for conducting an IHC assay.
  • the IHC assay is an automated IHC assay.
  • the means for detecting a tumor mutation burden (TMB) status of a biological sample comprises an automated DNA sequencer.
  • the automated DNA sequencer is capable of whole exome sequencing (WES).
  • Certain aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject is identified as having a tumor suitable for a checkpoint inhibitor therapy, wherein the subject is identified as suitable by obtaining a biological sample from the subject and applying the biological sample to a device disclosed herein.
  • Certain aspects of the present disclosure are directed to a method of identifying a subject suitable for an immunotherapy, comprising obtaining a biological sample from the subject and applying the biological sample to the device a device disclosed herein. In some aspects, the method further comprises administering an immunotherapy to the subject.
  • FIGs.2A-2B are graphical representations of progression free survival (PFS; FIG. 2A) and overall survival (FIG.2B) in patients stratified by parenchymal signature score.
  • A low parenchymal score
  • B medium parenchymal score
  • C high parenchymal score. The number of patients at risk in each group is shown below the x-axis. (P ⁇ 0.01).
  • FIGs. 4A-4B are graphs that show the association of the parenchymal signature scores (log hazard ration) with progression-free survival (PFS; FIG. 4A) and overall survival (OS; FIGs.4B) at PD-L1 expression levels of 0% (FIGs.4A-4B), at least 1% (FIGs.4C-4D), and at least 5% as indicated.
  • FIGs.4C and 4D are graphical representations of overall survival (OS) in subjects identified as having ⁇ 1% tumor PD-L1 expression (FIG.
  • FIGs. 5A-5B are graphs that show the association of the parenchymal signature scores (log hazard ration) with progression-free survival (PFS; FIG. 5A) and overall survival (OS; FIGs. 5B) based on TMB scores, as indicated.
  • FIG. 5C is a chart showing the cutoffs for TMB used for the statistical analysis for FIGs. 5A-5B.
  • OS overall survival
  • the present disclosure provides a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.
  • TMB tumor mutation burden
  • Some aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising (a) determining whether the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a TMB status of at least about 10 mutations per megabase of genes examined; and administering an effective amount of a checkpoint inhibitor to the subject.
  • Some aspects of the present disclosure are directed to a method of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy, comprising any two or more of the following: determining whether the subject has a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; detecting a level of PD-L1 expression in tumor cells of the tumor; and measuring a TMB status of a biological sample obtained from the subject.
  • 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 intrasternal 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" (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.
  • 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 V H ) and a heavy chain constant region.
  • the heavy chain constant region comprises three constant domains, CH1, CH2 and CH3.
  • Each light chain comprises a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chain constant region comprises one constant domain, C L .
  • V H and V L 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 VH and VL 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 (C1q) 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 IgG1, IgG2, IgG3 and IgG4.
  • “Isotype” refers to the antibody class or subclass (e.g., IgM or IgG1) 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.
  • the term “antibody” also includes an antigen-binding fragment or an antigen- binding 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.
  • 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. For example, an anti-PD-1 antibody binds specifically to PD-1, an anti-PD-L1 antibody binds specifically to PD-L1, and 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 antigen- binding 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 CH1 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 CH1 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 V H domain; (vi) an isolated complementarity determining region (CDR) and
  • 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.
  • 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- L1), variants, isoforms, and species homologs of hPD-L1, and analogs having at least one common epitope with hPD-L1.
  • the complete hPD-L1 sequence can be found under GenBank Accession No. Q9NZQ7.
  • the human PD-L1 protein is encoded by the human CD274 gene (NCBI Gene ID: 29126).
  • 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).
  • LAG-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 P18627.
  • 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, that 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).
  • 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.
  • subject and patient are used interchangeably herein.
  • the use of the term “flat dose” with regard to the methods and dosages of the disclosure means a dose that is administered to a patient without regard for the weight or body surface area (BSA) of the patient. The flat dose is therefore not provided as a mg/kg dose, but rather as an absolute amount of the agent (e.g., the anti-PD-1 antibody).
  • the use of the term "fixed dose" with regard to a method of the disclosure means that two or more different antibodies in a single composition (e.g., anti-PD-1 antibody and anti- CTLA-4 antibody or an anti-PD-L1 antibody and an anti-CTLA-4 antibody) are present in the composition in particular (fixed) ratios with each other.
  • the fixed dose is based on the weight (e.g., mg) of the antibodies.
  • the fixed dose is based on the concentration (e.g., mg/ml) of the antibodies.
  • the ratio is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:15, about 1:20, about 1:30, about 1:40, about 1:50, about 1:60, about 1:70, about 1:80, about 1:90, about 1:100, about 1:120, about 1:140, about 1:160, about 1:180, about 1:200, about 200:1, about 180:1, about 160:1, about 140:1, about 120:1, about 100:1, about 90:1, about 80:1, about 70:1, about 60:1, about 50:1, about 40:1, about 30:1, about 20:1, about 15:1, about 10:1, about 9:1, about 8:1, about 7:1, about 6:1, about 5:1, about 4:1, about 3:1, or about 2:1 mg first antibody (e.g., anti-PD-1 antibody or an anti-PD-L1 antibody) to mg second antibody (e.g., anti-CTLA-4 antibody).
  • first antibody e.g
  • the 3:1 ratio of an anti-PD-1 antibody and an anti-CTLA-4 antibody can mean that a vial can contain about 240 mg of the anti-PD-1 antibody and 80 mg of the anti-CTLA-4 antibody or about 3 mg/ml of the anti-PD-1 antibody and 1 mg/ml of the anti- CTLA-4 antibody.
  • weight-based dose means that a dose that is administered to a patient is calculated based on the weight of the patient. For example, when a patient with 60 kg body weight requires 3 mg/kg of an anti-PD-1 antibody, one can calculate and use the appropriate amount of the anti-PD-1 antibody (i.e., 180 mg) for administration.
  • 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.
  • Promote 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.
  • 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.
  • 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.
  • a T cell e.g., an effector T cell, a Th cell, a CD4 + cell, a CD8 + T cell, or a Treg 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 long- term monitoring of the effects of these agents on the target disease.
  • 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).
  • 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.
  • 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.
  • 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.
  • tumor mutation burden refers to the number of somatic mutations in a tumor’s genome and/or the number of somatic mutations per area of the tumor’s genome. Germline (inherited) variants are excluded when determining TMB, because the immune system has a higher likelihood of recognizing these as self.
  • TMB Tumor mutation burden
  • TMB Tumor mutation burden
  • NGS next-generation sequencing
  • the NGS-based method is selected from whole genome sequencing (WGS), whole exome sequencing (WES), or comprehensive genomic profiling (CGP) of cancer gene panels such as FOUNDATIONONE CDXTM and MSK-IMPACT clinical tests.
  • TMB refers to the number of somatic mutations per megabase (Mb) of DNA sequenced.
  • Mb megabase
  • TMB is measured using the total number of nonsynonymous mutations, e.g., missense mutation (i.e. changing a particular amino acid in the protein) and/or nonsense (causing premature termination and thus truncation of the protein sequence), identified by normalizing matched tumor with germline samples to exclude any inherited germline genetic alterations.
  • TMB is measured using the total number of missense mutations in a tumor.
  • tissue sample for example, a minimum of 10 slides
  • TMB is expressed as NsMs per megabase (NsM/Mb).1 megabase represents 1 million bases.
  • the TMB status can be a numerical value or a relative value, e.g., high, medium, or low; within the highest fractile, or within the top tertile, of a reference set.
  • a TMB has a score of at least 210, at least 215, at least 220, at least 225, at least 230, at least 235, at least 240, at least 245, at least 250, at least 255, at least 260, at least 265, at least 270, at least 275, at least 280, at least 285, at least 290, at least 295, at least 300, at least 305, at least 310, at least 315, at least 320, at least 325, at least 330, at least 335, at least 340, at least 345, at least 350, at least 355, at least 360, at least 365, at least 370, at least 375, at least 380, at least 385, at least 390, at least 395, at least 400, at least 405, at least 210, at least 215, at least 220, at least 225, at least 230, at least 235, at least 240, at least 245, at least 250, at least 255, at least 260, at least 265,
  • a "high TMB” refers to a TMB within the highest fractile of the reference TMB value.
  • all subjects with evaluable TMB data are grouped according to fractile distribution of TMB, i.e., subjects are rank ordered from highest to lowest number of genetic alterations and divided into a defined number of groups.
  • all subjects with evaluable TMB data are ranked ordered and divided into thirds, and a "high TMB" is within the top tertile of the reference TMB value.
  • the tertile boundaries are 0 ⁇ 100 genetic alterations; 100 to 243 genetic alterations; and > 243 genetic alterations.
  • a "high TMB” refers to a TMB of at least about 20 mutations/tumor, at least about 25 mutations/tumor, at least about 30 mutations/tumor, at least about 35 mutations/tumor, at least about 40 mutations/tumor, at least about 45 mutations/tumor, at least about 50 mutations/tumor, at least about 55 mutations/tumor, at least about 60 mutations/tumor, at least about 65 mutations/tumor, at least about 70 mutations/tumor, at least about 75 mutations/tumor, at least about 80 mutations/tumor, at least about 85 mutations/tumor, at least about 90 mutations/tumor, at least about 95 mutations/tumor, or at least about 100 mutations/tum
  • a "high TMB” refers to a TMB of at least about 105 mutations/tumor, at least about 110 mutations/tumor, at least about 115 mutations/tumor, at least about 120 mutations/tumor, at least about 125 mutations/tumor, at least about 130 mutations/tumor, at least about 135 mutations/tumor, at least about 140 mutations/tumor, at least about 145 mutations/tumor, at least about 150 mutations/tumor, at least about 175 mutations/tumor, or at least about 200 mutations/tumor.
  • a tumor having a high TMB has at least about 100 mutations/tumor.
  • the "high TMB” can also be referred to as the number of mutations per megabase of tumor genome sequenced, e.g., as measured by a mutation assay, e.g., FOUNDATIONONE® CDXTM assay.
  • the high TMB refers to at least about 9, at least about 10, at least about 11, at least 12, at least about 13, at least about 14, at least about 15, at least about 16, at least about 17, at least about 18, at least about 19, or at least about 20 mutations per megabase of genome as measured by a FOUNDATIONONE® CDXTM assay.
  • the "high TMB” refers to at least 10 mutations per megabase of genome sequenced by a FOUNDATIONONE® CDXTM assay.
  • the term “medium TMB” refers to a number of somatic mutations in a tumor’s genome that is at or around a number of somatic mutations that is normal or average and the term “low TMB” refers to a number of somatic mutations in a tumor’s genome that is below a number of somatic mutations that is normal or average.
  • a “high TMB” has a score of at least 243
  • a “medium TMB” has a score of between 100 and 242
  • a “low TMB” has a score of less than 100 (or between 0 and 100).
  • the "medium or low TMB” refers to less than 9 mutations per megabase of genome sequenced, e.g., as measured by a FOUNDATIONONE® CDXTM assay.
  • the term "reference TMB value" as referred to herein can be the TMB value shown in Table 9.
  • TMB status can correlate with smoking status. In particular, subjects who currently or formerly smoke(d) often have more genetic alterations, e.g., missense mutations, than subjects who never smoke(d).
  • a tumor with a high TMB can also have a high neoantigen load.
  • the term "neoantigen” refers to a newly formed antigen that has not been previously recognized by the immune system.
  • a neoantigen can be a protein or peptide that is recognized as foreign (or non-self) by the immune system. Transcription of a gene in the tumor genome harboring a somatic mutation results in mutated mRNA that, when translated, gives rise to a mutated protein, which is then processed and transported to the ER lumen and binds to MHC class I complex, facilitating T-cell recognition of the neoantigen.
  • Neoantigen recognition can promote T-cell activation, clonal expansion, and differentiation into effector and memory T-cells.
  • Neoantigen load can correlate with TMB. In some aspects, TMB is assessed as a surrogate for measuring tumor neoantigen load.
  • the TMB status of a tumor can be used as a factor, alone or in combination with other factors, in determining whether a patient is likely to benefit from a particular anti-cancer agent or type of treatment or therapy, e.g., a combination therapy comprising (a) an anti-PD-1 antibody or an anti-PD-L1 antibody and (b) an anti-CTLA-4 antibody.
  • a high TMB status indicates an enhanced likelihood of benefit from immuno-oncology and, thus, can be used to identify patients more likely to benefit from therapy of a combination therapy comprising (a) an anti-PD-1 antibody or an anti-PD-L1 antibody and (b) an anti-CTLA-4 antibody.
  • a high TMB status and a high neoantigen load indicate an enhanced likelihood of benefit from immuno-oncology, e.g., a combination therapy comprising (a) an anti-PD-1 antibody or an anti-PD-L1 antibody and (b) an anti-CTLA-4 antibody.
  • the term "benefit from therapy” refers to an improvement in one or more of overall survival, progression-free survival, partial response, complete response, and overall response rate and can also include a reduction in tumor growth or size, 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.
  • Other factors e.g., environmental factors, can associate with TMB status. For example, smoking status of patients with NSCLC was correlated with TMB distribution, whereby current and former smokers had higher median TMB compared with those patients who had never smoked. See Peters et al., AACR, April 1-5, 2017, Washington, D.C.
  • driver mutation refers to an acquired alteration in DNA that occurs after conception. Somatic mutations can occur in any of the cells of the body except the germ cells (sperm and egg) and therefore are not passed on to children.
  • germline mutation refers to a gene change in a body's reproductive cell (egg or sperm) that becomes incorporated into the DNA of every cell in the body of the offspring. Germline mutations are passed on from parents to offspring. Also called a "hereditary mutation.” In the analysis of TMB, germline mutations are considered as a "baseline,” and are subtracted from the number of mutations found in the tumor biopsy to determine the TMB within the tumor. As germline mutations are found in every cell in the body, their presence can be determined via less invasive sample collections than tumor biopsies, such as blood or saliva.
  • Germline mutations can increase the risk of developing certain cancers, and can play a role in the response to chemotherapy.
  • the term “measuring” or “measured” or “measurement” when referring to TMB status means determining a measurable quantity of somatic mutations in a biological sample of the subject. It will be appreciated that measuring can be performed by sequencing nucleic acids, e.g., cDNA, mRNA, exoRNA, ctDNA, and cfDNA, in the sample. The measuring is performed on a subject's sample and/or a reference sample or samples and can, for example, be detected de novo or correspond to a previous determination.
  • the measuring can be performed, for example, using PCR methods, qPCR methods, Sanger sequencing methods, genomic profiling methods (including comprehensive gene panels), exome sequencing methods, genome sequencing methods, and/or any other method disclosed herein, as is known to a person of skill in the art.
  • the measuring identifies a genomic alteration in the sequenced nucleic acids.
  • the genomic (or gene) profiling methods can involve panels of a predetermined set of genes, e.g., 150-500 genes, and in some instances the genomic alterations evaluated in the panel of genes are correlated with total somatic mutations evaluated.
  • the term “gene” includes DNA coding regions (e.g., exons), DNA non-coding regions associated with a coding region (e.g., introns and promoters), and mRNA transcripts.
  • genomic alteration refers to a change (or mutation) in the nucleotide sequence of the genome of a tumor, which change is not present in the germline nucleotide sequence, and which in some aspects is a nonsynonymous mutation including, but not limited to, a base pair substitution, a base pair insertion, a base pair deletion, a copy number alteration (CNA), a gene rearrangement, and any combination thereof.
  • the genomic alterations measured in the biological sample are missense mutations.
  • the term “whole genome sequencing” or “WGS,” as used herein, refers to a method of sequencing the entire genome.
  • the term “whole exome sequencing” or “WES,” as used herein, refers to a method of sequencing all the protein-coding regions (exons) of the genome.
  • 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.
  • genomic profiling assay “comprehensive genomic profiling,” or “CGP” refers to an assay that analyzes a panel of genes and select introns for in vitro diagnosis.
  • CGP is a combination of NGS and targeted bioinformatics analysis to screen for mutations in known clinically relevant cancer genes. This method can be used to catch mutations that are missed by testing "hotspots" (e.g., BRCA1/BRCA2 mutations or microsatellite markers).
  • the CGP further includes one or more mRNA transcript, non-coding RNA, and/or promoter region.
  • the genes in the panel are cancer-related genes.
  • a genomic profiling assay is a FOUNDATIONONE ® assay.
  • the term "concordance,” as used herein, refers to a degree of agreement between two measurements and/or diagnostic tests. Concordance can be established using both qualitative and quantitative methods. Quantitative methods to assess concordance differ based on the type of measurement. A particular measurement can be expressed either as 1) a categorical/dichotomized variable or 2) a continuous variable. A "categorical/dichotomized variable" (e.g., above or below TMB cut-off) may use percent agreements, such as overall percent agreement (OPA), positive percent agreement (PPA), or negative percent agreement (NPA), to assess concordance.
  • OPA overall percent agreement
  • PPA positive percent agreement
  • NPA negative percent agreement
  • the term “analytical concordance” refers to the degree of agreement in the performance (e.g., identification of biomarkers, genomic alteration types, and genomic signatures, and assessment of test reproducibility) of two assays or diagnostic tests to support clinical use.
  • the term “clinical concordance” refers to the degree of agreement in how the two assays or diagnostic tests correlate with clinical outcome.
  • 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 TMB, 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 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.
  • “Once about every week” can include every seven days ⁇ one day, i.e., every six days to every eight days.
  • “Once about every two weeks” can include every fourteen days ⁇ three days, i.e., every eleven days to every seventeen days. Similar approximations apply, for example, to once about every three weeks, once about every four weeks, once about every five weeks, once about every six weeks, and once about every twelve weeks.
  • a dosing interval of once about every six weeks or once about every twelve weeks means that the first dose can be administered any day in the first week, and then the next dose can be administered any day in the sixth or twelfth week, respectively.
  • a dosing interval of once about every six weeks or once about every twelve weeks means that the first dose is administered on a particular day of the first week (e.g., Monday) and then the next dose is administered on the same day of the sixth or twelfth weeks (i.e., Monday), respectively.
  • the use of the alternative should be understood to mean either one, both, or any combination thereof of the alternatives.
  • the indefinite articles "a” or “an” should be understood to refer to "one or more" of any recited or enumerated component.
  • 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.
  • “about” or “comprising essentially of” can mean within 1 or more than 1 standard deviation per the practice in the art.
  • “about” or “comprising essentially of” can mean a range of up to 10%.
  • the terms can mean up to an order of magnitude or up to 5-fold of a value.
  • any concentration range, percentage range, ratio range or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated.
  • Abbreviations used herein are defined throughout the present disclosure. A list of additional abbreviations is provided in Table 1. Table 1: List of Abbreviations
  • the present disclosure is directed to methods of treating a tumor in a human subject, comprising administering an effective amount of a checkpoint inhibitor to the subject, e.g., an anti-PD-1 antibody, anti-PD-L1 antibody, and/or anti-LAG-3 antibody, wherein the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.
  • TMB tumor mutation burden
  • Certain aspects of the present disclosure are directed to methods of identifying a subject having a parenchymal inflammatory phenotype, determined by measuring the expression of a panel of genes comprising at least about 5 genes in a sample obtained from the subject.
  • the panel of genes comprises at least about 5 genes, at least about 6 genes, at least about 7 genes, at least about 8 genes, at least about 9 genes, at least about 10 genes, at least about 11 genes, at least about 12 genes, at least about 13 genes, at least about 14 genes, at least about 15 genes, at least about 16 genes, at least about 17 genes, at least about 18 genes, at least about 19 genes, at least about 20 genes, at least about 25 genes, at least about 30 genes, at least about 35 genes, at least about 40 genes, at least about 45 genes, at least about 50 genes, at least about 55 genes, at least about 60 genes, at least about 63 genes, at least about 65 genes, at least about 70 genes, at least about 80 genes, at least about 85 genes, at least about 90 genes, or at least about 95 genes.
  • the panel of genes comprises at least about 10 genes. In some aspects, the panel of genes comprises at least about 20 genes. In some aspects, the panel of genes comprises at least about 30 genes. In some aspects, the panel of genes comprises at least about 40 genes. In some aspects, the panel of genes comprises at least about 50 genes. In some aspects, the panel of genes comprises at least about 60 genes. In some aspects, the panel of genes comprises at least about 70 genes. In some aspects, the panel of genes comprises at least about 80 genes. In some aspects, the panel of genes comprises at least about 90 genes. In some aspects, the panel of genes comprises at least about 63 genes. In some aspects, the panel of genes comprises at least about 95 genes.
  • the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA- 4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6,
  • the panel of genes comprises at least about 5 additional genes, at least about 10 additional genes, at least about 15 additional genes, at least about 20 additional genes, at least about 25 additional genes, at least about 30 additional genes, at least about 35 additional genes, at least about 40 additional genes, at least about 45 additional genes, at least about 50 additional genes, at least about 55 additional genes, at least about 60 additional genes, at least about 65 additional genes, at least about 70 additional genes, at least about 75 additional genes, at least about 80 additional genes, at least about 85 additional genes, at least about 90 additional genes, at least about 95 additional genes, at least about 100 additional genes, at least about 110 additional genes, at least about 120 additional genes, at least about 130 additional genes, at least about 140 additional genes, at least about 150 additional genes, at least about 160 additional genes, at least about 170 additional genes, at least about 180 additional genes, at least about 190 additional genes, or at least about 200 additional genes.
  • At least one of the additional genes is selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin-9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA- DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1
  • the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA- 4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) 2 additional genes, 3 additional genes, 4 additional genes, 5 additional genes, 6 additional genes, 7 additional genes, 8 additional genes, 9 additional genes, 10 additional genes, 11 additional genes, 12 additional genes, 13 additional genes, 14 additional genes, 15 additional genes, 16 additional genes, or 17 additional genes.
  • the gene panel consists essentially of (or consists of) (i) CD274 (PD-L1) and LAG3, and (ii) 2 additional genes, 3 additional genes, 4 additional genes, 5 additional genes, 6 additional genes, 7 additional genes, 8 additional genes, 9 additional genes, 10 additional genes, 11 additional genes, 12 additional genes, 13 additional genes, 14 additional genes, 15 additional genes, 16 additional genes, 17 additional genes, 18 additional genes, 19 additional genes, 20 additional genes, 21 additional genes, 22 additional genes, 23 additional genes, 24 additional genes, 25 additional genes, 26 additional genes, 27 additional genes, 28 additional genes, 29 additional genes, 30 additional genes, 31 additional genes, 32 additional genes, 33 additional genes, 34 additional genes, 35 additional genes, 36 additional genes, 37 additional genes, 38 additional genes, 39 additional genes, 40 additional genes, 41 additional genes, 42 additional genes, 43 additional genes, 44 additional genes, 45 additional genes, 46 additional genes, 47 additional genes, 48 additional genes, 49 additional genes, 50 additional genes, 51 additional genes, 52 additional genes, 53 additional genes, 54 additional genes, 55 additional genes, 56
  • 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.
  • the panel of genes comprises at least 14 housekeeping genes. In some aspects, the panel of genes comprises at least 15 housekeeping genes. [0127] 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, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof. In some aspects, 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 70 control genes, at least 75 control genes, at least 80 control genes, at least
  • 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 ANT1, 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 panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1,
  • the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1,
  • the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA- DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG
  • the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PR
  • a parenchymal inflammatory phenotype is characterized by a low parenchymal signature score ("signature score").
  • signature score is a measurement of the composite expression of the genes in the gene panel in a tumor sample form the subject, and comparing the composite expression of the genes in the gene panel to the average composite expression of the panel of genes in tumor samples obtained from a population of subjects afflicted with the tumor.
  • the average composite signature score is determined by measuring the composite expression of the panel of genes in tumor samples obtained from the population of subjects.
  • a low signature score is characterized by a signature score of less than about 50, less than about 45, less than about 40, less than about 35, less than about 30, less than about 25, less than about 20, less than about 15, less than about 10, or less than about 5; wherein a signature score of 100 indicates that the subject has no parenchymal inflammatory phenotype; and wherein a signature score of 1 indicates that a subject has a high parenchymal phenotype.
  • a low signature score is characterized by a signature score of less than about 50.
  • a low signature score is characterized by a signature score of less than about 45.
  • a low signature score is characterized by a signature score of less than about 40.
  • a low signature score is characterized by a signature score of less than about 35. In some aspects, a low signature score is characterized by a signature score of less than about 30. In some aspects, a low signature score is characterized by a signature score of less than about 25. In some aspects, a low signature score is characterized by a signature score of less than about 20. In some aspects, a low signature score is characterized by a signature score of less than about 15. In some aspects, a low signature score is characterized by a signature score of less than about 10. In some aspects, a low signature score is characterized by a signature score of less than about 9. In some aspects, a low signature score is characterized by a signature score of less than about 8.
  • a low signature score is characterized by a signature score of less than about 7. In some aspects, a low signature score is characterized by a signature score of less than about 6. In some aspects, a low signature score is characterized by a signature score of less than about 5. In some aspects, a low signature score is characterized by a signature score of less than about 4. In some aspects, a low signature score is characterized by a signature score of less than about 3. In some aspects, a low signature score is characterized by a signature score of less than about 2. In some aspects, a low signature score is characterized by a signature score of about 1. [0133] 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 the checkpoint inhibitor therapy.
  • 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.
  • the expression of one or more of the genes in the panel of genes is determined by detecting the presence of mRNA transcribed from the gene, the presence of a protein encoded by the gene, or both. [0135] In some aspects, the expression of one or more of the genes is determined by measuring the level of gene mRNA in a sample obtained from the subject. 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 RNA in situ hybridization. [0136] In some aspects, the expression of one or more of the genes is determined by measuring the level of protein expressed from the gene in a sample obtained from the subject.
  • the protein is measured using an immunohistochemistry (IHC) assay.
  • IHC immunohistochemistry
  • the IHC is an automated IHC.
  • the expression of one or more of the genes of the inflammatory gene panel is normalized relative to the expression of one or more housekeeping genes.
  • the one or more housekeeping genes comprises any housekeeping gene disclosed herein or any combination thereof.
  • raw gene expression values are normalized following standard gene expression profiling (GEP) protocols.
  • GEP standard gene expression profiling
  • gene expression signature scores 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.
  • scores have positive or negative values, depending on whether gene expression is up- or down-regulated under a particular condition.
  • a low signature score is characterized by a signature score that is lower than a reference signature score.
  • the reference inflammatory signature score is an average signature score.
  • the average signature score is determined by measuring the expression of the genes present in the panel of genes 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 checkpoint inhibitor, e.g., the anti-PD-1 antibody, the anti-PD-L1 antibody, the anti-CTLA-4 antibody, the anti-LAG-3 antibody, or any combination thereof.
  • the checkpoint inhibitor e.g., the anti-PD-1 antibody, the anti-PD-L1 antibody, the anti-CTLA-4 antibody, the anti-LAG-3 antibody, or any combination thereof.
  • a low signature score is characterized by a signature score that is at less than about 95%, less than about 90%, less than about 85%, less than about 80%, less than about 80%, less than about 75%, less than about 70%, less than about 65%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% that of the average signature score.
  • a low signature score is characterized by a signature score that is at less than about 95% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about % that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 90% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 85% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 80% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 75% that of the average signature score.
  • a low signature score is characterized by a signature score that is at less than about 70% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 65% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 60% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 55% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 50% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 45% that of the average signature score.
  • a low signature score is characterized by a signature score that is at less than about 40% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 35% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 30% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 25% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 20% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 15% that of the average signature score.
  • a low signature score is characterized by a signature score that is at less than about 10% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 9% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 8% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 7% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 6% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 5% that of the average signature score.
  • a low signature score is characterized by a signature score that is at less than about 4% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 3% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 2% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 1% that of the average signature score. II.B.
  • TMB Tumor Mutation Burden
  • TMB refers to the number of somatic mutations in a tumor’s genome and/or the number of somatic mutations per area of the tumor genome (after taking into account germline variant DNA).
  • the acquisition of somatic mutations and, thus, a higher TMB can be influenced by distinct mechanisms, such as exogenous mutagen exposure (e.g., tobacco smoking) and DNA mismatch repair mutations (e.g., MSI in colorectal and esophageal cancers).
  • exogenous mutagen exposure e.g., tobacco smoking
  • DNA mismatch repair mutations e.g., MSI in colorectal and esophageal cancers.
  • MSI in colorectal and esophageal cancers
  • a "nonsynonymous mutation” herein refers to a nucleotide mutation that alters the amino acid sequence of a protein.
  • Missense mutations and nonsense mutations can be both nonsynonymous mutations.
  • a "missense mutation” herein refers to a nonsynonymous point mutation in which a single nucleotide change results in a codon that codes for a different amino acid.
  • a “nonsense mutation” herein refers to a nonsynonymous point mutation in which a codon is changed to a premature stop codon that leads to truncation of the resulting protein.
  • somatic mutations can be expressed at the RNA and/or protein level, resulting in neoantigens (also referred to as neoepitopes). Neoantigens can influence an immune-mediated anti-tumor response.
  • neoantigen recognition can promote T-cell activation, clonal expansion, and differentiation into effector and memory T-cells.
  • early clonal mutations or "trunk mutations”
  • late mutations or “branch mutations”
  • neoantigens derived from clonal "trunk” mutations are more widespread in the tumor genome than "branch” mutations and, thus, can lead to a high number of T cells reactive against the clonal neoantigen.
  • McGranahan et al., (2016) 351:1463-1469. tumors with a high TMB can also have a high neoantigen load, which can lead to high tumor immunogenicity and increased T-cell reactivity and anti-tumor response.
  • cancers with a high TMB can respond well to treatment with immunotherapies, e.g., an anti-PD-1 antibody or anti-PD-L1 antibody.
  • immunotherapies e.g., an anti-PD-1 antibody or anti-PD-L1 antibody.
  • Advances in sequencing technologies allow for evaluation of the tumor’s genomic mutation landscape. Any sequencing methods known to those of skill in the art can be used to sequence nucleic acids from the tumor genome (e.g., obtained from a biological sample from a subject afflicted with a tumor).
  • PCR or qPCR methods, Sanger sequencing methods, or next-generation sequencing (“NGS”) methods can be used to measure TMB.
  • the TMB status is measured using genomic profiling.
  • Genomic profiling involves analyzing nucleic acids from tumor samples, including coding and non-coding regions, and can be performed using methods having integrated optimized nucleic acid selection, read alignment, and mutation calling.
  • gene profiling provides next generation sequencing (NGS)-based analysis of tumors that can be optimized on a cancer-by-cancer, gene-by-gene, and/or site-by-site basis.
  • GGS next generation sequencing
  • Genome profiling can integrate the use of multiple, individually tuned, alignment methods or algorithms to optimize performance in sequencing methods, particularly in methods that rely on massively parallel sequencing of a large number of diverse genetic events in a large number of diverse genes.
  • Genomic profiling provides for a comprehensive analysis of a subject's cancer genome, with clinical grade quality, and the output of the genetic analysis can be contextualized with relevant scientific and medical knowledge to increase the quality and efficiency of cancer therapy.
  • II.B.1. Genomic Profiling involves a panel of a predefined set of genes comprising as few as five genes or as many as 1000 genes, about 25 genes to about 750 genes, about 100 genes to about 800 genes, about 150 genes to about 500 genes, about 200 genes to about 400 genes, about 250 genes to about 350 genes.
  • the genomic profile comprises at least 300 genes, at least 305 genes, at least 310 genes, at least 315 genes, at least 320 genes, at least 325 genes, at least 330 genes, at least 335 genes, at least 340 genes, at least 345 genes, at least 350 genes, at least 355 genes, at least 360 genes, at least 365 genes, at least 370 genes, at least 375 genes, at least 380 genes, at least 385 genes, at least 390 genes, at least 395 genes, or at least 400 genes.
  • the genomic profile comprises at least 325 genes.
  • the genomic profile comprises at least 315 cancer-related genes and introns in 28 genes (FOUNDATIONONE®) or the complete DNA coding sequence of 406 genes, introns in 31 genes with rearrangements, and the RNA sequence (cDNA) of 265 genes (FOUNDATIONONE® Heme).
  • the genomic profile comprises 26 genes and 1000 associated mutations (EXODX® Solid Tumor).
  • the genomic profile comprises 76 genes (Guardant360).
  • the genomic profile comprises 73 genes (Guardant360).
  • the genomic profile comprises 354 genes and introns in 28 genes for rearrangements (FOUNDATIONONE® CDX TM ).
  • the genomic profile is FOUNDATIONONE® F1CDx.
  • the genomic profile comprises 468 genes (MSK-IMPACT TM ).
  • One or more genes can be added to the genome profile as more genes are identified to be related to oncology.
  • II.B.1.a. FOUNDATIONONE® Assay is comprehensive genomic profiling assay for solid tumors, including but not limited to solid tumors of the lung, colon, and breast, melanoma, and ovarian cancer.
  • the FOUNDATIONONE® assay uses a hybrid-capture, next-generation sequencing test to identify genomic alterations (base substitutions, insertions and deletions, copy number alterations, and rearrangements) and select genomic signatures (e.g., TMB and microsatellite instability).
  • the assay covers 322 unique genes, including the entire coding region of 315 cancer-related genes, and selected introns from 28 genes.
  • the full list of FOUNDATIONONE® assay genes is provided in Tables 2 and 3. See FOUNDATIONONE: Technical Specifications, Foundation Medicine, Inc., available at FoundationMedicine.com, last visited March 16, 2018, which is incorporated by reference herein in its entirety.
  • Table 2 List of genes wherein entire coding sequences are assayed in the FOUNDATIONONE® assay.
  • EXODX® Solid Tumor Assay [0148] In one aspect, TMB is measured using the EXODX® Solid Tumor assay.
  • the EXODX® Solid Tumor assay is an exoRNA- and cfDNA-based assay, which detects actionable mutations in cancer pathways.
  • the EXODX® Solid Tumor assay is a plasma-based assay that does not require a tissue sample.
  • the EXODX® Solid Tumor assay covers 26 genes and 1000 mutations. The specific genes covered by the EXODX® Solid Tumor assay are shown in Table 4.
  • TMB status is determined using the Guardant360 assay.
  • the Guardant360 assay measures mutations in at least 73 genes (Table 5), 23 indels (Table 6), 18 CNVs (Table 7), and 6 fusion genes (Table 8). See GuardantHealth.com, last accessed on March 25, 2019.
  • Table 5 Guardant360 assay genes.
  • Table 6 Guardant360 assay indels.
  • Table 7 Guardant360 assay amplifications (CNVs).
  • TMB is determined using the TruSight Tumor 170 assay (ILLUMINA).
  • the TruSight Tumor 170 assay is a next-generation sequencing assay that covers 170 genes associated with common solid tumors, which simultaneously analyzes DNA and RNA.
  • the TruSight Tumor 170 assay assesses fusions, splice variants, insertions/deletions, single nucleotide variants (SNVs), and amplifications.
  • the TruSight Tumor 170 assay gene lists are shown in Tables 12-14.
  • Table 9 TruSight Tumor 170 assay genes (amplifications).
  • Table 10 TruSight Tumor 170 assay genes (fusions).
  • Table 11 TruSight Tumor 170 assay genes (small variants).
  • FOUNDATIONONE® CDX TM (“F1CDx”) is a next generation sequencing based in vitro diagnostic device for detection of substitutions, insertion and deletion alterations (indels), and copy number alterations (CNAs) in 324 genes and select gene rearrangements, as well as genomic signatures including microsatellite instability (MSI) and tumor mutation burden (TMB) using DNA isolated from formalin-fixed paraffin embedded (FFPE) tumor tissue specimens.
  • F1CDx is approved by the United States Food and Drug Administration (FDA) for several tumor indications, including NSCLC, melanoma, breast cancer, colorectal cancer, and ovarian cancer.
  • the F1CDx assay employs a single DNA extraction method from routine FFPE biopsy or surgical resection specimens, 50-1000 ng of which will undergo whole-genome shotgun library construction and hybridization-based capture of all coding exons from 309 cancer-related genes, one promoter region, one non-coding (ncRNA), and selected intronic regions from 34 commonly rearranged genes, 21 of which also include the coding exons.
  • Tables 12 and 13 provide the complete list of genes included in F1CDx. In total, the assay detects alterations in a total of 324 genes.
  • hybrid capture— selected libraries are sequenced to high uniform depth (targeting >500X median coverage with >99% of exons at coverage >100X). Sequence data is then processed using a customized analysis pipeline designed to detect all classes of genomic alterations, including base substitutions, indels, copy number alterations (amplifications and homozygous gene deletions), and selected genomic rearrangements (e.g., gene fusions). Additionally, genomic signatures including microsatellite instability (MSI) and tumor mutation burden (TMB) are reported. Table 12: Genes with full coding exonic regions included in FOUNDATIONONE® CDXTM for the detection of substitutions, insertions and deletions (indels), and copy number alterations (CNAs).
  • MSI microsatellite instability
  • TMB tumor mutation burden
  • the F1CDx assay identifies various alterations in the gene and/or intron sequences, including substitutions, insertions/deletions, and CNAs.
  • the F1CDx assay was previously identifies as having concordance with an externally validated NGS assay and the FOUNDATIONONE® (F1 LDT) assay. See FOUNDATIONONE® CDXTM: Technical Information, Foundation Medicine, Inc., available at FoundationMedicine.com, last visited March 25, 2019, which is incorporated by reference herein in its entirety. II.B.1.f.
  • MSK-IMPACT TM [0154] In some aspects, TMB status is assessed using the MSK-IMPACTTM assay.
  • the MSK-IMPACTTM assay uses next-generation sequencing to analyze the mutation status of 468 genes. Target genes are captured and sequenced on an ILLUMINA HISEQTM instrument. The MSK-IMPACTTM assay is approved by the US FDA for detection of somatic mutations and microsatellite instability in solid malignant neoplasms. The full list of 468 genes analyzed by the MSK-IMPACTTM assay is shown in Table 14. See Evaluation of Automatic Class III Designation for MSK-IMPACT (Integrated Mutation Profiling of Actionable Cancer Targets): Decision Summary, United States Food and Drug Administration, November 15, 2017, available at accessdata.fda.gov.
  • Table 14 Genes analyzed by the MSK-IMPACTTM assay.
  • the TMB is determined using a NEOTYPE Solid Tumor Profile.
  • the NEOGENOMICS assays measure the number of non-synonymous DNA coding sequence changes per megabase of sequenced DNA.
  • II.B.1.h. ONCOMINETM Tumor Mutation Load Assay [0156]
  • TMB is determined using a THERMOFISHER SCIENTIFIC® ONCOMINETM Tumor Mutation assay.
  • TMB is determined using a THERMOFISHER SCIENTIFIC® ION TORRENTTM ONCOMINETM Tumor Mutation assay.
  • the ION TORRENTTM ONCOMINETM Tumor Mutation assay is a targeted NGS assay that quantitates somatic mutations to determine tumor mutation load.
  • the assay covers 1.7 Mb of DNA.
  • the full list of 408 genes analyzed by the THERMOFISHER SCIENTIFIC® ION TORRENTTM ONCOMINETM Tumor Mutation assay is shown in Table 15 (see Iontorrent, Oncomine Tumor Mutation Load Assay Flyer, available at assets.thermofisher.com/TFS- Assets/CSD/Flyers/ oncomine-tumor-mutation-load-assay-flyer.pdf, last visited March 25, 2019).
  • Table 15 Genes analyzed by the THERMOFISHER SCIENTIFIC® ION TORRENTTM ONCOMINETM Tumor Mutation assay.
  • TMB is determined using a NOVOGENETM NOVOPMTM assay. In some aspects, TMB is determined using a NOVOGENETM NOVOPMTM Cancer Panel assay.
  • the NOVOGENETM NOVOPMTM Cancer Panel assay is a comprehensive NGS cancer panel that analyzes the complete coding regions of 548 genes and the introns of 21 genes, representing about 1.5 Mb of DNA, and that are relevant for the diagnosis and/or treatment of solid tumors according to the National Comprehensive Cancer Network (NCCN) guidelines and medical literature. The assay detects SNV, InDel, fusion, and copy number variation (CNV) genomic abnormalities. II.B.1.j.
  • TMB is determined using a TMB assay provided by CARIS® Life Sciences. In some aspects, TMB is determined using the PESONALIS® ACE ImmunoID assay. In some aspects, TMB is determined using the PGDX® CANCERXOMETM-R assay.
  • the genomic profiling detects all mutation types, i.e., single nucleotide variants, insertions/deletions (indels), copy number variations, and rearrangements, e.g., translocations, expression, and epigenetic markers. [0160] Comprehensive gene panels often contain predetermined genes selected based on the type of tumor to be analyzed.
  • the genomic profile used to measure TMB status can be selected based on the type of tumor the subject has.
  • the genomic profile can include a set of genes particular to a solid tumor.
  • the genomic profile can include a set of genes particular to hematologic malignancies and sarcomas.
  • the genomic profile comprises one or more genes selected from the group consisting of ABL1, BRAF, CHEK1, FANCC, GATA3, JAK2, MITF, PDCD1LG2, RBM10, STAT4, ABL2, BRCA1, CHEK2, FANCD2, GATA4, JAK3, MLH1, PDGFRA, RET, STK11, ACVR1B, BRCA2, CIC, FANCE, GATA6, JUN, MPL, PDGFRB, RICTOR, SUFU, AKT1, BRD4, CREBBP, FANCF, GID4 (C17orf39), KAT6A (MYST3), MRE11A, PDK1, RNF43, SYK, AKT2, BRIP1, CRKL, FANCG, GLI1, KDM5A, MSH2, PIK3C2B, ROS1, TAF1, AKT3, BTG1, CRLF2, FANCL, GNA11, KDM5C, MSH6, PIK3CA
  • the TMB analysis further comprises identifying a genomic alteration in one or more of ETV4, TMPRSS2, ETV5, BCR, ETV1, ETV6, and MYB.
  • the genomic profile comprises one or more genes selected from the group consisting of ABL1, 12B, ABL2, ACTB, ACVR1, ACVR1B, AGO2, AKT1, AKT2, AKT3, ALK, ALOX, ALOX12B, AMER1, AMER1 (FAM123B or WTX), AMER1 (FAM123B), ANKRD11, APC, APH1A, AR, ARAF, ARFRP1, ARHGAP26 (GRAF), ARID1A, ARID1B, ARID2, ARID5B, ARv7, ASMTL, ASXL1, ASXL2, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, AXIN2, AXL, B2M, BABAM1, BAP1, BARD1, BBC3, BCL10, B
  • the genomic profiling assay comprises at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, at least about 100, at least about 110, at least about 120, at least about 130, at least about 140, at least about 150, at least about 160, at least about 170, at least about 180, at least about 190, at least about 200, at least about 210, at least about 220, at least about 230, at least about 240, at least about 250, at least about 260, at least about 270, at least about 280, at least about 290, or at least about 300 genes selected from the group consisting of ABL1, 12B, ABL2, ACTB, ACVR1, ACVR1B, AGO2, AKT1, AKT2, AKT3, ALK, ALOX, ALOX12B, AMER1, AMER1 (FAM123B or WTX), AMER1 (FAM123B), ANKRD11, APC,
  • the genomic profile comprises one or more genes selected from the genes listed in Tables 2-15. II.B.2.
  • TMB Status [0165]
  • TMB status based on genomic profiling is highly correlated with TMB status based on whole-exome or whole-genome sequencing.
  • Evidence provided herein shows that the use of genomic profiling assays, such as the F1CDx assay, have concordance with whole-exome and/or whole genome sequencing assays.
  • TMB can be measured using a tissue biopsy sample or, alternatively, circulating tumor DNA (ctDNA), cfDNA (cell-free DNA), and/or a liquid biopsy sample.
  • ctDNA can be used to measure TMB status according to whole-exome or whole-genome sequencing or genomic profiling using available methodologies, e.g., GRAIL, Inc.
  • a subject is identified as suitable for an anti-PD-1 therapy, as disclosed herein, based on the measurement of TMB status and identification of a high TMB.
  • a TMB score is calculated as the total number of nonsynonymous missense mutations in a tumor, as measured by whole exome sequencing or whole genome sequencing.
  • the high TMB has a score of at least 210, at least 215, at least 220, at least 225, at least 230, at least 235, at least 240, at least 245, at least 250, at least 255, at least 260, at least 265, at least 270, at least 275, at least 280, at least 285, at least 290, at least 295, at least 300, at least 305, at least 310, at least 315, at least 320, at least 325, at least 330, at least 335, at least 340, at least 345, at least 350, at least 355, at least 360, at least 365, at least 370, at least 375, at least 380, at least 385, at least 390, at least 395, at least 400, at least 405, at least 410, at least 415, at least 420, at least 425, at least 430, at least 435, at least 440, at least 445, at least 450, at least 455, at least 460
  • the high TMB has a score of at least 215, at least 220, at least 221, at least 222, at least 223, at least 224, at least 225, at least 226, at least 227, at least 228, at least 229, at least 230, at least 231, at least 232, at least 233, at least 234, at least 235, at least 236, at least 237, at least 238, at least 239, at least 240, at least 241, at least 242, at least 243, at least 244, at least 245, at least 246, at least 247, at least 248, at least 249, or at least 250.
  • the high TMB has a score of at least 243.
  • the high TMB has a score of at least 244. In some aspects, the high TMB has a score of at least 245. In other aspects, the high TMB has a score of at least 246. In other aspects, the high TMB has a score of at least 247. In other aspects, the high TMB has a score of at least 248. In other aspects, the high TMB has a score of at least 249. In other aspects, the high TMB has a score of at least 250. In other aspects, the high TMB has a score of any integer between 200 and 300 or higher. In other aspects, the high TMB has a score of any integer between 210 and 290 or higher. In other aspects, the high TMB has a score of any integer between 220 and 280 or higher.
  • the high TMB has a score of any integer between 230 and 270 or higher. In other aspects, the high TMB has a score of any integer between 235 and 265 or higher. [0168] Alternatively, the high TMB can be a relative value rather than an absolute value.
  • the subject’s TMB status is compared to a reference TMB value. In one aspect, the subject’s TMB status is within the highest fractile of the reference TMB value. In another aspect, the subject’s TMB status is within the top tertile of the reference TMB value. [0169] In some aspects, TMB status is expressed as the number of mutations per sample, per cell, per exome, or per length of DNA (e.g., Mb).
  • a tumor has a high TMB status if the tumor has at least about 50 mutations/tumor, at least about 55 mutations/tumor, at least about 60 mutations/tumor, at least about 65 mutations/tumor, at least about 70 mutations/tumor, at least about 75 mutations/tumor, at least about 80 mutations/tumor, at least about 85 mutations/tumor, at least about 90 mutations/tumor, at least about 95 mutations/tumor, at least about 100 mutations/tumor, at least about 105 mutations/tumor, at least about 110 mutations/tumor, at least about 115 mutations/tumor, or at least about 120 mutations/tumor.
  • a tumor has a high TMB status if the tumor has at least about 125 mutations/tumor, at least about 150 mutations/tumor, at least about 175 mutations/tumor, at least about 200 mutations/tumor, at least about 225 mutations/tumor, at least about 250 mutations/tumor, at least about 275 mutations/tumor, at least about 300 mutations/tumor, at least about 350 mutations/tumor, at least about 400 mutations/tumor, or at least about 500 mutations/tumor.
  • a tumor has a high TMB status if the tumor has at least about 100 mutations/tumor.
  • a tumor has a high TMB status if the tumor has at least about 5 mutations per megabase of genes, e.g., genome sequenced according to a TMB assay, e.g., genome sequenced according to a FOUNDATIONONE® CDXTM assay, (mutations/Mb), at least about 6 mutations/Mb, at least about 7 mutations/Mb, at least about 8 mutations/Mb, at least about 9 mutations/Mb, at least about 10 mutations/Mb, at least about 11 mutations/Mb, at least about 12 mutations/Mb, at least about 13 mutations/Mb, at least about 14 mutations/Mb, at least about 15 mutations/Mb, at least about 20 mutations/Mb, at least about 25 mutations/Mb, at least about 30 mutations/Mb, at least about 35 mutations/Mb, at least about 40 mutations/Mb, at least about 45 mutations/Mb, at least about 50 mutations/Mb, at least about 5 mutations per mega
  • a tumor has a high TMB status if the tumor has at least about 5 mutations/Mb. In certain aspects, a tumor has a high TMB status if the tumor has at least about 10 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 11 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 12 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 13 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 14 mutations/Mb.
  • a tumor has a high TMB status if the tumor has at least about 15 mutations/Mb.
  • the values associated with "TMB high” and “TMB low” can differ across tumor types.
  • II.C. PD-L1 Expression Certain aspects of the present disclosure are directed to methods of measuring tumor PD-L1 expression in a sample obtained from a subject.
  • "PD-L1 positive” as used herein can be interchangeably used with "PD-L1 expression of at least about 1%.”
  • the PD-L1 expression can be used by any methods known in the art.
  • the PD-L1 expression is measured by an automated IHC.
  • PD-L1 positive tumors can thus have at least about 1%, at least about 2%, at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, 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 about 100% of the tumor cells expressing PD-L1 as measured by an automated IHC.
  • "PD-L1 positive” means that there are at least 100 cells that express PD-L1 on the surface of the cells.
  • a test tissue sample can be obtained from the patient who is in need of the therapy.
  • the assessment of PD-L1 expression can be achieved without obtaining a test tissue sample.
  • selecting a suitable patient includes (i) optionally providing a test tissue sample obtained from a patient with cancer of the tissue, the test tissue sample comprising tumor cells and/or tumor-infiltrating inflammatory cells; and (ii) assessing the proportion of cells in the test tissue sample that express PD-L1 on the surface of the cells based on an assessment that the proportion of cells in the test tissue sample that express PD-L1 on the cell surface is higher than a predetermined threshold level.
  • the step comprising the provision of a test tissue sample obtained from a patient is an optional step.
  • the "measuring” or “assessing” step to identify, or determine the number or proportion of, cells in the test tissue sample that express PD-L1 is performed by a transformative method of assaying for PD-L1 expression, for example by performing a reverse transcriptase-polymerase chain reaction (RT-PCR) assay or an IHC assay.
  • RT-PCR reverse transcriptase-polymerase chain reaction
  • no transformative step is involved and PD-L1 expression is assessed by, for example, reviewing a report of test results from a laboratory.
  • the steps of the methods up to, and including, assessing PD-L1 expression provides an intermediate result that may be provided to a physician or other healthcare provider for use in selecting a suitable candidate for the anti-PD-1 antibody or anti-PD-L1 antibody therapy.
  • the steps that provide the intermediate result is performed by a medical practitioner or someone acting under the direction of a medical practitioner. In other embodiments, these steps are performed by an independent laboratory or by an independent person such as a laboratory technician.
  • the proportion of cells that express PD-L1 is assessed by performing an assay to determine the presence of PD-L1 RNA.
  • the presence of PD-L1 RNA is determined by RT-PCR, in situ hybridization or RNase protection.
  • the proportion of cells that express PD-L1 is assessed by performing an assay to determine the presence of PD-L1 polypeptide.
  • the presence of PD-L1 polypeptide is determined by immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA), in vivo imaging, or flow cytometry.
  • IHC immunohistochemistry
  • ELISA enzyme-linked immunosorbent assay
  • PD-L1 expression is assayed by IHC.
  • cell surface expression of PD-L1 is assayed using, e.g., IHC or in vivo imaging.
  • Imaging techniques have provided important tools in cancer research and treatment. Recent developments in molecular imaging systems, including positron emission tomography (PET), single-photon emission computed tomography (SPECT), fluorescence reflectance imaging (FRI), fluorescence-mediated tomography (FMT), bioluminescence imaging (BLI), laser-scanning confocal microscopy (LSCM), and multiphoton microscopy (MPM) will likely herald even greater use of these techniques in cancer research.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • FMT fluorescence reflectance imaging
  • FMT fluorescence-mediated tomography
  • BLI laser-scanning confocal microscopy
  • MCM multiphoton microscopy
  • PD-L1 expression is assayed by immunoPET imaging.
  • the proportion of cells in a test tissue sample that express PD-L1 is assessed by performing an assay to determine the presence of PD-L1 polypeptide on the surface of cells in the test tissue sample.
  • the test tissue sample is a FFPE tissue sample.
  • the presence of PD-L1 polypeptide is determined by IHC assay.
  • the IHC assay is performed using an automated process.
  • the IHC assay is performed using an anti-PD- L1 monoclonal antibody to bind to the PD-L1 polypeptide.
  • an automated IHC method is used to assay the expression of PD-L1 on the surface of cells in FFPE tissue specimens.
  • This disclosure provides methods for detecting the presence of human PD-L1 antigen in a test tissue sample, or quantifying the level of human PD-L1 antigen or the proportion of cells in the sample that express the antigen, which methods comprise contacting the test sample, and a negative control sample, with a monoclonal antibody that specifically binds to human PD- L1, under conditions that allow for formation of a complex between the antibody or portion thereof and human PD-L1.
  • the test and control tissue samples are FFPE samples. The formation of a complex is then detected, wherein a difference in complex formation between the test sample and the negative control sample is indicative of the presence of human PD-L1 antigen in the sample.
  • Various methods are used to quantify PD-L1 expression.
  • the automated IHC method comprises: (a) deparaffinizing and rehydrating mounted tissue sections in an autostainer; (b) retrieving antigen using a decloaking chamber and pH 6 buffer, heated to 110°C for 10 min; (c) setting up reagents on an autostainer; and (d) running the autostainer to include steps of neutralizing endogenous peroxidase in the tissue specimen; blocking non-specific protein-binding sites on the slides; incubating the slides with primary antibody; incubating with a postprimary blocking agent; incubating with NovoLink Polymer; adding a chromogen substrate and developing; and counterstaining with hematoxylin.
  • a pathologist examines the number of membrane PD-L1 + + tumor cells in each field under a microscope and mentally estimates the percentage of cells that are positive, then averages them to come to the final percentage.
  • the different staining intensities are defined as 0/negative, l+/weak, 2+/moderate, and 3+/strong. Typically, percentage values are first assigned to the 0 and 3+ buckets, and then the intermediate 1+ and 2+ intensities are considered.
  • the specimen is divided into zones, and each zone is scored separately and then combined into a single set of percentage values.
  • the threshold number of cells that needs to be PD-L1 positive is at least about 100, at least about 125, at least about 150, at least about 175, or at least about 200 cells. In certain embodiments, the threshold number of cells that need to be PD-L1 positive is at least about 100 cells.
  • Staining is also assessed in tumor-infiltrating inflammatory cells such as macrophages and lymphocytes.
  • Macrophages and lymphocytes are assessed for plasma membrane staining and only recorded for all samples as being positive or negative for each cell category. Staining is also characterized according to an outside/inside tumor immune cell designation. "Inside” means the immune cell is within the tumor tissue and/or on the boundaries of the tumor region without being physically intercalated among the tumor cells. "Outside” means that there is no physical association with the tumor, the immune cells being found in the periphery associated with connective or any associated adjacent tissue.
  • the samples are scored by two pathologists operating independently, and the scores are subsequently consolidated.
  • the identification of positive and negative cells is scored using appropriate software.
  • AIS adjusted inflammation score
  • Checkpoint Inhibitors [0185] The present disclosure is directed to methods for treating a human subject afflicted with a cancer comprising administering to the subject a checkpoint inhibitor. Any checkpoint inhibitor known in the art can be used in the methods disclosed herein. In some aspects, the checkpoint inhibitor is any reagent that blocks, inhibits, or reduces the activity of one or more checkpoint protein.
  • the checkpoint protein is selected from the group selected from the group consisting of PD-1, CTLA-4, LAG3, TIGIT, TIM3, NKG2a, OX40, ICOS, CD137, KIR, TGF ⁇ , IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, and any combination thereof.
  • the checkpoint inhibitor blocks inhibits, or reduces the activity of PD-1.
  • the checkpoint inhibitor blocks inhibits, or reduces the activity of CTLA-4.
  • the checkpoint inhibitor blocks inhibits, or reduces the activity of LAG3.
  • the checkpoint inhibitor blocks inhibits, or reduces the activity of TIGIT. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of TIM3. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of NKG2a. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of OX40. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of ICOS. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CD137. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of KIR. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of TGF ⁇ .
  • the checkpoint inhibitor blocks inhibits, or reduces the activity of IL-10. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of IL-8. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of IL-2. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CD96. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of VISTA. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of B7-H4. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of Fas ligand. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CXCR4.
  • the checkpoint inhibitor blocks inhibits, or reduces the activity of mesothelin. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CD27. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of GITR. [0186] Any inhibitor can be used in the methods disclosed herein. In some aspects, the inhibitor is a small molecule. In some aspects, the inhibitor is a protein. In some aspects, the inhibitor is an antibody or an antigen-binding portion thereof. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds PD-1. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CTLA-4.
  • the checkpoint inhibitor is an antibody or antigen- binding portion thereof that specifically binds LAG3. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds TIGIT. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds TIM3. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds NKG2a. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds OX40. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds ICOS.
  • the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CD137. In some aspects, the checkpoint inhibitor is an antibody or antigen- binding portion thereof that specifically binds KIR. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds TGF ⁇ . In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds IL- 10. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds IL-8. In some aspects, the checkpoint inhibitor is an antibody or antigen- binding portion thereof that specifically binds IL-2. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CD96.
  • the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds VISTA. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds B7-H4. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds Fas ligand. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CXCR4. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds mesothelin. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CD27.
  • the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds GITR.
  • the subject is administered a monotherapy, e.g., an anti-PD-1 monotherapy, e.g., wherein the subject is not administered one or more additional anti-cancer agent.
  • the subject is administered a combination therapy, e.g., wherein the subject is administered a first checkpoint inhibitor, e.g., an anti-PD-1 antibody, and one or more additional anti-cancer agents.
  • the subject is administered a combination therapy comprising an anti-PD-1 antibody and an anti-CTLA-4 antibody.
  • the subject is administered a combination therapy comprising an anti-PD-1 antibody and an anti- LAG-3 antibody.
  • an anti-PD-L1 antibody is substituted for the anti-PD-1 antibody.
  • the methods comprise administering an anti-PD-L1 antibody to a subject.
  • the subject is administered an anti-PD-L1 monotherapy.
  • the subject is administered a combination therapy comprising an anti-PD-L1 antibody and a second anti-cancer agent, e.g., an anti-CTLA-4 antibody.
  • the subject is administered a combination therapy comprising an anti-PD-L1 antibody and a second anti-cancer agent, e.g., an anti-LAG-3 antibody.
  • the antibody is a chimeric, humanized or human monoclonal antibody or a portion thereof. II.D.1.
  • Anti-PD-1 Antibodies Useful for the Disclosure [0190] 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.
  • 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. [0191] 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.
  • 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. 10:136 (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. Oncol.
  • 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).
  • 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).
  • the anti-PD-1 antibody binds the same epitope as any of the anti-PD-1 antibodies described herein, e.g., nivolumab.
  • 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 antigen- binding 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 cross- competes 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, is administered at a dose of about 3 mg/kg body weight about once every 2 weeks.
  • the anti-PD-1 antibody e.g., pembrolizumab
  • the anti-PD-1 antibody is administered at a dose of about 2 mg/kg body weight about once every 3 weeks.
  • 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 380 mg, at least about 400 mg, at least about 420 mg, at least about 440 mg, at
  • 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.
  • the anti-PD-1 antibody is administered as a flat dose of about 200 mg at about once every 2 weeks.
  • the anti-PD-1 antibody is administered as a flat dose of about 240 mg at about once every 2 weeks.
  • the anti-PD-1 antibody is administered as a flat dose of about 480 mg at about once every 4 weeks.
  • 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. [0203] 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.
  • pembrolizumab is administered at a flat dose of about 400 mg once about every 4 weeks. II.D.2.
  • Anti-PD-L1 Antibodies Useful for the Disclosure [0204] In certain aspects, an anti-PD-L1 antibody is substituted for the anti-PD-1 antibody in any of the methods disclosed herein.
  • Anti-PD-L1 antibodies that are known in the art can be used in the compositions and methods of the present disclosure. Examples of anti-PD-L1 antibodies useful in the compositions and methods of the present disclosure include the antibodies disclosed in US Patent No. 9,580,507. Anti-PD-L1 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- ⁇ 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-L1 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-L1 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 IgG1 monoclonal anti-PD-L1 antibody.
  • the PD-L1 antibody is durvalumab (IMFINZITM). Durvalumab is a human IgG1 kappa monoclonal anti-PD-L1 antibody.
  • the PD-L1 antibody is avelumab (BAVENCIO®). Avelumab is a human IgG1 lambda monoclonal anti-PD-L1 antibody.
  • Anti-PD-L1 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-L1 antibody disclosed herein, e.g., atezolizumab, durvalumab, and/or avelumab.
  • the anti-PD-L1 antibody binds the same epitope as any of the anti-PD-L1 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 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., 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.
  • Anti-PD-L1 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-L1 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-L1 "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-L1 antibody or antigen-binding portion thereof cross-competes with atezolizumab, durvalumab, and/or avelumab for binding to human PD-L1.
  • the anti-PD-L1 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-L1 antibody is durvalumab.
  • the anti-PD-L1 antibody is avelumab.
  • the anti-PD-L1 antibody is atezolizumab.
  • the anti-PD-L1 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-L1 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-L1 antibody is administered at a dose of about 10 mg/kg body weight at about once every 2 weeks. [0216] In other aspects, the anti-PD-L1 antibody useful for the present disclosure is a flat dose.
  • the anti-PD-L1 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-L1 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-L1 antibody is administered as a flat dose of about 1200 mg at about once every 3 weeks. In other aspects, the anti-PD-L1 antibody is administered as a flat dose of about 800 mg at about once every 2 weeks. In other aspects, the anti-PD-L1 antibody is administered as a flat dose of about 840 mg at about once every 2 weeks. [0217] 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.
  • durvalumab is administered as a flat dose of about 800 mg/kg once about every 2 weeks.
  • durvalumab is administered as a flat dose of about 1200 mg/kg once about every 3 weeks.
  • Anti-CTLA-4 Antibodies [0220] 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.
  • 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.
  • 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 (ka) of at least about 10 3 , about 10 4 , or about 10 5 m -1 s -1 ; (c) a kinetic disassociation constant (kd) 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).
  • K a equilibrium association constant
  • 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.
  • ipilimumab also known as YERVOY®, MDX-010, 10D1; see U.S. Patent No.6,984,720
  • MK- 1308 Merck
  • AGEN-1884 Agenus Inc.
  • tremelimumab tremel
  • 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, IgG1 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.
  • the 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 cross- competing 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 cross- compete 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 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-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 antigen- binding 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 antigen- binding 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 antigen- binding 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 antigen- binding 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
  • 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.
  • 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.D.4.
  • a LAG-3 antagonist includes, but is not limited to, LAG-3 binding agents, e.g., a LAG-3 antibody, and soluble LAG-3 polypeptides, e.g., a fusion protein comprising the extracellular portion of LAG-3.
  • the LAG-3 inhibitor is a soluble LAG-3 polypeptide, for example, a LAG-3-Fc fusion polypeptide capable of binding to MHC Class II.
  • the LAG-3 antagonist comprises IMP321 (eftilagimod alpha).
  • the LAG-3 antagonist is an anti-LAG-3 antibody or an antigen binding fragment thereof that specifically binds to LAG-3 ("anti-LAG-3 antibody").
  • Anti-LAG-3 antibodies (or VH/VL domains derived therefrom) suitable for use herein can be generated using methods well known in the art. Alternatively, art recognized anti- LAG-3 antibodies can be used.
  • the anti-LAG-3 antibody is a chimeric, humanized, or human monoclonal antibody, or a portion thereof. In other aspects, the anti-LAG-3 antibody is a bispecific antibody or a multispecific antibody.
  • the anti-LAG-3 antibody is relatlimab, e.g., BMS-986016 as described in PCT/US13/48999, the teachings of which are hereby incorporated by reference.
  • the antibody has the heavy and light chain CDRs or variable regions of relatlimab. Accordingly, in one aspect, the antibody comprises CDR1, CDR2, and CDR3 domains of the VH region of relatlimab, and CDR1, CDR2 and CDR3 domains of the VL region of relatlimab. In another aspect, the antibody comprises VH and/or VL regions of relatlimab.
  • the anti-LAG-3 antibody cross-competes with relatlimab for binding to human LAG-3.
  • the ability of antibodies to cross-compete for binding to an antigen indicates that the 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., relatlimab, by virtue of their binding to the same epitope region.
  • Cross- competing antibodies can be readily identified based on their ability to cross-compete in standard binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223).
  • the anti-LAG-3 antibody binds to the same epitope as relatlimab.
  • the anti-LAG-3 antibody is a biosimilar of relatlimab.
  • the anti- LAG-3 antibody is LAG-525, MK-4280, REGN3767, TSR-033, TSR-075, Sym022, FS-118, or any combination thereof.
  • art recognized anti-LAG-3 antibodies can be used in the therapeutic methods of the disclosure.
  • the anti-human LAG-3 antibody described in US2011/0150892 A1 which is herein incorporated by reference, and referred to as monoclonal antibody 25F7 (also known as “25F7” and "LAG-3.1) can be used.
  • monoclonal antibody 25F7 also known as “25F7” and "LAG-3.1
  • Other art recognized anti- LAG-3 antibodies that can be used include IMP731 (H5L7BW) described in US 2011/007023, MK-4280 (28G-10) described in WO2016028672, REGN3767 described in Journal for ImmunoTherapy of Cancer, (2016) Vol.4, Supp.
  • anti-LAG-3 antibodies useful in the claimed invention can be found in, for example: US 10,188,730, WO2016/028672, WO2017/106129, WO2017/062888, WO2009/044273, WO2018/069500, WO2016/126858, WO2014/179664, WO2016/200782, WO2015/200119, WO2017/019846, WO2017/198741, WO2017/220555, WO2017/220569, WO2018/071500, WO2017/015560, WO2017/025498, WO2017/087589, WO2017/087901, WO2018/083087, WO2017/149143, WO2017/219995, US2017/0260271, WO2017/086367, WO2017/086419, WO2018/034227, WO18/185046, WO18/185043, WO2018/217940, WO19/011306, WO2018/208868, and WO
  • an anti-LAG-3 antibody cross-competes with, binds to the same epitope as, or is a biosimilar of an anti-LAG-3 antibody that is described herein or that is known in the art.
  • an anti-LAG-3 antibody is used to determine LAG-3 expression.
  • an anti-LAG-3 antibody is selected for its ability to bind to LAG-3 in formalin-fixed, paraffin-embedded (FFPE) tissue specimens.
  • an anti-LAG-3 antibody is capable of binding to LAG-3 in frozen tissues. In further aspects, an anti-LAG-3 antibody is capable of distinguishing membrane bound, cytoplasmic, and/or soluble forms of LAG-3. [0249] In some aspects, an anti-LAG-3 antibody useful for assaying, detecting, and/or quantifying LAG-3 expression in accordance with the methods described herein is the 17B4 mouse IgG1 anti-human LAG-3 monoclonal antibody, or an antigen binding fragment thereof. See, e.g., J. Matsuzaki, et al.; PNAS 107, 7875 (2010).
  • the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a dose ranging from about 0.1 mg/kg to about 10.0 mg/kg body weight once about every 1, 2, 3, 4, 5, 6, 7, or 8 weeks. In some aspects, the anti-LAG-3 antibody or antigen- binding portion thereof is administered at a dose of at least about 1 mg/kg, at least about 2 mg/kg, at least about 3 mg/kg, at least about 4 mg/kg, at least about 5 mg/kg, at least about 6 mg/kg, at least about 7 mg/kg, at least about 8 mg/kg, at least about 9 mg/kg, or at least about 10 mg/kg body weight about once every 1, 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose.
  • the anti-LAG-3 antibody is administered at a flat dose of from about 20 mg to about 2000 mg.
  • the anti-LAG-3 antibody or antigen- binding portion thereof is administered as a flat dose of at least about 80 mg or at least about 160 mg.
  • the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose once about every 1, 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose.
  • the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose of about 80 mg.
  • the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose of about 160 mg. II.D.5.
  • Combination Therapies [0255]
  • the anti-PD-1 antibody, the anti-PD-L1 antibody, the anti-LAG- 3 antibody, and/or the anti-CTLA-4 antibody are administered at a therapeutically effective amount.
  • the method comprises administering a therapeutically effective amount of anti-PD-1 antibody and an anti-CTLA-4 antibody.
  • the method comprises administering a therapeutically effective amount of anti-PD-1 antibody and an anti-LAG-3 antibody.
  • the method comprises administering a therapeutically effective amount of anti-PD-L1 antibody and an anti-CTLA-4 antibody. In other aspects, the method comprises administering a therapeutically effective amount of anti-PD-L1 antibody and an anti- LAG-3 antibody. Any anti-PD-1, anti-PD-L1, anti-LAG-3 antibody, or anti-CTLA-4 antibody disclosed herein can be used in the method.
  • the anti-PD-1 antibody comprises nivolumab.
  • the anti-PD-1 antibody comprises pembrolizumab.
  • the anti-PD-L1 antibody comprises atezolizumab.
  • the anti-PD-L1 antibody comprises durvalumab.
  • the anti-PD-L1 antibody comprises avelumab. In some aspects, the anti-CTLA-4 antibody comprises ipilimumab. In some aspects, the anti-CTLA-4 antibody comprises ipilimumab tremelimumab. [0256] In some aspects, the (a) anti-PD-1 antibody or the anti-PD-L1 antibody and the (b) anti-CTLA-4 antibody are each administered once about every 2 weeks, once about every 3 weeks, once about every 4 weeks, once about every 5 weeks, or once about every 6 weeks.
  • the anti-PD-1 antibody or the anti-PD-L1 antibody is administered once about every 2 weeks, once about every 3 weeks or once about every 4 weeks, and the anti-CTLA-4 antibody is administered once about every 6 weeks. In some aspects, the anti-PD-1 antibody or anti-PD-L1 antibody is administered on the same day as the anti-CTLA-4 antibody. In some aspects, the anti-PD-1 antibody or the anti-PD-L1 antibody is administered on a different day than the anti-CTLA-4 antibody. [0257] In some aspects, the anti-CTLA-4 antibody is administered at a dose ranging from about 0.1 mg/kg to about 20.0 mg/kg body weight once about every 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the anti-CTLA-4 antibody is administered at a dose of about 0.1 mg/kg, about 0.3 mg/kg, about 0.6 mg/kg, about 0.9 mg/kg, about 1 mg/kg, about 3 mg/kg, about 6 mg/kg, about 9 mg/kg, about 10 mg/kg, about 12 mg/kg, about 15 mg/kg, about 18 mg/kg, or about 20 mg/kg.
  • the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 4 weeks.
  • the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks.
  • the anti-CTLA-4 antibody is administered at a flat dose.
  • the anti-CTLA-4 antibody is administered at a flat dose ranging from at least about 40 mg to at least about 1600 mg. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose of at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least about 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg, at least about 180 mg, at least about 190 mg, or at least about 200 mg.
  • the CTLA-4 antibody is administered at a flat dose of at least about 220 mg, at least about 230 mg, at least about 240 mg, at least about 250 mg, at least about 260 mg, at least about 270 mg, at least about 280 mg, at least about 290 mg, at least about 300 mg, at least about 320 mg, at least about 360 mg, at least about 400 mg, at least about 440 mg, at least about 480 mg, at least about 520 mg, at least about 560 mg, or at least about 600 mg.
  • the CTLA-4 antibody is administered at a flat dose of at least about 640 mg, at least about 720 mg, at least about 800 mg, at least about 880 mg, at least about 960 mg, at least about 1040 mg, at least about 1120 mg, at least about 1200 mg, at least about 1280 mg, at least about 1360 mg, at least about 1440 mg, or at least about 1600 mg.
  • the anti-CTLA-4 antibody is administered in a flat dose at least once about every 2, 3, 4, 5, 6, 7, or 8 weeks.
  • the anti-PD-1 antibody is administered at a dose of about 2 mg/kg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks.
  • the anti-PD-1 antibody is administered at a dose of about 3 mg/kg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti- PD-1 antibody is administered at a dose of about 6 mg/kg once about every 4 weeks and the anti- CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0260] In certain aspects, the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks.
  • the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 240 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 480 mg once about every 4 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks.
  • the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a flat dose of about 80 mg once about every 6 weeks.
  • the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks.
  • the anti-PD-1 antibody is administered at a flat dose of about 240 mg once about every 2 weeks and the anti- CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks.
  • the anti-PD-1 antibody is administered at a flat dose of about 480 mg once about every 4 weeks and the anti-CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks.
  • the anti-PD-L1 antibody is administered at a dose of about 10 mg/kg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks.
  • the anti-PD-L1 antibody is administered at a dose of about 15 mg/kg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks.
  • the anti-PD-L1 antibody is administered at a flat dose of about 800 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-L1 antibody is administered at a flat dose of about 1200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0264] In certain aspects, the anti-PD-L1 antibody is administered at a flat dose of about 800 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a flat dose of about 80 mg once about every 6 weeks.
  • the anti-PD-L1 antibody is administered at a flat dose of about 1200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks.
  • the anti-PD-1 antibody e.g., nivolumab
  • the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg on the same day, once about every 3 weeks for 4 doses, then the anti-PD-1 antibody, e.g., nivolumab, is administered at a flat dose of 240 mg once about every 2 weeks or 480 mg once about every 4 weeks.
  • the anti-PD-1 antibody e.g., nivolumab
  • the anti-CTLA-4 antibody is administered at a dose of about 3 mg/kg on the same day, once about every 3 weeks for 4 doses, then the anti-PD-1 antibody, e.g., nivolumab, is administered at a flat dose of 240 mg once about every 2 weeks or 480 mg once about every 4 weeks.
  • the methods disclosed herein further comprise administering an anti-PD-1 antibody (or an anti-PD-L1 antibody) and an additional anticancer therapy.
  • the method comprising administering an anti-PD-1 antibody (or an anti-PD-L1 antibody), an anti-CTLA-4 antibody, and an additional anticancer therapy
  • the additional anticancer 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 additional anticancer therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof.
  • the additional anticancer therapy comprises a chemotherapy, including any chemotherapy disclosed herein.
  • the additional anticancer therapy comprises an immunotherapy.
  • the additional anticancer therapy comprises administration of an antibody or antigen-binding portion thereof that specifically binds TIGIT, TIM3, NKG2a, OX40, ICOS, MICA, CD137, KIR, TGF ⁇ , 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, kidney cancer, head and neck cancer, colon cancer, and any combination thereof.
  • 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 hepatocellular cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 gastroesophageal cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined.
  • 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 melanoma, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 bladder cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined.
  • 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 lung cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 kidney cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. In certain aspects, 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 head and neck cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined.
  • the tumor is derived from a colon 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, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined.
  • the subject has received one, two, three, four, five or more prior cancer treatments.
  • the subject is treatment-na ⁇ ve.
  • the subject has progressed on other cancer treatments.
  • 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. [0269] 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.
  • 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-L1 antibody therapy.
  • the previous line of therapy comprises a chemotherapy. In some aspects, the chemotherapy comprises a platinum-based therapy.
  • the platinum- based 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 anticancer 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 subject has experienced disease progression after the 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 co- formulation of an antibody with recombinant human hyaluronidase enzyme (rHuPH20), which removes traditional limitations on the volume of biologics 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, anti- oxidant, 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.
  • the method comprises administering an anti-PD-1 antibody (or an anti-PD-L1 antibody) and an anti-CTLA-4 antibody, wherein the anti-PD-1 antibody (or the anti- PD-L1 antibody) is administered in a fixed dose with the anti-CTLA-4 antibody in a single composition.
  • the anti-PD-1 antibody is administered in a fixed dose with the anti-CTLA-4 antibody.
  • the anti-PD-L1 antibody is administered in a fixed dose with the anti-CTLA-4 antibody in a single composition.
  • the ratio of the anti-PD- 1 antibody (or the anti-PD-L1 antibody) to the anti-CTLA-4 antibody is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:15, about 1:20, about 1:30, about 1:40, about 1:50, about 1:60, about 1:70, about 1:80, about 1:90, about 1:100, about 1:120, about 1:140, about 1:160, about 1:180, about 1:200, about 200:1, about 180:1, about 160:1, about 140:1, about 120:1, about 100:1, about 90:1, about 80:1, about 70:1, about 60:1, about 50:1, about 40:1, about 30:1, about 20:1, about 15:1, about 10:1, about 9:1, about 8:1, about 7:1, about 6:1, about 5:1, about 4:1, about 3:1, or about 2:1.
  • the dosages of the anti- PD-1 antibody, the anti-PD-L1 antibody, and/or the anti-CTLA-4 antibody administered are significantly lower than the approved dosage, i.e., a subtherapeutic dosage, of the agent.
  • the anti- PD-1 antibody, the anti-PD-L1 antibody, and/or the anti-CTLA-4 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.
  • kits comprising (a) one or more checkpoint inhibitors described elsewhere herein, e.g., an anti-PD-1 antibody or an anti-PD-L1 antibody for therapeutic uses, wherein the kits are suitable for the subjects identified as described herein.
  • 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 therapeutically effective dosage of a checkpoint inhibitor, e.g., 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-L1 antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-L1 antibody in the methods disclosed herein.
  • a checkpoint inhibitor e.g., 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-L1 antibody
  • kits for treating a subject identified as a suitable candidate for a checkpoint inhibitor therapy as described herein comprising: (a) a therapeutically effective dosage of a checkpoint inhibitor, e.g., 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-L1 antibody; and (b) instructions for using the checkpoint inhibitor, e.g., the anti- PD-1 antibody or the anti-PD-L1 antibody, in the methods disclosed herein.
  • a therapeutically effective dosage of a checkpoint inhibitor e.g., 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-L1 antibody
  • instructions for using the checkpoint inhibitor e.g., the anti- PD-1 antibody or the anti-PD-L1 antibody
  • this disclosure provides a kit for treating a subject identified as a suitable candidate for a checkpoint inhibitor therapy as described herein, the kit comprising: (a) a therapeutically effective dosage of a checkpoint inhibitor, e.g., 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-L1 antibody; and (b) instructions for using the checkpoint inhibitor, e.g., the anti-PD-1 antibody or the anti-PD-L1 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. [0281] 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. [0282] In some aspects, the kit further includes an inflammatory gene panel assay disclosed herein.
  • the kit further includes instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having a high inflammatory signature score, according to the methods disclosed herein.
  • the kit further includes an anti-CTLA-4 antibody and instructions to administer (a) the anti-PD-1 antibody or the anti-PD-L1 antibody and (b) the anti-CTLA-4 antibody to a subject identified as having a high inflammatory signature score, according to the methods disclosed herein.
  • the kit further includes a comprehensive genomic profiling assay disclosed herein.
  • the kit includes a FOUNDATIONONE® CDXTM genomic profiling assay.
  • the kit further includes instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein.
  • the kit further includes an anti-CTLA-4 antibody and instructions to administer (a) the anti-PD-1 antibody or the anti-PD-L1 antibody and (b) the anti- CTLA-4 antibody to a subject identified as having a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a comprehensive genomic profiling assay disclosed herein, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a high inflammatory signature score and (b) a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an anti-CTLA-4 antibody, (c) an inflammatory gene panel assay disclosed herein, (d) a comprehensive genomic profiling assay disclosed herein, and (e) instructions to administer (a) the anti-PD-1 antibody or the anti-PD-L1 antibody and (b) the anti-CTLA-4 antibody to a subject identified as having (a) a high inflammatory signature score and (b) a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein.
  • the kit further comprises an anti-PD-L1 antibody.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) positive tumor PD-L1 expression, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 1%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD- L1 expression of at least about 5%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 10%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 15%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 20%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti- PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD- L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 25%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 30%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 35%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 40%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti- PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD- L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 45%, as disclosed herein.
  • the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 50%, as disclosed herein. IV.
  • Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, (b) a PD-L1 expression pattern in a biological sample, and (c) a tumor mutation burden (TMB) status of a biological sample.
  • Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, and (b) a PD-L1 expression pattern in a biological sample.
  • Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, and (b) a tumor mutation burden (TMB) status of a biological sample.
  • the device further comprises a means for detecting a level of expression of the transcript.
  • the means for detecting a level of expression of a panel of genes in the biological sample comprises a first biological probe, wherein the first biological probe specifically interacts with a transcript of at least one gene in the panel of genes.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe, a third biological probe, a fourth biological probe, a fifth biological probe, a sixth biological probe, a seventh biological probe, an eight biological probe, a ninth biological probe, or a tenth biological probe.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises a third biological probe.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises a fourth biological probe.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises a fifth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a sixth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a seventh biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises an eight biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a ninth biological probe.
  • the means for detecting a level of expression of a panel of genes in the biological sample further comprises or a tenth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample comprises more than ten biological probes.
  • the first biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a first gene of the at least 5 genes.
  • the second biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a second gene of the at least 5 genes.
  • the third biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a third gene of the at least 5 genes.
  • the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes. In some aspects, the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes. In some aspects, the fifth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fifth gene of the at least 5 genes.
  • the transcript comprises an mRNA. In some aspects, the transcript comprises a cDNA. [0290] All of the references cited above, as well as all references cited herein, are incorporated herein by reference in their entireties.
  • TME tumor microenvironment
  • I-O immune oncology
  • PD-L1 Programmed death ligand 1
  • IHC immunohistochemistry
  • RNA transcripts by gene expression profiling (GEP)
  • GEP gene expression profiling
  • a parenchymal inflammatory signature score (parenchymal signature) that correlates with CD8 expression by IHC can accurately assess inflammation in the TME.
  • the parenchymal signature has been analytically validated using an investigational use only (IUO) GEP-based inflammation panel across multiple tumor types, including urothelial carcinoma (UC).
  • TMB Tumor mutational burden
  • parenchymal signature scores did not correlate with other potential biomarkers of response (PD-L1 expression and TMB), and associations of the parenchymal signature with survival were independent of PD- L1 expression and TMB. [0308] These data suggest that the parenchymal signature can be used alone and/or in combination with other relevant biomarkers such as PD-L1 expression or TMB to identify patients more likely to benefit from I-O therapies.

Abstract

The disclosure provides a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.

Description

METHODS OF IDENTIFYING A SUBJECT WITH A TUMOR SUITABLE FOR A CHECKPOINT INHIBITOR THERAPY
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This PCT application claims the priority benefit of U.S. Provisional Application No. 62/931,744, filed November 6, 2019, 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 a checkpoint inhibitor, e.g, 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] Until recently, cancer immunotherapy had focused substantial effort on approaches that enhance anti-tumor immune responses by adoptive-transfer of activated effector cells, immunization against relevant antigens, or providing non-specific immune-stimulatory agents such as cytokines. In the past decade, however, intensive efforts to develop specific immune checkpoint pathway inhibitors have begun to provide new immunotherapeutic approaches for treating cancer, including the development of antibodies such as nivolumab and pembrolizumab (formerly lambrolizumab; US AN Council Statement, 2013) that bind specifically to the Programmed Death -1 (PD-1) receptor and block the inhibitory PD-l/PD-1 ligand pathway (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 are complex, however. 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] Certain aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined. [0007] Certain aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising: (a) (a) determining whether the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD- L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined; and (b) administering an effective amount of a checkpoint inhibitor to the subject. [0008] Certain aspects of the present disclosure are directed to a method of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy, comprising any two or more of the following: (i) determining whether the subject has a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) detecting a level of PD-L1 expression in tumor cells of the tumor; and (iii) measuring a TMB status of a biological sample obtained from the subject. [0009] In some aspects, the method further comprises administering to the subject a checkpoint inhibitor if the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression, and (iii) the subject exhibits a TMB status of at least about 10 mutations per megabase of genes examined. [0010] Certain aspects of the present disclosure are directed to a checkpoint inhibitor for treating a tumor in a human subject in need thereof, wherein the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined. [0011] Certain aspects of the present disclosure are directed to a combination of assays for identifying a tumor in a human subject in need thereof, wherein the combination of assays comprises at least two of the following: (i) the subject is to be determined for having a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject [0012] (ii) a level of PD-L1 expression in tumor cells of the tumor is to be detected; and (iii) the subject is to be measured for a TMB status of a biological sample obtained from the subject. [0013] In some aspects, the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA-4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA- DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3. [0014] In some aspects, the panel of genes comprises at least about 5 additional genes, at least about 10 additional genes, at least about 15 additional genes, at least about 20 additional genes, at least about 25 additional genes, at least about 30 additional genes, at least about 35 additional genes, at least about 40 additional genes, at least about 45 additional genes, at least about 50 additional genes, at least about 55 additional genes, at least about 60 additional genes, at least about 65 additional genes, at least about 70 additional genes, at least about 75 additional genes, at least about 80 additional genes, at least about 85 additional genes, at least about 90 additional genes, at least about 95 additional genes, at least about 100 additional genes, at least about 110 additional genes, at least about 120 additional genes, at least about 130 additional genes, at least about 140 additional genes, at least about 150 additional genes, at least about 160 additional genes, at least about 170 additional genes, at least about 180 additional genes, at least about 190 additional genes, or at least about 200 additional genes. In some aspects, at least one of the additional genes is selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof. In some aspects, the panel of genes comprises at least about 63 genes. [0015] In some aspects, the panel of genes comprises at least one housekeeping gene. In some aspects, the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 housekeeping genes. In some aspects, the housekeeping genes are selected from the group consisting of ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof. [0016] In some aspects, the panel of genes comprises at least one control gene. In some aspects, the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 control genes. In some aspects, the control genes are selected from the group consisting of ANT1, 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. [0017] In some aspects, the panel of genes comprises at least about 95 genes. In some aspects, the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3. [0018] In some aspects, the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0019] In some aspects, the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR- Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0020] In some aspects, the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0021] In some aspects, a parenchymal inflammatory phenotype is characterized by a low signature score, wherein the signature score is determined by measuring the composite expression of the genes in the gene panel in a tumor sample form the subject, and comparing the composite expression of the genes in the gene panel to the average composite expression of the panel of genes in tumor samples obtained from a population of subjects afflicted with the tumor. In some aspects, the average composite signature score is determined by measuring the composite expression of the panel of genes in tumor samples obtained from the population of subjects. In some aspects, the low signature score is characterized by a signature score of less than about 50, less than about 45, less than about 40, less than about 35, less than about 30, less than about 25, less than about 20, less than about 15, less than about 10, or less than about 5; wherein a signature score of 100 indicates that the subject has no parenchymal inflammatory phenotype; and wherein a signature score of 1 indicates that a subject has a high parenchymal phenotype. [0022] In some aspects, the TMB status is determined by sequencing nucleic acids in the tumor and identifying a genomic alteration in the sequenced nucleic acids. In some aspects, alteration comprises one or more somatic mutations. In some aspects, the genomic alteration comprises one or more nonsynonymous mutations. In some aspects, the genomic alteration comprises one or more missense mutations. In some aspects, the genomic alteration comprises one or more alterations selected from the group consisting of a base pair substitution, a base pair insertion, a base pair deletion, a copy number alteration (CNAs), a gene rearrangement, and any combination thereof. [0023] In some aspects, the TMB status of the tumor comprises at least 10 mutations, at least about 11 mutations, at least about 12 mutations, at least about 13 mutations, at least about 14 mutations, at least about 15 mutations, at least about 16 mutations, at least about 17 mutations, at least about 18 mutations, at least about 19 mutations, at least about 20 mutations, at least about 21 mutations, at least about 22 mutations, at least about 23 mutations, at least about 24 mutations, at least about 25 mutations, at least about 26 mutations, at least about 27 mutations, at least about 28 mutations, at least about 29 mutations, or at least about 30 mutations per megabase of genome examined as measured by a FOUNDATIONONE® CDX™ assay. [0024] In some aspects, PD-L1 expression is determined using an IHC assay. In some aspects, the IHC assay is an automated IHC assay. In some aspects, the tumor sample is a tumor tissue biopsy. In some aspects, the tumor sample is a formalin-fixed, paraffin-embedded tumor tissue or a fresh-frozen tumor tissue. [0025] In some aspects, the expression of the genes in the panel of genes is determined by measuring the level of mRNA, the level of a protein encoded by the gene, or both. In some aspects, the level of mRNA is determined using reverse transcriptase PCR. In some aspects, the level of the protein encoded by the gene is determined using an IHC assay. In some aspects, the IHC assay is an automated IHC assay. [0026] In some aspects, the checkpoint inhibitor comprises a polypeptide, a small molecule, a polynucleotide, or any combination thereof. In some aspects, the checkpoint inhibitor comprises an antibody or an antigen-binding fragment thereof. In some aspects, the antibody or antigen-binding fragment thereof specifically binds a target selected from PD-1 ("anti-PD-1 antibody"), PD-L1 ("anti-PD-L1 antibody"), CTLA-4, LAG3, TIGIT, TIM3, NKG2a, OX40, ICOS, CD137, KIR, TGFβ, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, and any combination thereof. In some aspects, the antibody is a chimeric, humanized or human monoclonal antibody or a portion thereof. [0027] In some aspects, the anti-PD-1 antibody cross-competes with nivolumab for binding to human PD-1. In some aspects, the anti-PD-1 antibody binds to the same epitope as nivolumab. In some aspects, the anti-PD-1 antibody comprises a heavy chain constant region which is of a human IgG1 or IgG4 isotype. In some aspects, the anti-PD-1 antibody is nivolumab. In some aspects, the anti-PD-1 antibody is pembrolizumab. [0028] In some aspects, the anti-PD-1 antibody is administered at a dose ranging from at least about 0.1 mg/kg to at least about 10.0 mg/kg body weight once about every 1, 2 or 3 weeks. In some aspects, the anti-PD-1 antibody is administered at a dose of at least about 3 mg/kg body weight once about every 2 weeks. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose. In some aspects, the anti-PD-1 antibody or antigen- binding portion thereof is administered at a flat dose of at least about 200, at least about 220, at least about 240, at least about 260, at least about 280, at least about 300, at least about 320, at least about 340, at least about 360, at least about 380, at least about 400, at least about 420, at least about 440, at least about 460, at least about 480, at least about 500 or at least about 550 mg. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 240 mg. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 480 mg. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose about once every 1, 2, 3 or 4 weeks. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose or about 240 mg once about every two weeks. In some aspects, the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 480 mg once about every four weeks. In some aspects, the anti-PD-1 antibody is administered for as long as clinical benefit is observed or until unmanageable toxicity or disease progression occurs. [0029] In some aspects, the antibody is formulated for intravenous administration. In some aspects, the antibody is administered at a subtherapeutic dose. [0030] In some aspects, the methods disclosed herein further comprise administering an antibody or an antigen binding fragment thereof that binds specifically to CTLA-4 ("an anti- CTLA-4 antibody"). In some aspects, the anti-CTLA-4 antibody cross-competes with ipilimumab or tremelimumab for binding to human CTLA-4. In some aspects, the anti-CTLA-4 antibody binds to the same epitope as ipilimumab or tremelimumab. In some aspects, the anti-CTLA-4 antibody is ipilimumab. In some aspects, the anti-CTLA-4 antibody is tremelimumab. In some aspects, the anti-CTLA-4 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. In some aspects, the anti-CTLA-4 antibody is administered at a dose of 1 mg/kg body weight once every 6 weeks. In some aspects, the anti-CTLA-4 antibody is administered at a dose of 1 mg/kg body weight once every 4 weeks. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose of at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least about 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg,at least about 180 mg, at least about 190 mg, or at least about 200 mg. In some aspects, the anti-CTLA-4 antibody is administered as a flat dose about once every 2, 3, 4, 5, 6, 7, or 8 weeks. [0031] 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, kidney cancer, head and neck cancer, colon cancer, and any combination thereof. In some aspects, the tumor is derived from a melanoma. In some aspects, the tumor is derived from a small cell carcinoma of the head and neck. In some aspects, the tumor is relapsed. In some aspects, the tumor is refractory. [0032] In some aspects, the tumor is refractory following at least one prior therapy comprising administration of at least one anticancer agent. In some aspects, the at least one anticancer agent comprises a standard of care therapy. In some aspects, the at least one anticancer agent comprises an immunotherapy. In some aspects, the tumor is locally advanced. In some aspects, the tumor is metastatic. In some aspects, the administering treats the tumor. [0033] In some aspects, the administering reduces the size of the tumor. In some aspects, the size of the tumor is reduced by at least about 10%, about 20%, about 30%, about 40%, or about 50% compared to the tumor size prior to the administration. In some aspects, the subject exhibits progression-free survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about eighteen months, at least about two years, at least about three years, at least about four years, or at least about five years after the initial administration. In some aspects, the subject exhibits stable disease after the administration. In some aspects, the subject exhibits a partial response after the administration. In some aspects, the subject exhibits a complete response after the administration. [0034] Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, (b) a PD-L1 expression pattern in a biological sample, and (c) a tumor mutation burden (TMB) status of a biological sample. In some aspects, the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA-4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample comprises a first biological probe, wherein the first biological probe specifically interacts with a transcript of at least one gene in the panel of genes. In some aspects, the device further comprises a means for detecting a level of expression of the transcript. In some aspects, the transcript comprises an mRNA or a cDNA. [0035] In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe, a third biological probe, a fourth biological probe, and a fifth biological probe. In some aspects, the first biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a first gene of the at least 5 genes; wherein the second biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a second gene of the at least 5 genes; wherein the third biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a third gene of the at least 5 genes; wherein the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes; and wherein the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes. In some aspects, the transcript of the first gene, the transcript of the second gene, the transcript of the third gene, and/or the transcript of the fourth gene comprises an mRNA or a cDNA. [0036] In some aspects, the biological sample comprises a tumor sample from a human subject. In some aspects, the tumor sample is a tumor tissue biopsy. In some aspects, the tumor sample is a formalin-fixed, paraffin-embedded tumor tissue or a fresh-frozen tumor tissue. [0037] In some aspects, the panel of genes comprises 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 110, at least about 120, at least about 130, at least about 140, at least about 150, at least about 160, at least about 170, at least about 180, at least about 190, or at least about 110 additional genes. In some aspects, at least one of the additional genes are selected from CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof. In some aspects, the panel of genes comprises at least about 63 genes. [0038] In some aspects, the panel of genes comprises at least one housekeeping gene. In some aspects, the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 housekeeping genes. In some aspects, the housekeeping genes are selected from the group consisting of ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof. [0039] In some aspects, the panel of genes comprises at least one control gene. In some aspects, the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 control genes. In some aspects, the control genes are selected from the group consisting of ANT1, 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. [0040] In some aspects, the panel of genes comprises at least about 95 genes. In some aspects, the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3. [0041] In some aspects, the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0042] In some aspects, the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR- Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0043] In some aspects, the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0044] In some aspects, the means for detecting a PD-L1 expression pattern in a biological sample comprises means for conducting an IHC assay. In some aspects, the IHC assay is an automated IHC assay. [0045] In some aspects, the means for detecting a tumor mutation burden (TMB) status of a biological sample comprises an automated DNA sequencer. In some aspects, the automated DNA sequencer is capable of whole exome sequencing (WES). [0046] Certain aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject is identified as having a tumor suitable for a checkpoint inhibitor therapy, wherein the subject is identified as suitable by obtaining a biological sample from the subject and applying the biological sample to a device disclosed herein. Certain aspects of the present disclosure are directed to a method of identifying a subject suitable for an immunotherapy, comprising obtaining a biological sample from the subject and applying the biological sample to the device a device disclosed herein. In some aspects, the method further comprises administering an immunotherapy to the subject. BRIEF DESCRIPTION OF THE DRAWINGS [0047] FIG. 1 is a graphical representation showing the distribution of parenchymal inflammatory signature scores ("parenchymal score") relative to objective response in patients. SD = stable disease; PD = progressive disease; NE = not evaluated; CR = complete response; PR = partial response (P<0.01). [0048] FIGs.2A-2B are graphical representations of progression free survival (PFS; FIG. 2A) and overall survival (FIG.2B) in patients stratified by parenchymal signature score. A = low parenchymal score; B = medium parenchymal score; C = high parenchymal score. The number of patients at risk in each group is shown below the x-axis. (P < 0.01). [0049] FIGs. 3A-3B are scatter plots comparing parenchymal score to PD-L1 expression in tumor cells (FIG.3A), and TMB score (FIG.3B). [0050] FIGs. 4A-4B are graphs that show the association of the parenchymal signature scores (log hazard ration) with progression-free survival (PFS; FIG. 4A) and overall survival (OS; FIGs.4B) at PD-L1 expression levels of 0% (FIGs.4A-4B), at least 1% (FIGs.4C-4D), and at least 5% as indicated. [0051] FIGs.4C and 4D are graphical representations of overall survival (OS) in subjects identified as having < 1% tumor PD-L1 expression (FIG. 4C) or ≥ 1% PD-L1 tumor expression (FIG.4D) stratified by parenchymal signature score (A = low; B = medium; C = high). [0052] FIGs. 5A-5B are graphs that show the association of the parenchymal signature scores (log hazard ration) with progression-free survival (PFS; FIG. 5A) and overall survival (OS; FIGs. 5B) based on TMB scores, as indicated. FIG. 5C is a chart showing the cutoffs for TMB used for the statistical analysis for FIGs. 5A-5B. FIGs. 5D and 5E are graphical representations of overall survival (OS) in subjects identified as having a TMB score that is less than the median TMB score (FIG.4D) or a TMB score that is greater than or equal to the median TMB score (FIG. 4E) stratified by parenchymal signature score (A = low; B = medium; C = high). DETAILED DESCRIPTION OF THE DISCLOSURE [0053] The present disclosure provides a method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined. [0054] Some aspects of the present disclosure are directed to a method of treating a tumor in a human subject in need thereof, comprising (a) determining whether the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a TMB status of at least about 10 mutations per megabase of genes examined; and administering an effective amount of a checkpoint inhibitor to the subject. [0055] Some aspects of the present disclosure are directed to a method of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy, comprising any two or more of the following: determining whether the subject has a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; detecting a level of PD-L1 expression in tumor cells of the tumor; and measuring a TMB status of a biological sample obtained from the subject. I. Terms [0056] 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. [0057] 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. [0058] 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. [0059] Units, prefixes, and symbols are denoted in their Système International de Unites (SI) accepted form. Numeric ranges are inclusive of the numbers defining the range. Unless otherwise indicated, nucleotide sequences are written left to right in 5' to 3' orientation. Amino acid sequences are written left to right in amino to carboxy orientation. The headings provided herein are not limitations of the various aspects of the disclosure, which can be had by reference to the specification as a whole. Accordingly, the terms defined immediately below are more fully defined by reference to the specification in its entirety. [0060] "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-L1 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 intrasternal 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. [0061] 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. [0062] 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 VH) and a heavy chain constant region. The heavy chain constant region comprises three constant domains, CH1, CH2 and CH3. Each light chain comprises a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region comprises one constant domain, CL. The VH and VL 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 VH and VL 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 (C1q) 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. [0063] 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 IgG1, IgG2, IgG3 and IgG4. "Isotype" refers to the antibody class or subclass (e.g., IgM or IgG1) 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 antigen- binding portion of any of the aforementioned immunoglobulins, and includes a monovalent and a divalent fragment or portion, and a single chain antibody. [0064] 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. [0065] 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. [0066] 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. [0067] 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. [0068] 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. [0069] 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-L1 antibody binds specifically to PD-L1, and an anti-CTLA-4 antibody binds specifically to CTLA-4. [0070] 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 antigen- binding 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-L1 antibody described herein, include (i) a Fab fragment (fragment from papain cleavage) or a similar monovalent fragment consisting of the VL, VH, LC and CH1 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 CH1 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. [0071] 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. [0072] 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. [0073] "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. [0074] "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- L1), variants, isoforms, and species homologs of hPD-L1, and analogs having at least one common epitope with hPD-L1. The complete hPD-L1 sequence can be found under GenBank Accession No. Q9NZQ7. The human PD-L1 protein is encoded by the human CD274 gene (NCBI Gene ID: 29126). [0075] "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). [0076] "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 P18627. The human LAG-3 protein is encoded by the human LAG3 gene (NCBI Gene ID: 3902). [0077] "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, that 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). [0078] "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. [0079] 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. [0080] The use of the term "flat dose" with regard to the methods and dosages of the disclosure means a dose that is administered to a patient without regard for the weight or body surface area (BSA) of the patient. The flat dose is therefore not provided as a mg/kg dose, but rather as an absolute amount of the agent (e.g., the anti-PD-1 antibody). For example, a 60 kg person and a 100 kg person would receive the same dose of an antibody (e.g., 240 mg of an anti- PD-1 antibody). [0081] The use of the term "fixed dose" with regard to a method of the disclosure means that two or more different antibodies in a single composition (e.g., anti-PD-1 antibody and anti- CTLA-4 antibody or an anti-PD-L1 antibody and an anti-CTLA-4 antibody) are present in the composition in particular (fixed) ratios with each other. In some aspects, the fixed dose is based on the weight (e.g., mg) of the antibodies. In certain aspects, the fixed dose is based on the concentration (e.g., mg/ml) of the antibodies. In some aspects, the ratio is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:15, about 1:20, about 1:30, about 1:40, about 1:50, about 1:60, about 1:70, about 1:80, about 1:90, about 1:100, about 1:120, about 1:140, about 1:160, about 1:180, about 1:200, about 200:1, about 180:1, about 160:1, about 140:1, about 120:1, about 100:1, about 90:1, about 80:1, about 70:1, about 60:1, about 50:1, about 40:1, about 30:1, about 20:1, about 15:1, about 10:1, about 9:1, about 8:1, about 7:1, about 6:1, about 5:1, about 4:1, about 3:1, or about 2:1 mg first antibody (e.g., anti-PD-1 antibody or an anti-PD-L1 antibody) to mg second antibody (e.g., anti- CTLA-4 antibody). For example, the 3:1 ratio of an anti-PD-1 antibody and an anti-CTLA-4 antibody can mean that a vial can contain about 240 mg of the anti-PD-1 antibody and 80 mg of the anti-CTLA-4 antibody or about 3 mg/ml of the anti-PD-1 antibody and 1 mg/ml of the anti- CTLA-4 antibody. [0082] The term "weight-based dose" as referred to herein means that a dose that is administered to a patient is calculated based on the weight of the patient. For example, when a patient with 60 kg body weight requires 3 mg/kg of an anti-PD-1 antibody, one can calculate and use the appropriate amount of the anti-PD-1 antibody (i.e., 180 mg) for administration. [0083] 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. [0084] 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. [0085] 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. [0086] 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. [0087] 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 long- term monitoring of the effects of these agents on the target disease. [0088] 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). [0089] 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. [0090] 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. [0091] 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. [0092] The term "tumor mutation burden" (TMB) as used herein refers to the number of somatic mutations in a tumor’s genome and/or the number of somatic mutations per area of the tumor’s genome. Germline (inherited) variants are excluded when determining TMB, because the immune system has a higher likelihood of recognizing these as self. Tumor mutation burden (TMB) can also be used interchangeably with "tumor mutation load," "tumor mutational burden," or "tumor mutational load." [0093] TMB is a genetic analysis of a tumor’s genome and, thus, can be measured by applying sequencing methods well known to those of skill in the art. The tumor DNA can be compared with DNA from patient-matched normal tissue to eliminate germline mutations or polymorphisms. [0094] In some aspects, TMB is determined by sequencing tumor DNA using a high- throughput sequence technique, e.g., next-generation sequencing (NGS) or an NGS-based method. In some aspects, the NGS-based method is selected from whole genome sequencing (WGS), whole exome sequencing (WES), or comprehensive genomic profiling (CGP) of cancer gene panels such as FOUNDATIONONE CDX™ and MSK-IMPACT clinical tests. In some aspects, TMB, as used herein, refers to the number of somatic mutations per megabase (Mb) of DNA sequenced. In one aspect, TMB is measured using the total number of nonsynonymous mutations, e.g., missense mutation (i.e. changing a particular amino acid in the protein) and/or nonsense (causing premature termination and thus truncation of the protein sequence), identified by normalizing matched tumor with germline samples to exclude any inherited germline genetic alterations. In another aspect, TMB is measured using the total number of missense mutations in a tumor. In order to measure TMB, a sufficient amount of sample is required. In one aspect, tissue sample (for example, a minimum of 10 slides) is used for evaluation. In some aspects, TMB is expressed as NsMs per megabase (NsM/Mb).1 megabase represents 1 million bases. [0095] The TMB status can be a numerical value or a relative value, e.g., high, medium, or low; within the highest fractile, or within the top tertile, of a reference set. [0096] The term "high TMB" as used herein refers to a number of somatic mutations in a tumor’s genome that is above a number of somatic mutations that is normal or average. In some aspects, a TMB has a score of at least 210, at least 215, at least 220, at least 225, at least 230, at least 235, at least 240, at least 245, at least 250, at least 255, at least 260, at least 265, at least 270, at least 275, at least 280, at least 285, at least 290, at least 295, at least 300, at least 305, at least 310, at least 315, at least 320, at least 325, at least 330, at least 335, at least 340, at least 345, at least 350, at least 355, at least 360, at least 365, at least 370, at least 375, at least 380, at least 385, at least 390, at least 395, at least 400, at least 405, at least 410, at least 415, at least 420, at least 425, at least 430, at least 435, at least 440, at least 445, at least 450, at least 455, at least 460, at least 465, at least 470, at least 475, at least 480, at least 485, at least 490, at least 495, or at least 500; in other aspects a high TMB has a score of at least at least 221, at least 222, at least 223, at least 224, at least 225, at least 226, at least 227, at least 228, at least 229, at least 230, at least 231, at least 232, at least 233, at least 234, at least 235, at least 236, at least 237, at least 238, at least 239, at least 240, at least 241, at least 242, at least 243, at least 244, at least 245, at least 246, at least 247, at least 248, at least 249, or at least 250; and, in a particular aspect, a high TMB has a score of at least 243. [0097] In other aspects, a "high TMB" refers to a TMB within the highest fractile of the reference TMB value. For example, all subjects with evaluable TMB data are grouped according to fractile distribution of TMB, i.e., subjects are rank ordered from highest to lowest number of genetic alterations and divided into a defined number of groups. In one aspect, all subjects with evaluable TMB data are ranked ordered and divided into thirds, and a "high TMB" is within the top tertile of the reference TMB value. In a particular aspect, the tertile boundaries are 0 < 100 genetic alterations; 100 to 243 genetic alterations; and > 243 genetic alterations. It should be understood that, once rank ordered, subjects with evaluable TMB data can be divided into any number of groups, e.g., quartiles, quintiles, etc. [0098] In some aspects, a "high TMB" refers to a TMB of at least about 20 mutations/tumor, at least about 25 mutations/tumor, at least about 30 mutations/tumor, at least about 35 mutations/tumor, at least about 40 mutations/tumor, at least about 45 mutations/tumor, at least about 50 mutations/tumor, at least about 55 mutations/tumor, at least about 60 mutations/tumor, at least about 65 mutations/tumor, at least about 70 mutations/tumor, at least about 75 mutations/tumor, at least about 80 mutations/tumor, at least about 85 mutations/tumor, at least about 90 mutations/tumor, at least about 95 mutations/tumor, or at least about 100 mutations/tumor. In some aspects, a "high TMB" refers to a TMB of at least about 105 mutations/tumor, at least about 110 mutations/tumor, at least about 115 mutations/tumor, at least about 120 mutations/tumor, at least about 125 mutations/tumor, at least about 130 mutations/tumor, at least about 135 mutations/tumor, at least about 140 mutations/tumor, at least about 145 mutations/tumor, at least about 150 mutations/tumor, at least about 175 mutations/tumor, or at least about 200 mutations/tumor. In certain aspects, a tumor having a high TMB has at least about 100 mutations/tumor. [0099] The "high TMB" can also be referred to as the number of mutations per megabase of tumor genome sequenced, e.g., as measured by a mutation assay, e.g., FOUNDATIONONE® CDX™ assay. In one aspect, the high TMB refers to at least about 9, at least about 10, at least about 11, at least 12, at least about 13, at least about 14, at least about 15, at least about 16, at least about 17, at least about 18, at least about 19, or at least about 20 mutations per megabase of genome as measured by a FOUNDATIONONE® CDX™ assay. In a particular aspect, the "high TMB" refers to at least 10 mutations per megabase of genome sequenced by a FOUNDATIONONE® CDX™ assay. [0100] As used herein, the term "medium TMB" refers to a number of somatic mutations in a tumor’s genome that is at or around a number of somatic mutations that is normal or average and the term "low TMB" refers to a number of somatic mutations in a tumor’s genome that is below a number of somatic mutations that is normal or average. In a particular aspect, a "high TMB" has a score of at least 243, a "medium TMB" has a score of between 100 and 242, and a "low TMB" has a score of less than 100 (or between 0 and 100). The "medium or low TMB" refers to less than 9 mutations per megabase of genome sequenced, e.g., as measured by a FOUNDATIONONE® CDX™ assay. [0101] The term "reference TMB value" as referred to herein can be the TMB value shown in Table 9. [0102] In some aspects, TMB status can correlate with smoking status. In particular, subjects who currently or formerly smoke(d) often have more genetic alterations, e.g., missense mutations, than subjects who never smoke(d). [0103] A tumor with a high TMB can also have a high neoantigen load. As used herein, the term "neoantigen" refers to a newly formed antigen that has not been previously recognized by the immune system. A neoantigen can be a protein or peptide that is recognized as foreign (or non-self) by the immune system. Transcription of a gene in the tumor genome harboring a somatic mutation results in mutated mRNA that, when translated, gives rise to a mutated protein, which is then processed and transported to the ER lumen and binds to MHC class I complex, facilitating T-cell recognition of the neoantigen. Neoantigen recognition can promote T-cell activation, clonal expansion, and differentiation into effector and memory T-cells. Neoantigen load can correlate with TMB. In some aspects, TMB is assessed as a surrogate for measuring tumor neoantigen load. The TMB status of a tumor, can be used as a factor, alone or in combination with other factors, in determining whether a patient is likely to benefit from a particular anti-cancer agent or type of treatment or therapy, e.g., a combination therapy comprising (a) an anti-PD-1 antibody or an anti-PD-L1 antibody and (b) an anti-CTLA-4 antibody. In one aspect, a high TMB status (or a high TMB) indicates an enhanced likelihood of benefit from immuno-oncology and, thus, can be used to identify patients more likely to benefit from therapy of a combination therapy comprising (a) an anti-PD-1 antibody or an anti-PD-L1 antibody and (b) an anti-CTLA-4 antibody. Similarly, tumors with high tumor neoantigen load and high TMB are more likely to be immunogenic than tumors with low neoantigen load and low TMB. In addition, high-neoantigen/high-TMB tumors are more likely to be recognized as non- self by the immune system, thus triggering an immune-mediated antitumor response. In one aspect, a high TMB status and a high neoantigen load indicate an enhanced likelihood of benefit from immuno-oncology, e.g., a combination therapy comprising (a) an anti-PD-1 antibody or an anti-PD-L1 antibody and (b) an anti-CTLA-4 antibody. As used herein, the term "benefit from therapy" refers to an improvement in one or more of overall survival, progression-free survival, partial response, complete response, and overall response rate and can also include a reduction in tumor growth or size, 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. [0104] Other factors, e.g., environmental factors, can associate with TMB status. For example, smoking status of patients with NSCLC was correlated with TMB distribution, whereby current and former smokers had higher median TMB compared with those patients who had never smoked. See Peters et al., AACR, April 1-5, 2017, Washington, D.C. The presence of a driver mutation in NSCLC tumors was associated with younger age, female sex, and non-smoker status. See Singal et al., ASCO, June 1-5, 2017; Chicago, IL. A trend associating the presence of driver mutations, such as EGFR, ALK, or KRAS, with lower TMB was observed (P = 0.06). Davis et al., AACR, April 1-5, 2017, Washington, D.C. [0105] The term "somatic mutation" as used herein refers to an acquired alteration in DNA that occurs after conception. Somatic mutations can occur in any of the cells of the body except the germ cells (sperm and egg) and therefore are not passed on to children. These alterations can, but do not always, cause cancer or other diseases. The term "germline mutation" refers to a gene change in a body's reproductive cell (egg or sperm) that becomes incorporated into the DNA of every cell in the body of the offspring. Germline mutations are passed on from parents to offspring. Also called a "hereditary mutation." In the analysis of TMB, germline mutations are considered as a "baseline," and are subtracted from the number of mutations found in the tumor biopsy to determine the TMB within the tumor. As germline mutations are found in every cell in the body, their presence can be determined via less invasive sample collections than tumor biopsies, such as blood or saliva. Germline mutations can increase the risk of developing certain cancers, and can play a role in the response to chemotherapy. [0106] The term "measuring" or "measured" or "measurement" when referring to TMB status means determining a measurable quantity of somatic mutations in a biological sample of the subject. It will be appreciated that measuring can be performed by sequencing nucleic acids, e.g., cDNA, mRNA, exoRNA, ctDNA, and cfDNA, in the sample. The measuring is performed on a subject's sample and/or a reference sample or samples and can, for example, be detected de novo or correspond to a previous determination. The measuring can be performed, for example, using PCR methods, qPCR methods, Sanger sequencing methods, genomic profiling methods (including comprehensive gene panels), exome sequencing methods, genome sequencing methods, and/or any other method disclosed herein, as is known to a person of skill in the art. In some aspects, the measuring identifies a genomic alteration in the sequenced nucleic acids. The genomic (or gene) profiling methods can involve panels of a predetermined set of genes, e.g., 150-500 genes, and in some instances the genomic alterations evaluated in the panel of genes are correlated with total somatic mutations evaluated. As used herein when referring to sequencing, the term "gene" includes DNA coding regions (e.g., exons), DNA non-coding regions associated with a coding region (e.g., introns and promoters), and mRNA transcripts. [0107] The term "genomic alteration" as used herein refers to a change (or mutation) in the nucleotide sequence of the genome of a tumor, which change is not present in the germline nucleotide sequence, and which in some aspects is a nonsynonymous mutation including, but not limited to, a base pair substitution, a base pair insertion, a base pair deletion, a copy number alteration (CNA), a gene rearrangement, and any combination thereof. In a particular aspect, the genomic alterations measured in the biological sample are missense mutations. [0108] The term "whole genome sequencing" or "WGS," as used herein, refers to a method of sequencing the entire genome. The term "whole exome sequencing" or "WES," as used herein, refers to a method of sequencing all the protein-coding regions (exons) of the genome. [0109] 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. [0110] The term "genomic profiling assay," "comprehensive genomic profiling," or "CGP" refers to an assay that analyzes a panel of genes and select introns for in vitro diagnosis. CGP is a combination of NGS and targeted bioinformatics analysis to screen for mutations in known clinically relevant cancer genes. This method can be used to catch mutations that are missed by testing "hotspots" (e.g., BRCA1/BRCA2 mutations or microsatellite markers). In some aspects, the CGP further includes one or more mRNA transcript, non-coding RNA, and/or promoter region. In one aspect, the genes in the panel are cancer-related genes. In another aspect, a genomic profiling assay is a FOUNDATIONONE® assay. [0111] The term "harmonization" refers to a study conducted to determine the comparability between two or more measures and/or diagnostic tests. Harmonization studies provide a systematic approach to address questions of how diagnostic tests compare with each other, as well as their interchangeability when used to determine the biomarker status of a patient’s tumor. In general, at least one well-characterized measure and/or diagnostic test is used as a standard for comparison with others. Concordance assessment is often utilized in harmonization studies. [0112] The term "concordance," as used herein, refers to a degree of agreement between two measurements and/or diagnostic tests. Concordance can be established using both qualitative and quantitative methods. Quantitative methods to assess concordance differ based on the type of measurement. A particular measurement can be expressed either as 1) a categorical/dichotomized variable or 2) a continuous variable. A "categorical/dichotomized variable" (e.g., above or below TMB cut-off) may use percent agreements, such as overall percent agreement (OPA), positive percent agreement (PPA), or negative percent agreement (NPA), to assess concordance. A "continuous variable" (e.g., TMB by WES) uses Spearman’s rank correlation or Pearson’s correlation coefficient (r), which takes on values -1 ≤ r ≤ +1, to assess concordance across a spectrum of values (Note r = +1 or -1 means that each of the variables is perfectly correlated). The term "analytical concordance" refers to the degree of agreement in the performance (e.g., identification of biomarkers, genomic alteration types, and genomic signatures, and assessment of test reproducibility) of two assays or diagnostic tests to support clinical use. The term "clinical concordance" refers to the degree of agreement in how the two assays or diagnostic tests correlate with clinical outcome. [0113] 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. [0114] 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 TMB, 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 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. [0115] The terms "once about every week," "once about every two weeks," or any other similar dosing interval terms as used herein mean approximate numbers. "Once about every week" can include every seven days ± one day, i.e., every six days to every eight days. "Once about every two weeks" can include every fourteen days ± three days, i.e., every eleven days to every seventeen days. Similar approximations apply, for example, to once about every three weeks, once about every four weeks, once about every five weeks, once about every six weeks, and once about every twelve weeks. In some aspects, a dosing interval of once about every six weeks or once about every twelve weeks means that the first dose can be administered any day in the first week, and then the next dose can be administered any day in the sixth or twelfth week, respectively. In other aspects, a dosing interval of once about every six weeks or once about every twelve weeks means that the first dose is administered on a particular day of the first week (e.g., Monday) and then the next dose is administered on the same day of the sixth or twelfth weeks (i.e., Monday), respectively. [0116] 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. [0117] 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. [0118] As described herein, any concentration range, percentage range, ratio range or integer range is to be understood to include the value of any integer within the recited range and, when appropriate, fractions thereof (such as one tenth and one hundredth of an integer), unless otherwise indicated. [0119] Abbreviations used herein are defined throughout the present disclosure. A list of additional abbreviations is provided in Table 1. Table 1: List of Abbreviations
Figure imgf000033_0001
Figure imgf000034_0001
[0120] Various aspects of the disclosure are described in further detail in the following subsections. II. Methods of the Disclosure [0121] The present disclosure is directed to methods of treating a tumor in a human subject, comprising administering an effective amount of a checkpoint inhibitor to the subject, e.g., an anti-PD-1 antibody, anti-PD-L1 antibody, and/or anti-LAG-3 antibody, wherein the subject meets at least two of the following conditions: the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined. II.A. Parenchymal Gene Panel [0122] Certain aspects of the present disclosure are directed to methods of identifying a subject having a parenchymal inflammatory phenotype, determined by measuring the expression of a panel of genes comprising at least about 5 genes in a sample obtained from the subject. In some aspects, the panel of genes comprises at least about 5 genes, at least about 6 genes, at least about 7 genes, at least about 8 genes, at least about 9 genes, at least about 10 genes, at least about 11 genes, at least about 12 genes, at least about 13 genes, at least about 14 genes, at least about 15 genes, at least about 16 genes, at least about 17 genes, at least about 18 genes, at least about 19 genes, at least about 20 genes, at least about 25 genes, at least about 30 genes, at least about 35 genes, at least about 40 genes, at least about 45 genes, at least about 50 genes, at least about 55 genes, at least about 60 genes, at least about 63 genes, at least about 65 genes, at least about 70 genes, at least about 80 genes, at least about 85 genes, at least about 90 genes, or at least about 95 genes. In some aspects, the panel of genes comprises at least about 10 genes. In some aspects, the panel of genes comprises at least about 20 genes. In some aspects, the panel of genes comprises at least about 30 genes. In some aspects, the panel of genes comprises at least about 40 genes. In some aspects, the panel of genes comprises at least about 50 genes. In some aspects, the panel of genes comprises at least about 60 genes. In some aspects, the panel of genes comprises at least about 70 genes. In some aspects, the panel of genes comprises at least about 80 genes. In some aspects, the panel of genes comprises at least about 90 genes. In some aspects, the panel of genes comprises at least about 63 genes. In some aspects, the panel of genes comprises at least about 95 genes. [0123] In some aspects, the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA- 4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin-9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA- DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3. [0124] In some aspects, wherein the panel of genes comprises at least about 5 additional genes, at least about 10 additional genes, at least about 15 additional genes, at least about 20 additional genes, at least about 25 additional genes, at least about 30 additional genes, at least about 35 additional genes, at least about 40 additional genes, at least about 45 additional genes, at least about 50 additional genes, at least about 55 additional genes, at least about 60 additional genes, at least about 65 additional genes, at least about 70 additional genes, at least about 75 additional genes, at least about 80 additional genes, at least about 85 additional genes, at least about 90 additional genes, at least about 95 additional genes, at least about 100 additional genes, at least about 110 additional genes, at least about 120 additional genes, at least about 130 additional genes, at least about 140 additional genes, at least about 150 additional genes, at least about 160 additional genes, at least about 170 additional genes, at least about 180 additional genes, at least about 190 additional genes, or at least about 200 additional genes. In some aspects, at least one of the additional genes is selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin-9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA- DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof. [0125] In some aspects, the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA- 4, Pan-KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) 2 additional genes, 3 additional genes, 4 additional genes, 5 additional genes, 6 additional genes, 7 additional genes, 8 additional genes, 9 additional genes, 10 additional genes, 11 additional genes, 12 additional genes, 13 additional genes, 14 additional genes, 15 additional genes, 16 additional genes, or 17 additional genes. In some aspects, the gene panel consists essentially of (or consists of) (i) CD274 (PD-L1) and LAG3, and (ii) 2 additional genes, 3 additional genes, 4 additional genes, 5 additional genes, 6 additional genes, 7 additional genes, 8 additional genes, 9 additional genes, 10 additional genes, 11 additional genes, 12 additional genes, 13 additional genes, 14 additional genes, 15 additional genes, 16 additional genes, 17 additional genes, 18 additional genes, 19 additional genes, 20 additional genes, 21 additional genes, 22 additional genes, 23 additional genes, 24 additional genes, 25 additional genes, 26 additional genes, 27 additional genes, 28 additional genes, 29 additional genes, 30 additional genes, 31 additional genes, 32 additional genes, 33 additional genes, 34 additional genes, 35 additional genes, 36 additional genes, 37 additional genes, 38 additional genes, 39 additional genes, 40 additional genes, 41 additional genes, 42 additional genes, 43 additional genes, 44 additional genes, 45 additional genes, 46 additional genes, 47 additional genes, 48 additional genes, 49 additional genes, 50 additional genes, 51 additional genes, 52 additional genes, 53 additional genes, 54 additional genes, 55 additional genes, 56 additional genes, 57 additional genes, or 58 additional genes, wherein the additional genes are selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin-9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof. [0126] 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. [0127] 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, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof. 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 ANT1, 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. [0128] In some aspects, the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3. [0129] In some aspects, the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0130] In some aspects, the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA- DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. [0131] In some aspects, the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA- DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ. II.B.1. Parenchymal Inflammatory Phenotype [0132] In certain aspects of the present disclosure, a parenchymal inflammatory phenotype is characterized by a low parenchymal signature score ("signature score"). The signature score, as used herein, is a measurement of the composite expression of the genes in the gene panel in a tumor sample form the subject, and comparing the composite expression of the genes in the gene panel to the average composite expression of the panel of genes in tumor samples obtained from a population of subjects afflicted with the tumor. In some aspects, the average composite signature score is determined by measuring the composite expression of the panel of genes in tumor samples obtained from the population of subjects. In some aspects, a low signature score is characterized by a signature score of less than about 50, less than about 45, less than about 40, less than about 35, less than about 30, less than about 25, less than about 20, less than about 15, less than about 10, or less than about 5; wherein a signature score of 100 indicates that the subject has no parenchymal inflammatory phenotype; and wherein a signature score of 1 indicates that a subject has a high parenchymal phenotype. In some aspects, a low signature score is characterized by a signature score of less than about 50. In some aspects, a low signature score is characterized by a signature score of less than about 45. In some aspects, a low signature score is characterized by a signature score of less than about 40. In some aspects, a low signature score is characterized by a signature score of less than about 35. In some aspects, a low signature score is characterized by a signature score of less than about 30. In some aspects, a low signature score is characterized by a signature score of less than about 25. In some aspects, a low signature score is characterized by a signature score of less than about 20. In some aspects, a low signature score is characterized by a signature score of less than about 15. In some aspects, a low signature score is characterized by a signature score of less than about 10. In some aspects, a low signature score is characterized by a signature score of less than about 9. In some aspects, a low signature score is characterized by a signature score of less than about 8. In some aspects, a low signature score is characterized by a signature score of less than about 7. In some aspects, a low signature score is characterized by a signature score of less than about 6. In some aspects, a low signature score is characterized by a signature score of less than about 5. In some aspects, a low signature score is characterized by a signature score of less than about 4. In some aspects, a low signature score is characterized by a signature score of less than about 3. In some aspects, a low signature score is characterized by a signature score of less than about 2. In some aspects, a low signature score is characterized by a signature score of about 1. [0133] 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 the checkpoint inhibitor therapy. 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. [0134] 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 panel of genes is determined by detecting the presence of mRNA transcribed from the gene, the presence of a protein encoded by the gene, or both. [0135] In some aspects, the expression of one or more of the genes is determined by measuring the level of gene mRNA in a sample obtained from the subject. 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 RNA in situ hybridization. [0136] In some aspects, the expression of one or more of the genes is determined by measuring the level of protein expressed from the gene 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 protein is measured using an immunohistochemistry (IHC) assay. In certain aspects, the IHC is an automated IHC. [0137] In some aspects, the expression of one or more of the genes of the inflammatory gene panel is normalized relative to the expression of one or more housekeeping genes. In some aspects, the one or more housekeeping genes comprises any housekeeping gene disclosed herein or any combination thereof. [0138] In some aspects, raw gene expression values are normalized following standard gene expression profiling (GEP) protocols. In these aspects, gene expression signature scores 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, scores have positive or negative values, depending on whether gene expression is up- or down-regulated under a particular condition. [0139] In certain aspects, a low signature score is characterized by a signature score that is lower than a reference signature score. In some aspects, the reference inflammatory signature score is an average signature score. In some aspects, the average signature score is determined by measuring the expression of the genes present in the panel of genes 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 checkpoint inhibitor, e.g., the anti-PD-1 antibody, the anti-PD-L1 antibody, the anti-CTLA-4 antibody, the anti-LAG-3 antibody, or any combination thereof. [0140] In some aspects, a low signature score is characterized by a signature score that is at less than about 95%, less than about 90%, less than about 85%, less than about 80%, less than about 80%, less than about 75%, less than about 70%, less than about 65%, less than about 60%, less than about 55%, less than about 50%, less than about 45%, less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 95% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about % that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 90% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 85% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 80% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 75% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 70% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 65% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 60% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 55% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 50% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 45% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 40% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 35% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 30% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 25% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 20% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 15% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 10% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 9% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 8% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 7% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 6% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 5% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 4% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 3% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 2% that of the average signature score. In some aspects, a low signature score is characterized by a signature score that is at less than about 1% that of the average signature score. II.B. Tumor Mutation Burden (TMB) [0141] Certain aspects of the present disclosure are directed to a method for treating a human subject afflicted with a tumor comprising administering a checkpoint inhibitor, e.g., an anti-PD-1 antibody or an anti-PD-L1 antibody, to the subject, wherein the subject is identified as exhibiting a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined prior to the administration. The disclosure is based on the fact that tumor immunogenicity is directly related to TMB and/or neoantigen load. [0142] As a tumor grows, it accumulates somatic mutations not present in germline DNA. TMB refers to the number of somatic mutations in a tumor’s genome and/or the number of somatic mutations per area of the tumor genome (after taking into account germline variant DNA). The acquisition of somatic mutations and, thus, a higher TMB can be influenced by distinct mechanisms, such as exogenous mutagen exposure (e.g., tobacco smoking) and DNA mismatch repair mutations (e.g., MSI in colorectal and esophageal cancers). In solid tumors, about 95% of mutations are single-base substitutions. (Vogelstein et al., Science (2013) 339:1546-1558.) A "nonsynonymous mutation" herein refers to a nucleotide mutation that alters the amino acid sequence of a protein. Missense mutations and nonsense mutations can be both nonsynonymous mutations. A "missense mutation" herein refers to a nonsynonymous point mutation in which a single nucleotide change results in a codon that codes for a different amino acid. A "nonsense mutation" herein refers to a nonsynonymous point mutation in which a codon is changed to a premature stop codon that leads to truncation of the resulting protein. [0143] In some aspects, somatic mutations can be expressed at the RNA and/or protein level, resulting in neoantigens (also referred to as neoepitopes). Neoantigens can influence an immune-mediated anti-tumor response. For example, neoantigen recognition can promote T-cell activation, clonal expansion, and differentiation into effector and memory T-cells. [0144] As a tumor develops, early clonal mutations (or "trunk mutations") can be carried by most or all tumor cells, while late mutations (or "branch mutations") can occur in only a subset of tumor cells or regions. (Yap et al., Sci Tranl Med (2012) 4:1-5; Jamai-Hanjani et al., (2015) Clin Cancer Res 21:1258-1266.) As a result, neoantigens derived from clonal "trunk" mutations are more widespread in the tumor genome than "branch" mutations and, thus, can lead to a high number of T cells reactive against the clonal neoantigen. (McGranahan et al., (2016) 351:1463-1469.) Generally, tumors with a high TMB can also have a high neoantigen load, which can lead to high tumor immunogenicity and increased T-cell reactivity and anti-tumor response. As such, cancers with a high TMB can respond well to treatment with immunotherapies, e.g., an anti-PD-1 antibody or anti-PD-L1 antibody. [0145] Advances in sequencing technologies allow for evaluation of the tumor’s genomic mutation landscape. Any sequencing methods known to those of skill in the art can be used to sequence nucleic acids from the tumor genome (e.g., obtained from a biological sample from a subject afflicted with a tumor). In one aspect, PCR or qPCR methods, Sanger sequencing methods, or next-generation sequencing ("NGS") methods (such as genomic profiling, exome sequencing, or genome sequencing) can be used to measure TMB. In some aspects, the TMB status is measured using genomic profiling. Genomic profiling involves analyzing nucleic acids from tumor samples, including coding and non-coding regions, and can be performed using methods having integrated optimized nucleic acid selection, read alignment, and mutation calling. In some aspects, gene profiling provides next generation sequencing (NGS)-based analysis of tumors that can be optimized on a cancer-by-cancer, gene-by-gene, and/or site-by-site basis. Genome profiling can integrate the use of multiple, individually tuned, alignment methods or algorithms to optimize performance in sequencing methods, particularly in methods that rely on massively parallel sequencing of a large number of diverse genetic events in a large number of diverse genes. Genomic profiling provides for a comprehensive analysis of a subject's cancer genome, with clinical grade quality, and the output of the genetic analysis can be contextualized with relevant scientific and medical knowledge to increase the quality and efficiency of cancer therapy. II.B.1. Genomic Profiling [0146] Genomic profiling involves a panel of a predefined set of genes comprising as few as five genes or as many as 1000 genes, about 25 genes to about 750 genes, about 100 genes to about 800 genes, about 150 genes to about 500 genes, about 200 genes to about 400 genes, about 250 genes to about 350 genes. In one aspect, the genomic profile comprises at least 300 genes, at least 305 genes, at least 310 genes, at least 315 genes, at least 320 genes, at least 325 genes, at least 330 genes, at least 335 genes, at least 340 genes, at least 345 genes, at least 350 genes, at least 355 genes, at least 360 genes, at least 365 genes, at least 370 genes, at least 375 genes, at least 380 genes, at least 385 genes, at least 390 genes, at least 395 genes, or at least 400 genes. In another aspect, the genomic profile comprises at least 325 genes. In a particular aspect, the genomic profile comprises at least 315 cancer-related genes and introns in 28 genes (FOUNDATIONONE®) or the complete DNA coding sequence of 406 genes, introns in 31 genes with rearrangements, and the RNA sequence (cDNA) of 265 genes (FOUNDATIONONE® Heme). In another aspect, the genomic profile comprises 26 genes and 1000 associated mutations (EXODX® Solid Tumor). In yet another aspect, the genomic profile comprises 76 genes (Guardant360). In yet another aspect, the genomic profile comprises 73 genes (Guardant360). In another aspect, the genomic profile comprises 354 genes and introns in 28 genes for rearrangements (FOUNDATIONONE® CDXTM). In certain aspects, the genomic profile is FOUNDATIONONE® F1CDx. In another aspect, the genomic profile comprises 468 genes (MSK-IMPACTTM). One or more genes can be added to the genome profile as more genes are identified to be related to oncology. II.B.1.a. FOUNDATIONONE® Assay [0147] The FOUNDATIONONE® assay is comprehensive genomic profiling assay for solid tumors, including but not limited to solid tumors of the lung, colon, and breast, melanoma, and ovarian cancer. The FOUNDATIONONE® assay uses a hybrid-capture, next-generation sequencing test to identify genomic alterations (base substitutions, insertions and deletions, copy number alterations, and rearrangements) and select genomic signatures (e.g., TMB and microsatellite instability). The assay covers 322 unique genes, including the entire coding region of 315 cancer-related genes, and selected introns from 28 genes. The full list of FOUNDATIONONE® assay genes is provided in Tables 2 and 3. See FOUNDATIONONE: Technical Specifications, Foundation Medicine, Inc., available at FoundationMedicine.com, last visited March 16, 2018, which is incorporated by reference herein in its entirety. Table 2: List of genes wherein entire coding sequences are assayed in the FOUNDATIONONE® assay.
Figure imgf000046_0001
Figure imgf000047_0001
Table 3: List of genes wherein selected introns are assayed in the FOUNDATIONONE® assay.
Figure imgf000047_0002
II.B.1.b. EXODX® Solid Tumor Assay [0148] In one aspect, TMB is measured using the EXODX® Solid Tumor assay. The EXODX® Solid Tumor assay is an exoRNA- and cfDNA-based assay, which detects actionable mutations in cancer pathways. The EXODX® Solid Tumor assay is a plasma-based assay that does not require a tissue sample. The EXODX® Solid Tumor assay covers 26 genes and 1000 mutations. The specific genes covered by the EXODX® Solid Tumor assay are shown in Table 4. See Plasma-Based Solid Tumor Mutation Panel Liquid Biopsy, Exosome Diagnostics, Inc., available at exosomedx.com, last accessed on March 25, 2019. Table 4: Genes covered by the EXODX® Solid Tumor assay.
Figure imgf000048_0001
II.B.1.c. Guardant360 Assay [0149] In some aspects, TMB status is determined using the Guardant360 assay. The Guardant360 assay measures mutations in at least 73 genes (Table 5), 23 indels (Table 6), 18 CNVs (Table 7), and 6 fusion genes (Table 8). See GuardantHealth.com, last accessed on March 25, 2019. Table 5: Guardant360 assay genes.
Figure imgf000048_0002
Table 6: Guardant360 assay indels.
Figure imgf000048_0003
Table 7: Guardant360 assay amplifications (CNVs).
Figure imgf000049_0001
Table 8: Guardant360 assay fusions.
Figure imgf000049_0002
II.B.1.d. ILLUMINA® TruSight Assay [0150] In some aspects, TMB is determined using the TruSight Tumor 170 assay (ILLUMINA). The TruSight Tumor 170 assay is a next-generation sequencing assay that covers 170 genes associated with common solid tumors, which simultaneously analyzes DNA and RNA. The TruSight Tumor 170 assay assesses fusions, splice variants, insertions/deletions, single nucleotide variants (SNVs), and amplifications. The TruSight Tumor 170 assay gene lists are shown in Tables 12-14. Table 9: TruSight Tumor 170 assay genes (amplifications).
Figure imgf000049_0003
Table 10: TruSight Tumor 170 assay genes (fusions).
Figure imgf000049_0004
Table 11: TruSight Tumor 170 assay genes (small variants).
Figure imgf000049_0005
Figure imgf000050_0001
II.B.1.e. FOUNDATIONONE® F1CDx Assay [0151] FOUNDATIONONE® CDXTM ("F1CDx") is a next generation sequencing based in vitro diagnostic device for detection of substitutions, insertion and deletion alterations (indels), and copy number alterations (CNAs) in 324 genes and select gene rearrangements, as well as genomic signatures including microsatellite instability (MSI) and tumor mutation burden (TMB) using DNA isolated from formalin-fixed paraffin embedded (FFPE) tumor tissue specimens. F1CDx is approved by the United States Food and Drug Administration (FDA) for several tumor indications, including NSCLC, melanoma, breast cancer, colorectal cancer, and ovarian cancer. [0152] The F1CDx assay employs a single DNA extraction method from routine FFPE biopsy or surgical resection specimens, 50-1000 ng of which will undergo whole-genome shotgun library construction and hybridization-based capture of all coding exons from 309 cancer-related genes, one promoter region, one non-coding (ncRNA), and selected intronic regions from 34 commonly rearranged genes, 21 of which also include the coding exons. Tables 12 and 13 provide the complete list of genes included in F1CDx. In total, the assay detects alterations in a total of 324 genes. Using the ILLUMINA® HiSeq 4000 platform, hybrid capture– selected libraries are sequenced to high uniform depth (targeting >500X median coverage with >99% of exons at coverage >100X). Sequence data is then processed using a customized analysis pipeline designed to detect all classes of genomic alterations, including base substitutions, indels, copy number alterations (amplifications and homozygous gene deletions), and selected genomic rearrangements (e.g., gene fusions). Additionally, genomic signatures including microsatellite instability (MSI) and tumor mutation burden (TMB) are reported. Table 12: Genes with full coding exonic regions included in FOUNDATIONONE® CDX™ for the detection of substitutions, insertions and deletions (indels), and copy number alterations (CNAs).
Figure imgf000051_0001
Table 13: Genes with selected intronic regions for the detection of gene rearrangements, one with 3’UTR, one gene with a promoter region and one ncRNA gene.
Figure imgf000051_0002
[0153] The F1CDx assay identifies various alterations in the gene and/or intron sequences, including substitutions, insertions/deletions, and CNAs. The F1CDx assay was previously identifies as having concordance with an externally validated NGS assay and the FOUNDATIONONE® (F1 LDT) assay. See FOUNDATIONONE® CDX™: Technical Information, Foundation Medicine, Inc., available at FoundationMedicine.com, last visited March 25, 2019, which is incorporated by reference herein in its entirety. II.B.1.f. MSK-IMPACTTM [0154] In some aspects, TMB status is assessed using the MSK-IMPACT™ assay. The MSK-IMPACT™ assay uses next-generation sequencing to analyze the mutation status of 468 genes. Target genes are captured and sequenced on an ILLUMINA HISEQ™ instrument. The MSK-IMPACT™ assay is approved by the US FDA for detection of somatic mutations and microsatellite instability in solid malignant neoplasms. The full list of 468 genes analyzed by the MSK-IMPACT™ assay is shown in Table 14. See Evaluation of Automatic Class III Designation for MSK-IMPACT (Integrated Mutation Profiling of Actionable Cancer Targets): Decision Summary, United States Food and Drug Administration, November 15, 2017, available at accessdata.fda.gov. Table 14: Genes analyzed by the MSK-IMPACT™ assay. ABL1 CALR DDR2 FGF19 HIST3H3 LY KX21 PPARG RPTOR STK19 0 U 12 1 2 3 B1 3 7L2 T BR1 BR2 M12 RSS
Figure imgf000052_0001
AIP3 RSF 1 BP1 F2 F7 R F1 1 FA N1 C1 C1L R1 1 C2 1 X3 9
Figure imgf000053_0001
II.B.1.g. NEOGENOMICS® NEOTYPE™ Assays [0155] In some aspects, TMB is determined using a NEOGENOMICS® NEOTYOPE™ assay. In some aspects, the TMB is determined using a NEOTYPE™ Discovery Profile. In some aspects, the TMB is determined using a NEOTYPE Solid Tumor Profile. The NEOGENOMICS assays measure the number of non-synonymous DNA coding sequence changes per megabase of sequenced DNA. II.B.1.h. ONCOMINE™ Tumor Mutation Load Assay [0156] In some aspects, TMB is determined using a THERMOFISHER SCIENTIFIC® ONCOMINE™ Tumor Mutation assay. In some aspects, TMB is determined using a THERMOFISHER SCIENTIFIC® ION TORRENT™ ONCOMINE™ Tumor Mutation assay. The ION TORRENT™ ONCOMINE™ Tumor Mutation assay is a targeted NGS assay that quantitates somatic mutations to determine tumor mutation load. The assay covers 1.7 Mb of DNA. The full list of 408 genes analyzed by the THERMOFISHER SCIENTIFIC® ION TORRENT™ ONCOMINE™ Tumor Mutation assay is shown in Table 15 (see Iontorrent, Oncomine Tumor Mutation Load Assay Flyer, available at assets.thermofisher.com/TFS- Assets/CSD/Flyers/ oncomine-tumor-mutation-load-assay-flyer.pdf, last visited March 25, 2019). Table 15: Genes analyzed by the THERMOFISHER SCIENTIFIC® ION TORRENT™ ONCOMINE™ Tumor Mutation assay.
Figure imgf000054_0001
Figure imgf000055_0001
II.B.1.i. NOVOGENE™ NOVOPM™ Assay [0157] In some aspects, TMB is determined using a NOVOGENE™ NOVOPM™ assay. In some aspects, TMB is determined using a NOVOGENE™ NOVOPM™ Cancer Panel assay. The NOVOGENE™ NOVOPM™ Cancer Panel assay is a comprehensive NGS cancer panel that analyzes the complete coding regions of 548 genes and the introns of 21 genes, representing about 1.5 Mb of DNA, and that are relevant for the diagnosis and/or treatment of solid tumors according to the National Comprehensive Cancer Network (NCCN) guidelines and medical literature. The assay detects SNV, InDel, fusion, and copy number variation (CNV) genomic abnormalities. II.B.1.j. Other TMB Assays [0158] In some aspects, TMB is determined using a TMB assay provided by CARIS® Life Sciences. In some aspects, TMB is determined using the PESONALIS® ACE ImmunoID assay. In some aspects, TMB is determined using the PGDX® CANCERXOME™-R assay. [0159] In yet another particular aspect, the genomic profiling detects all mutation types, i.e., single nucleotide variants, insertions/deletions (indels), copy number variations, and rearrangements, e.g., translocations, expression, and epigenetic markers. [0160] Comprehensive gene panels often contain predetermined genes selected based on the type of tumor to be analyzed. Accordingly, the genomic profile used to measure TMB status can be selected based on the type of tumor the subject has. In one aspect, the genomic profile can include a set of genes particular to a solid tumor. In another aspect, the genomic profile can include a set of genes particular to hematologic malignancies and sarcomas. [0161] In one aspect, the genomic profile comprises one or more genes selected from the group consisting of ABL1, BRAF, CHEK1, FANCC, GATA3, JAK2, MITF, PDCD1LG2, RBM10, STAT4, ABL2, BRCA1, CHEK2, FANCD2, GATA4, JAK3, MLH1, PDGFRA, RET, STK11, ACVR1B, BRCA2, CIC, FANCE, GATA6, JUN, MPL, PDGFRB, RICTOR, SUFU, AKT1, BRD4, CREBBP, FANCF, GID4 (C17orf39), KAT6A (MYST3), MRE11A, PDK1, RNF43, SYK, AKT2, BRIP1, CRKL, FANCG, GLI1, KDM5A, MSH2, PIK3C2B, ROS1, TAF1, AKT3, BTG1, CRLF2, FANCL, GNA11, KDM5C, MSH6, PIK3CA, RPTOR, TBX3, ALK, BTK, CSF1R, FAS, GNA13, KDM6A, MTOR, PIK3CB, RUNX1, TERC, AMER1 (FAM123B), C11orf30 (EMSY), CTCF, FAT1, GNAQ, KDR, MUTYH, PIK3CG, RUNX1T1, TERT (promoter only), APC, CARD11, CTNNA1, FBXW7, GNAS, KEAP1, MYC, PIK3R1, SDHA, TET2, AR, CBFB, CTNNB1, FGF10, GPR124, KEL, MYCL (MYCL1), PIK3R2, SDHB, TGFBR2, ARAF, CBL, CUL3, FGF14, GRIN2A, KIT, MYCN, PLCG2, SDHC, TNFAIP3, ARFRP1, CCND1, CYLD, FGF19, GRM3, KLHL6, MYD88, PMS2, SDHD, TNFRSF14, ARID1A, CCND2, DAXX, FGF23, GSK3B, KMT2A (MLL), NF1, POLD1, SETD2, TOP1, ARID1B, CCND3, DDR2, FGF3, H3F3A, KMT2C (MLL3), NF2, POLE, SF3B1, TOP2A, ARID2, CCNE1, DICER1, FGF4, HGF, KMT2D (MLL2), NFE2L2, PPP2R1A, SLIT2, TP53, ASXL1, CD274, DNMT3A, FGF6, HNF1A, KRAS, NFKBIA, PRDM1, SMAD2, TSC1, ATM, CD79A, DOT1L, FGFR1, HRAS, LMO1, NKX2-1, PREX2, SMAD3, TSC2, ATR, CD79B, EGFR, FGFR2, HSD3B1, LRP1B, NOTCH1, PRKAR1A, SMAD4, TSHR, ATRX, CDC73, EP300, FGFR3, HSP90AA1, LYN, NOTCH2, PRKCI, SMARCA4, U2AF1, AURKA, CDH1, EPHA3, FGFR4, IDH1, LZTR1, NOTCH3, PRKDC, SMARCB1, VEGFA, AURKB, CDK12, EPHA5, FH, IDH2, MAGI2, NPM1, PRSS8, SMO, VHL, AXIN1, CDK4, EPHA7, FLCN, IGF1R, MAP2K1, NRAS, PTCH1, SNCAIP, WISP3, AXL, CDK6, EPHB1, FLT1, IGF2, MAP2K2, NSD1, PTEN, SOCS1, WT1, BAP1, CDK8, ERBB2, FLT3, IKBKE, MAP2K4, NTRK1, PTPN11, SOX10, XPO1, BARD1, CDKN1A, ERBB3, FLT4, IKZF1, MAP3K1, NTRK2, QKI, SOX2, ZBTB2, BCL2, CDKN1B, ERBB4, FOXL2, IL7R, MCL1, NTRK3, RAC1, SOX9, ZNF217, BCL2L1, CDKN2A, ERG, FOXP1, INHBA, MDM2, NUP93, RAD50, SPEN, ZNF703, BCL2L2, CDKN2B, ERRFI1, FRS2, INPP4B, MDM4, PAK3, RAD51, SPOP, BCL6, CDKN2C, ESR1, FUBP1, IRF2, MED12, PALB2, RAF1, SPTA1, BCOR, CEBPA, EZH2, GABRA6, IRF4, MEF2B, PARK2, RANBP2, SRC, BCORL1, CHD2, FAM46C, GATA1, IRS2, MEN1, PAX5, RARA, STAG2, BLM, CHD4, FANCA, GATA2, JAK1, MET, PBRM1, RB1, STAT3, and any combination thereof. In other aspects, the TMB analysis further comprises identifying a genomic alteration in one or more of ETV4, TMPRSS2, ETV5, BCR, ETV1, ETV6, and MYB. [0162] In another aspect, the genomic profile comprises one or more genes selected from the group consisting of ABL1, 12B, ABL2, ACTB, ACVR1, ACVR1B, AGO2, AKT1, AKT2, AKT3, ALK, ALOX, ALOX12B, AMER1, AMER1 (FAM123B or WTX), AMER1 (FAM123B), ANKRD11, APC, APH1A, AR, ARAF, ARFRP1, ARHGAP26 (GRAF), ARID1A, ARID1B, ARID2, ARID5B, ARv7, ASMTL, ASXL1, ASXL2, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, AXIN2, AXL, B2M, BABAM1, BAP1, BARD1, BBC3, BCL10, BCL11B, BCL2, BCL2L1, BCL2L11, BCL2L2, BCL6, BCL7A, BCOR, BCORL1, BIRC3, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRD4, BRIP1, BRIP1 (BACH1), BRSK1, BTG1, BTG2, BTK, BTLA, C11orf 30 (EMSY), C11orf30, C11orf30 (EMSY), CAD, CALR, CARD11, CARM1, CASP8, CBFB, CBL, CCND1, CCND2, CCND3, CCNE1, CCT6B, CD22, CD274, CD274 (PD-L1), CD276, CD36, CD58, CD70, CD79A, CD79B, CDC42, CDC73, CDH1, CDK12, CDK4, CDK6, CDK8, CDKN1A, CDKN1B, CDKN2A, CDKN2Ap14ARF, CDKN2Ap16INK4A, CDKN2B, CDKN2C, CEBPA, CENPA, CHD2, CHD4, CHEK1, CHEK2, CIC, CIITA, CKS1B, CPS1, CREBBP, CRKL, CRLF2, CSDE1, CSF1R, CSF3R, CTCF, CLTA-4, CTNN B1, CTNNA1, CTNNB1, CUL3, CUL4A, CUX1, CXCR4, CYLD, CYP17A1, CYSLTR2, DAXX, DCUN1D1, DDR1, DDR2, DDX3X, DH2, DICER1, DIS3, DNAJB1, DNM2, DNMT1, DNMT3A, DNMT3B, DOT1L, DROSHA, DTX1, DUSP2, DUSP4, DUSP9, E2F3, EBF1, ECT2L, EED, EGFL7, EGFR, EIF1AX, EIF4A2, EIF4E, ELF3, ELP2, EML4, EML4-ALK, EP300, EPAS1, EPCAM, EPHA3, EPHA5, EPHA7, EPHB1, EPHB4, ERBB2, ERBB3, ERBB4, ERCC1, ERCC2, ERCC3, ERCC4, ERCC5, ERF, ERG, ERRFI1, ERRFl1, ESR1, ETS1, ETV1, ETV4, ETV5, ETV6, EWSR1, EXOSC6, EZH1, EZH2, FAF1, FAM175A, FAM46C, FAM58A, FANCA, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCL, FAS, FAS (TNFRSF6), FAT1, FBXO11, FBXO31, FBXW7, FGF1, FGF10, FGF12, FGF14, FGF19, FGF2, FGF23, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, FGFR1, FGFR2, FGFR3, FGFR4, FH, FHIT, FLCN, FLI1, FLT1, FLT3, FLT4, FLYWCH1, FOXA1, FOXL2, FOXO1, FOXO3, FOXP1, FRS2, FUBP1, FYN, GABRA6, GADD45B, GATA1, GATA2, GATA3, GATA4, GATA6, GEN1, GID4 (C17orf 39), GID4 (C17orf39), GLI1, GLl1, GNA11, GNA12, GNA13, GNAQ, GNAS, GPR124, GPS2, GREM1, GRIN2A, GRM3, GSK3B, GTSE1, H3F3A, H3F3B, H3F3C, HDAC1, HDAC4, HDAC7, Hedgehog, HER-2/NEU; ERBB2, HGF, HIST1H1C, HIST1H1D, HIST1H1E, HIST1H2AC, HIST1H2AG, HIST1H2AL, HIST1H2AM, HIST1H2BC, HIST1H2BD, HIST1H2BJ, HIST1H2BK, HIST1H2BO, HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3I, HIST1H3J, HIST2H3C, HIST2H3D, HIST3H3, HLA-A, HLA-B, HNF1A, HOXB13, HRAS, HSD3B1, HSP90AA1, ICK, ICOSLG, ID3, IDH1, IDH2, IFNGR1, IGF1, IGF1R, IGF2, IKBKE, IKZF1, IKZF2, IKZF3, IL10, IL7R, INHA, INHBA, INPP4A, INPP4B, INPP5D (SHIP), INPPL1, INSR, IRF1, IRF2, IRF4, IRF8, IRS1, IRS2, JAK1, JAK2, JAK3, JARID2, JUN, K14, KAT6A (MYST 3), KAT6A (MYST3), KDM2B, KDM4C, KDM5A, KDM5C, KDM6A, KDR, KEAP1, KEL, KIF5B, KIT, KLF4, KLHL6, KMT2A, KMT2A (MLL), KMT2B, KMT2C, KMT2C (MLL3), KMT2D, KMT2D (MLL2), KNSTRN, KRAS, LAMP1, LATS1, LATS2, LEF1, LMO1, LRP1B, LRRK2, LTK, LYN, LZTR1, MAF, MAFB, MAGED1, MAGI2, MALT1, MAP2K1, MAP2K1 (MEK1), MAP2K2, MAP2K2 (MEK2), MAP2K4, MAP3, MAP3K1, MAP3K13, MAP3K14, MAP3K6, MAP3K7, MAPK1, MAPK3, MAPKAP1, MAX, MCL1, MDC1, MDM2, MDM4, MED12, MEF2B, MEF2C, MEK1, MEN1, MERTK, MET, MGA, MIB1, MITF, MKI67, MKNK1, MLH1, MLLT3, MPL, MRE 11A, MRE11A, MSH2, MSH3, MSH6, MSI1, MSI2, MST1, MST1R, MTAP, MTOR, MUTYH, MYC, MYCL, MYCL (MYC L1), MYCL (MYCL1), MYCL1, MYCN, MYD88, MYO18A, MYOD1, NBN, NCOA3, NCOR1, NCOR2, NCSTN, NEGR1, NF1, NF2, NFE2L2, NFKBIA, NKX2-1, NKX3-1, NOD1, NOTCH1, NOTCH2, NOTCH3, NOTCH4, NPM1, NRAS, NRG1, NSD1, NT5C2, NTHL1, NTRK1, NTRK2, NTRK3, NUF2, NUP93, NUP98, P2RY8, PAG1, PAK1, PAK3, PAK7, PALB2, PARK2, PARP1, PARP2, PARP3, PASK, PAX3, PAX5, PAX7, PBRM1, PC, PCBP1, PCLO, PDCD1, PDCD1 (PD-1), PDCD11, PDCD1LG2, PDCD1LG2 (PD-L2), PDGFRA, PDGFRB, PDK1, PDPK1, PGR, PHF6, PHOX2B, PIK3C2B, PIK3C2G, PIK3C3, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PIK3R1, PIK3R2, PIK3R3, PIM1, PLCG2, PLK2, PMAIP1, PMS1, PMS2, PNRC1, POLD1, POLE, POT1, PPARG, PPM1D, PPP2, PPP2R1A, PPP2R2A, PPP4R2, PPP6C, PRDM1, PRDM14, PREX2, PRKAR1A, PRKCI, PRKD1, PRKDC, PRSS8, PTCH1, PTEN, PTP4A1, PTPN11, PTPN2, PTPN6 (SHP-1), PTPRD, PTPRO, PTPRS, PTPRT, QKI, R1A, RAB35, RAC1, RAC2, RAD21, RAD50, RAD51, RAD51B, RAD51C, RAD51D, RAD52, RAD54L, RAF1, RANBP2, RARA, RASA1, RASGEF1A, RB1, RBM10, RECQL, RECQL4, REL, RELN, RET, RFWD2, RHEB, RHOA, RICTOR, RIT1, RNF43, ROS1, RPS6KA4, RPS6KB1, RPS6KB2, RPTOR, RRAGC, RRAS, RRAS2, RTEL1, RUNX1, RUNX1T1, RXRA, RYBP, S1PR2, SDHA, SDHAF2, SDHB, SDHC, SDHD, SERP2, SESN1, SESN2, SESN3, SETBP1, SETD2, SETD8, SF3B1, SGK1, SH2B3, SH2D1A, SHOC2, SHQ1, SLIT2, SLX4, SMAD2, SMAD3, SMAD4, SMARCA1, SMARCA4, SMARCB1, SMARCD1, SMC1A, SMC3, SMO, SMYD3, SNCAIP, SOCS1, SOCS2, SOCS3, SOS1, SOX10, SOX17, SOX2, SOX9, SPEN, SPOP, SPRED1, SPTA1, SRC, SRSF2, STAG2, STAT3, STAT4, STAT5A, STAT5B, STAT6, STK11, STK19, STK40, SUFU, SUZ12, SYK, TAF1, TAP1, TAP2, TBL1XR1, TBX3, TCEB1, TCF3, TCF3 (E2A), TCF7L2, TCL1A (TCL1), TEK, TERC, TERT, TERT Promoter, TET1, TET2, TFRC, TGFBR1, TGFBR2, TIPARP, TLL2, TMEM127, TMEM30A, TMPRSS2, TMSB4XP8 (TMSL3), TNFAIP3, TNFRSF11A, TNFRSF14, TNFRSF17, TOP1, TOP2A, TP53, TP53BP1, TP63, TRAF2, TRAF3, TRAF5, TRAF7, TSC1, TSC2, TSHR, TUSC3, TYK2, TYRO3, U2AF1, U2AF2, UPF1, VEGFA, VHL, VTCN1, WDR90, WHSC1, WHSC1 (MMSET or NSD2), WHSC1L1, WISP3, WT1, WWTR1, XBP1, XIAP, XPO1, XRCC2, YAP1, YES1, YY1AP1, ZBTB2, ZFHX3, ZMYM3, ZNF217, ZNF24 (ZSCAN3), ZNF703, ZRSR2, 0082, SEPT9, 81RC2, 81RC3, 81RC5, 8AI3, 8CL10, 8CL118, 8CL11A, 8CL2, 8CL2L1, 8CL2L2, 8CL3, 8CL6, 8CL9, 8CR, 8LM, 8LNK, 8MPR1A, 8RD3, 8TK, 8U818, A8L2, ACVR2A, ADAMTS2, AFF1, AFF3, AKAP9, ARNT, ATF1, AURK8, AURKC, CASCS, CDH11, CDH2, CDH20, CDH5, CMPK1, COL1A1, CRBN, CREB1, CRTC1, CSMD3, CYP2C19, CYP2D6, DCC, DDIT3, DEK, DPYD, DST, EP400, EXT1, EXT2, FAM123B, FANCJ, FLl1, FN1, FOX01, FOX03, FOXP4, FZR1, G6PD, GDNF, GRM8, HCAR1, HFN1A, HIF1A, HLF, HOOK3, HSP90A81, ICK, IGF2R, IKBKB, IL2, IL21R, IL6ST, ING4, ITGA10, ITGA9, ITGB2, ITGB3, KAT6A, KAT6B, KLF6, KOR, LCK, LIFR, LPHN3, LPP, LRP18, LTF, M8D1, MAF8, MAGEA1, MAGl1, MAML2, MAPK8, MARK1, MARK4, MLL, MLL2, MLL3, MLLT10, MMP2, MN1, MTC, MTOT, MTR, MTRR, MUC1, MY8, MYH11, MYH9, NCOA1, NCOA2, NCOA4, NFK81, NFK82, NIN, NLRP1, NUMA1, NUP214, P8RM1, P8X1, PAX?, PAX3, PAX8, PAXS, PDE4DIP, PDGF8, PER1, PGAP3, PHOX28, PIK3C28, PKHD1, PLAG1, PLCG1, PLEKHGS, PML, POU5F1, PSIP1, PTGS2, RADSO, RALGDS, RHOH, RNASEL, RNF2, RNF213, RPS6KA2, RRM1, SAMD9, SBDS, SMUG1, SOHO, SOX11, SSX1, STK36, SYNE1, T8X22, TAF1L, TAL1, TCF12, TCF7L1, TFE3, TGF8R2, TGM7, TH8S1, TIMP3, TLR4, TLX1, TNK2, TPR, TRIM24, TRIM33, TRIP11, TRRAP, U8R5, UGT1A1, USP9X, WAS, WRN, XP01, XPA, XPC, ZNF384, ZNF521, and any combination thereof. [0163] In another aspect, the genomic profiling assay comprises at least about 20, at least about 30, at least about 40, at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, at least about 100, at least about 110, at least about 120, at least about 130, at least about 140, at least about 150, at least about 160, at least about 170, at least about 180, at least about 190, at least about 200, at least about 210, at least about 220, at least about 230, at least about 240, at least about 250, at least about 260, at least about 270, at least about 280, at least about 290, or at least about 300 genes selected from the group consisting of ABL1, 12B, ABL2, ACTB, ACVR1, ACVR1B, AGO2, AKT1, AKT2, AKT3, ALK, ALOX, ALOX12B, AMER1, AMER1 (FAM123B or WTX), AMER1 (FAM123B), ANKRD11, APC, APH1A, AR, ARAF, ARFRP1, ARHGAP26 (GRAF), ARID1A, ARID1B, ARID2, ARID5B, ARv7, ASMTL, ASXL1, ASXL2, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, AXIN2, AXL, B2M, BABAM1, BAP1, BARD1, BBC3, BCL10, BCL11B, BCL2, BCL2L1, BCL2L11, BCL2L2, BCL6, BCL7A, BCOR, BCORL1, BIRC3, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRD4, BRIP1, BRIP1 (BACH1), BRSK1, BTG1, BTG2, BTK, BTLA, C11orf 30 (EMSY), C11orf30, C11orf30 (EMSY), CAD, CALR, CARD11, CARM1, CASP8, CBFB, CBL, CCND1, CCND2, CCND3, CCNE1, CCT6B, CD22, CD274, CD274 (PD-L1), CD276, CD36, CD58, CD70, CD79A, CD79B, CDC42, CDC73, CDH1, CDK12, CDK4, CDK6, CDK8, CDKN1A, CDKN1B, CDKN2A, CDKN2Ap14ARF, CDKN2Ap16INK4A, CDKN2B, CDKN2C, CEBPA, CENPA, CHD2, CHD4, CHEK1, CHEK2, CIC, CIITA, CKS1B, CPS1, CREBBP, CRKL, CRLF2, CSDE1, CSF1R, CSF3R, CTCF, CLTA-4, CTNN B1, CTNNA1, CTNNB1, CUL3, CUL4A, CUX1, CXCR4, CYLD, CYP17A1, CYSLTR2, DAXX, DCUN1D1, DDR1, DDR2, DDX3X, DH2, DICER1, DIS3, DNAJB1, DNM2, DNMT1, DNMT3A, DNMT3B, DOT1L, DROSHA, DTX1, DUSP2, DUSP4, DUSP9, E2F3, EBF1, ECT2L, EED, EGFL7, EGFR, EIF1AX, EIF4A2, EIF4E, ELF3, ELP2, EML4, EML4-ALK, EP300, EPAS1, EPCAM, EPHA3, EPHA5, EPHA7, EPHB1, EPHB4, ERBB2, ERBB3, ERBB4, ERCC1, ERCC2, ERCC3, ERCC4, ERCC5, ERF, ERG, ERRFI1, ERRFl1, ESR1, ETS1, ETV1, ETV4, ETV5, ETV6, EWSR1, EXOSC6, EZH1, EZH2, FAF1, FAM175A, FAM46C, FAM58A, FANCA, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCI, FANCL, FAS, FAS (TNFRSF6), FAT1, FBXO11, FBXO31, FBXW7, FGF1, FGF10, FGF12, FGF14, FGF19, FGF2, FGF23, FGF3, FGF4, FGF5, FGF6, FGF7, FGF8, FGF9, FGFR1, FGFR2, FGFR3, FGFR4, FH, FHIT, FLCN, FLI1, FLT1, FLT3, FLT4, FLYWCH1, FOXA1, FOXL2, FOXO1, FOXO3, FOXP1, FRS2, FUBP1, FYN, GABRA6, GADD45B, GATA1, GATA2, GATA3, GATA4, GATA6, GEN1, GID4 (C17orf 39), GID4 (C17orf39), GLI1, GLl1, GNA11, GNA12, GNA13, GNAQ, GNAS, GPR124, GPS2, GREM1, GRIN2A, GRM3, GSK3B, GTSE1, H3F3A, H3F3B, H3F3C, HDAC1, HDAC4, HDAC7, Hedgehog, HER-2/NEU; ERBB2, HGF, HIST1H1C, HIST1H1D, HIST1H1E, HIST1H2AC, HIST1H2AG, HIST1H2AL, HIST1H2AM, HIST1H2BC, HIST1H2BD, HIST1H2BJ, HIST1H2BK, HIST1H2BO, HIST1H3A, HIST1H3B, HIST1H3C, HIST1H3D, HIST1H3E, HIST1H3F, HIST1H3G, HIST1H3H, HIST1H3I, HIST1H3J, HIST2H3C, HIST2H3D, HIST3H3, HLA-A, HLA-B, HNF1A, HOXB13, HRAS, HSD3B1, HSP90AA1, ICK, ICOSLG, ID3, IDH1, IDH2, IFNGR1, IGF1, IGF1R, IGF2, IKBKE, IKZF1, IKZF2, IKZF3, IL10, IL7R, INHA, INHBA, INPP4A, INPP4B, INPP5D (SHIP), INPPL1, INSR, IRF1, IRF2, IRF4, IRF8, IRS1, IRS2, JAK1, JAK2, JAK3, JARID2, JUN, K14, KAT6A (MYST 3), KAT6A (MYST3), KDM2B, KDM4C, KDM5A, KDM5C, KDM6A, KDR, KEAP1, KEL, KIF5B, KIT, KLF4, KLHL6, KMT2A, KMT2A (MLL), KMT2B, KMT2C, KMT2C (MLL3), KMT2D, KMT2D (MLL2), KNSTRN, KRAS, LAMP1, LATS1, LATS2, LEF1, LMO1, LRP1B, LRRK2, LTK, LYN, LZTR1, MAF, MAFB, MAGED1, MAGI2, MALT1, MAP2K1, MAP2K1 (MEK1), MAP2K2, MAP2K2 (MEK2), MAP2K4, MAP3, MAP3K1, MAP3K13, MAP3K14, MAP3K6, MAP3K7, MAPK1, MAPK3, MAPKAP1, MAX, MCL1, MDC1, MDM2, MDM4, MED12, MEF2B, MEF2C, MEK1, MEN1, MERTK, MET, MGA, MIB1, MITF, MKI67, MKNK1, MLH1, MLLT3, MPL, MRE 11A, MRE11A, MSH2, MSH3, MSH6, MSI1, MSI2, MST1, MST1R, MTAP, MTOR, MUTYH, MYC, MYCL, MYCL (MYC L1), MYCL (MYCL1), MYCL1, MYCN, MYD88, MYO18A, MYOD1, NBN, NCOA3, NCOR1, NCOR2, NCSTN, NEGR1, NF1, NF2, NFE2L2, NFKBIA, NKX2-1, NKX3-1, NOD1, NOTCH1, NOTCH2, NOTCH3, NOTCH4, NPM1, NRAS, NRG1, NSD1, NT5C2, NTHL1, NTRK1, NTRK2, NTRK3, NUF2, NUP93, NUP98, P2RY8, PAG1, PAK1, PAK3, PAK7, PALB2, PARK2, PARP1, PARP2, PARP3, PASK, PAX3, PAX5, PAX7, PBRM1, PC, PCBP1, PCLO, PDCD1, PDCD1 (PD-1), PDCD11, PDCD1LG2, PDCD1LG2 (PD-L2), PDGFRA, PDGFRB, PDK1, PDPK1, PGR, PHF6, PHOX2B, PIK3C2B, PIK3C2G, PIK3C3, PIK3CA, PIK3CB, PIK3CD, PIK3CG, PIK3R1, PIK3R2, PIK3R3, PIM1, PLCG2, PLK2, PMAIP1, PMS1, PMS2, PNRC1, POLD1, POLE, POT1, PPARG, PPM1D, PPP2, PPP2R1A, PPP2R2A, PPP4R2, PPP6C, PRDM1, PRDM14, PREX2, PRKAR1A, PRKCI, PRKD1, PRKDC, PRSS8, PTCH1, PTEN, PTP4A1, PTPN11, PTPN2, PTPN6 (SHP-1), PTPRD, PTPRO, PTPRS, PTPRT, QKI, R1A, RAB35, RAC1, RAC2, RAD21, RAD50, RAD51, RAD51B, RAD51C, RAD51D, RAD52, RAD54L, RAF1, RANBP2, RARA, RASA1, RASGEF1A, RB1, RBM10, RECQL, RECQL4, REL, RELN, RET, RFWD2, RHEB, RHOA, RICTOR, RIT1, RNF43, ROS1, RPS6KA4, RPS6KB1, RPS6KB2, RPTOR, RRAGC, RRAS, RRAS2, RTEL1, RUNX1, RUNX1T1, RXRA, RYBP, S1PR2, SDHA, SDHAF2, SDHB, SDHC, SDHD, SERP2, SESN1, SESN2, SESN3, SETBP1, SETD2, SETD8, SF3B1, SGK1, SH2B3, SH2D1A, SHOC2, SHQ1, SLIT2, SLX4, SMAD2, SMAD3, SMAD4, SMARCA1, SMARCA4, SMARCB1, SMARCD1, SMC1A, SMC3, SMO, SMYD3, SNCAIP, SOCS1, SOCS2, SOCS3, SOS1, SOX10, SOX17, SOX2, SOX9, SPEN, SPOP, SPRED1, SPTA1, SRC, SRSF2, STAG2, STAT3, STAT4, STAT5A, STAT5B, STAT6, STK11, STK19, STK40, SUFU, SUZ12, SYK, TAF1, TAP1, TAP2, TBL1XR1, TBX3, TCEB1, TCF3, TCF3 (E2A), TCF7L2, TCL1A (TCL1), TEK, TERC, TERT, TERT Promoter, TET1, TET2, TFRC, TGFBR1, TGFBR2, TIPARP, TLL2, TMEM127, TMEM30A, TMPRSS2, TMSB4XP8 (TMSL3), TNFAIP3, TNFRSF11A, TNFRSF14, TNFRSF17, TOP1, TOP2A, TP53, TP53BP1, TP63, TRAF2, TRAF3, TRAF5, TRAF7, TSC1, TSC2, TSHR, TUSC3, TYK2, TYRO3, U2AF1, U2AF2, UPF1, VEGFA, VHL, VTCN1, WDR90, WHSC1, WHSC1 (MMSET or NSD2), WHSC1L1, WISP3, WT1, WWTR1, XBP1, XIAP, XPO1, XRCC2, YAP1, YES1, YY1AP1, ZBTB2, ZFHX3, ZMYM3, ZNF217, ZNF24 (ZSCAN3), ZNF703, ZRSR2, 0082, SEPT9, 81RC2, 81RC3, 81RC5, 8AI3, 8CL10, 8CL118, 8CL11A, 8CL2, 8CL2L1, 8CL2L2, 8CL3, 8CL6, 8CL9, 8CR, 8LM, 8LNK, 8MPR1A, 8RD3, 8TK, 8U818, A8L2, ACVR2A, ADAMTS2, AFF1, AFF3, AKAP9, ARNT, ATF1, AURK8, AURKC, CASCS, CDH11, CDH2, CDH20, CDH5, CMPK1, COL1A1, CRBN, CREB1, CRTC1, CSMD3, CYP2C19, CYP2D6, DCC, DDIT3, DEK, DPYD, DST, EP400, EXT1, EXT2, FAM123B, FANCJ, FLl1, FN1, FOX01, FOX03, FOXP4, FZR1, G6PD, GDNF, GRM8, HCAR1, HFN1A, HIF1A, HLF, HOOK3, HSP90A81, ICK, IGF2R, IKBKB, IL2, IL21R, IL6ST, ING4, ITGA10, ITGA9, ITGB2, ITGB3, KAT6A, KAT6B, KLF6, KOR, LCK, LIFR, LPHN3, LPP, LRP18, LTF, M8D1, MAF8, MAGEA1, MAGl1, MAML2, MAPK8, MARK1, MARK4, MLL, MLL2, MLL3, MLLT10, MMP2, MN1, MTC, MTOT, MTR, MTRR, MUC1, MY8, MYH11, MYH9, NCOA1, NCOA2, NCOA4, NFK81, NFK82, NIN, NLRP1, NUMA1, NUP214, P8RM1, P8X1, PAX?, PAX3, PAX8, PAXS, PDE4DIP, PDGF8, PER1, PGAP3, PHOX28, PIK3C28, PKHD1, PLAG1, PLCG1, PLEKHGS, PML, POU5F1, PSIP1, PTGS2, RADSO, RALGDS, RHOH, RNASEL, RNF2, RNF213, RPS6KA2, RRM1, SAMD9, SBDS, SMUG1, SOHO, SOX11, SSX1, STK36, SYNE1, T8X22, TAF1L, TAL1, TCF12, TCF7L1, TFE3, TGF8R2, TGM7, TH8S1, TIMP3, TLR4, TLX1, TNK2, TPR, TRIM24, TRIM33, TRIP11, TRRAP, U8R5, UGT1A1, USP9X, WAS, WRN, XP01, XPA, XPC, ZNF384, ZNF521, and any combination thereof. [0164] In another aspect, the genomic profile comprises one or more genes selected from the genes listed in Tables 2-15. II.B.2. TMB Status [0165] In one aspect, TMB status based on genomic profiling is highly correlated with TMB status based on whole-exome or whole-genome sequencing. Evidence provided herein shows that the use of genomic profiling assays, such as the F1CDx assay, have concordance with whole-exome and/or whole genome sequencing assays. These data support the use of genomic profiling assays as a more efficient means of measuring TMB status, without forfeiting the prognostic qualities of TMB status. [0166] TMB can be measured using a tissue biopsy sample or, alternatively, circulating tumor DNA (ctDNA), cfDNA (cell-free DNA), and/or a liquid biopsy sample. ctDNA can be used to measure TMB status according to whole-exome or whole-genome sequencing or genomic profiling using available methodologies, e.g., GRAIL, Inc. [0167] In some aspects, a subject is identified as suitable for an anti-PD-1 therapy, as disclosed herein, based on the measurement of TMB status and identification of a high TMB. In some aspects, a TMB score is calculated as the total number of nonsynonymous missense mutations in a tumor, as measured by whole exome sequencing or whole genome sequencing. In one aspect, the high TMB has a score of at least 210, at least 215, at least 220, at least 225, at least 230, at least 235, at least 240, at least 245, at least 250, at least 255, at least 260, at least 265, at least 270, at least 275, at least 280, at least 285, at least 290, at least 295, at least 300, at least 305, at least 310, at least 315, at least 320, at least 325, at least 330, at least 335, at least 340, at least 345, at least 350, at least 355, at least 360, at least 365, at least 370, at least 375, at least 380, at least 385, at least 390, at least 395, at least 400, at least 405, at least 410, at least 415, at least 420, at least 425, at least 430, at least 435, at least 440, at least 445, at least 450, at least 455, at least 460, at least 465, at least 470, at least 475, at least 480, at least 485, at least 490, at least 495, or at least 500. In another aspect, the high TMB has a score of at least 215, at least 220, at least 221, at least 222, at least 223, at least 224, at least 225, at least 226, at least 227, at least 228, at least 229, at least 230, at least 231, at least 232, at least 233, at least 234, at least 235, at least 236, at least 237, at least 238, at least 239, at least 240, at least 241, at least 242, at least 243, at least 244, at least 245, at least 246, at least 247, at least 248, at least 249, or at least 250. In a particular aspect, the high TMB has a score of at least 243. In other aspects, the high TMB has a score of at least 244. In some aspects, the high TMB has a score of at least 245. In other aspects, the high TMB has a score of at least 246. In other aspects, the high TMB has a score of at least 247. In other aspects, the high TMB has a score of at least 248. In other aspects, the high TMB has a score of at least 249. In other aspects, the high TMB has a score of at least 250. In other aspects, the high TMB has a score of any integer between 200 and 300 or higher. In other aspects, the high TMB has a score of any integer between 210 and 290 or higher. In other aspects, the high TMB has a score of any integer between 220 and 280 or higher. In other aspects, the high TMB has a score of any integer between 230 and 270 or higher. In other aspects, the high TMB has a score of any integer between 235 and 265 or higher. [0168] Alternatively, the high TMB can be a relative value rather than an absolute value. In some aspects, the subject’s TMB status is compared to a reference TMB value. In one aspect, the subject’s TMB status is within the highest fractile of the reference TMB value. In another aspect, the subject’s TMB status is within the top tertile of the reference TMB value. [0169] In some aspects, TMB status is expressed as the number of mutations per sample, per cell, per exome, or per length of DNA (e.g., Mb). In some aspects, a tumor has a high TMB status if the tumor has at least about 50 mutations/tumor, at least about 55 mutations/tumor, at least about 60 mutations/tumor, at least about 65 mutations/tumor, at least about 70 mutations/tumor, at least about 75 mutations/tumor, at least about 80 mutations/tumor, at least about 85 mutations/tumor, at least about 90 mutations/tumor, at least about 95 mutations/tumor, at least about 100 mutations/tumor, at least about 105 mutations/tumor, at least about 110 mutations/tumor, at least about 115 mutations/tumor, or at least about 120 mutations/tumor. In some aspects, a tumor has a high TMB status if the tumor has at least about 125 mutations/tumor, at least about 150 mutations/tumor, at least about 175 mutations/tumor, at least about 200 mutations/tumor, at least about 225 mutations/tumor, at least about 250 mutations/tumor, at least about 275 mutations/tumor, at least about 300 mutations/tumor, at least about 350 mutations/tumor, at least about 400 mutations/tumor, or at least about 500 mutations/tumor. In one particular aspect, a tumor has a high TMB status if the tumor has at least about 100 mutations/tumor. [0170] In some aspects, a tumor has a high TMB status if the tumor has at least about 5 mutations per megabase of genes, e.g., genome sequenced according to a TMB assay, e.g., genome sequenced according to a FOUNDATIONONE® CDX™ assay, (mutations/Mb), at least about 6 mutations/Mb, at least about 7 mutations/Mb, at least about 8 mutations/Mb, at least about 9 mutations/Mb, at least about 10 mutations/Mb, at least about 11 mutations/Mb, at least about 12 mutations/Mb, at least about 13 mutations/Mb, at least about 14 mutations/Mb, at least about 15 mutations/Mb, at least about 20 mutations/Mb, at least about 25 mutations/Mb, at least about 30 mutations/Mb, at least about 35 mutations/Mb, at least about 40 mutations/Mb, at least about 45 mutations/Mb, at least about 50 mutations/Mb, at least about 75 mutations/Mb, or at least about 100 mutations/Mb. In certain aspects, a tumor has a high TMB status if the tumor has at least about 5 mutations/Mb. In certain aspects, a tumor has a high TMB status if the tumor has at least about 10 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 11 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 12 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 13 mutations/Mb. In some aspects, a tumor has a high TMB status if the tumor has at least about 14 mutations/Mb. In certain aspects, a tumor has a high TMB status if the tumor has at least about 15 mutations/Mb. [0171] Because the number of mutations varies by tumor type and other ways (see Q4 and Q5), the values associated with "TMB high" and "TMB low" can differ across tumor types. II.C. PD-L1 Expression [0172] Certain aspects of the present disclosure are directed to methods of measuring tumor PD-L1 expression in a sample obtained from a subject. "PD-L1 positive" as used herein can be interchangeably used with "PD-L1 expression of at least about 1%." In one embodiment, the PD-L1 expression can be used by any methods known in the art. In another embodiment, the PD-L1 expression is measured by an automated IHC. PD-L1 positive tumors can thus have at least about 1%, at least about 2%, at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, 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 about 100% of the tumor cells expressing PD-L1 as measured by an automated IHC. In certain embodiments, "PD-L1 positive" means that there are at least 100 cells that express PD-L1 on the surface of the cells. [0173] In order to assess the PD-L1 expression expression, in one embodiment, a test tissue sample can be obtained from the patient who is in need of the therapy. In another embodiment, the assessment of PD-L1 expression can be achieved without obtaining a test tissue sample. In some embodiments, selecting a suitable patient includes (i) optionally providing a test tissue sample obtained from a patient with cancer of the tissue, the test tissue sample comprising tumor cells and/or tumor-infiltrating inflammatory cells; and (ii) assessing the proportion of cells in the test tissue sample that express PD-L1 on the surface of the cells based on an assessment that the proportion of cells in the test tissue sample that express PD-L1 on the cell surface is higher than a predetermined threshold level. [0174] In any of the methods comprising the measurement of PD-L1 expression in a test tissue sample, however, it should be understood that the step comprising the provision of a test tissue sample obtained from a patient is an optional step. It should also be understood that in certain embodiments the "measuring" or "assessing" step to identify, or determine the number or proportion of, cells in the test tissue sample that express PD-L1 (e.g., the expression of PD-L1 on the cell surface) is performed by a transformative method of assaying for PD-L1 expression, for example by performing a reverse transcriptase-polymerase chain reaction (RT-PCR) assay or an IHC assay. In certain other embodiments, no transformative step is involved and PD-L1 expression is assessed by, for example, reviewing a report of test results from a laboratory. In certain embodiments, the steps of the methods up to, and including, assessing PD-L1 expression provides an intermediate result that may be provided to a physician or other healthcare provider for use in selecting a suitable candidate for the anti-PD-1 antibody or anti-PD-L1 antibody therapy. In certain embodiments, the steps that provide the intermediate result is performed by a medical practitioner or someone acting under the direction of a medical practitioner. In other embodiments, these steps are performed by an independent laboratory or by an independent person such as a laboratory technician. [0175] In certain embodiments of any of the present methods, the proportion of cells that express PD-L1 is assessed by performing an assay to determine the presence of PD-L1 RNA. In further embodiments, the presence of PD-L1 RNA is determined by RT-PCR, in situ hybridization or RNase protection. In other embodiments, the proportion of cells that express PD-L1 is assessed by performing an assay to determine the presence of PD-L1 polypeptide. In further embodiments, the presence of PD-L1 polypeptide is determined by immunohistochemistry (IHC), enzyme-linked immunosorbent assay (ELISA), in vivo imaging, or flow cytometry. In some embodiments, PD-L1 expression is assayed by IHC. In other embodiments of all of these methods, cell surface expression of PD-L1 is assayed using, e.g., IHC or in vivo imaging. [0176] Imaging techniques have provided important tools in cancer research and treatment. Recent developments in molecular imaging systems, including positron emission tomography (PET), single-photon emission computed tomography (SPECT), fluorescence reflectance imaging (FRI), fluorescence-mediated tomography (FMT), bioluminescence imaging (BLI), laser-scanning confocal microscopy (LSCM), and multiphoton microscopy (MPM) will likely herald even greater use of these techniques in cancer research. Some of these molecular imaging systems allow clinicians to not only see where a tumor is located in the body, but also to visualize the expression and activity of specific molecules, cells, and biological processes that influence tumor behavior and/or responsiveness to therapeutic drugs (Condeelis and Weissleder, "In vivo imaging in cancer," Cold Spring Harb. Perspect. Biol.2(12):a003848 (2010)). Antibody specificity, coupled with the sensitivity and resolution of PET, makes immunoPET imaging particularly attractive for monitoring and assaying expression of antigens in tissue samples (McCabe and Wu, "Positive progress in immunoPET—not just a coincidence," Cancer Biother. Radiopharm. 25(3):253-61 (2010); Olafsen et al., "ImmunoPET imaging of B-cell lymphoma using 124I-anti-CD20 scFv dimers (diabodies)," Protein Eng. Des. Sel. 23(4):243-9 (2010)). In certain embodiments of any of the present methods, PD-L1 expression is assayed by immunoPET imaging. In certain embodiments of any of the present methods, the proportion of cells in a test tissue sample that express PD-L1 is assessed by performing an assay to determine the presence of PD-L1 polypeptide on the surface of cells in the test tissue sample. In certain embodiments, the test tissue sample is a FFPE tissue sample. In other embodiments, the presence of PD-L1 polypeptide is determined by IHC assay. In further embodiments, the IHC assay is performed using an automated process. In some embodiments, the IHC assay is performed using an anti-PD- L1 monoclonal antibody to bind to the PD-L1 polypeptide. [0177] In one embodiment of the present methods, an automated IHC method is used to assay the expression of PD-L1 on the surface of cells in FFPE tissue specimens. This disclosure provides methods for detecting the presence of human PD-L1 antigen in a test tissue sample, or quantifying the level of human PD-L1 antigen or the proportion of cells in the sample that express the antigen, which methods comprise contacting the test sample, and a negative control sample, with a monoclonal antibody that specifically binds to human PD- L1, under conditions that allow for formation of a complex between the antibody or portion thereof and human PD-L1. In certain embodiments, the test and control tissue samples are FFPE samples. The formation of a complex is then detected, wherein a difference in complex formation between the test sample and the negative control sample is indicative of the presence of human PD-L1 antigen in the sample. Various methods are used to quantify PD-L1 expression. [0178] In a particular embodiment, the automated IHC method comprises: (a) deparaffinizing and rehydrating mounted tissue sections in an autostainer; (b) retrieving antigen using a decloaking chamber and pH 6 buffer, heated to 110°C for 10 min; (c) setting up reagents on an autostainer; and (d) running the autostainer to include steps of neutralizing endogenous peroxidase in the tissue specimen; blocking non-specific protein-binding sites on the slides; incubating the slides with primary antibody; incubating with a postprimary blocking agent; incubating with NovoLink Polymer; adding a chromogen substrate and developing; and counterstaining with hematoxylin. [0179] For assessing PD-L1 expression in tumor tissue samples, a pathologist examines the number of membrane PD-L1+ + tumor cells in each field under a microscope and mentally estimates the percentage of cells that are positive, then averages them to come to the final percentage. The different staining intensities are defined as 0/negative, l+/weak, 2+/moderate, and 3+/strong. Typically, percentage values are first assigned to the 0 and 3+ buckets, and then the intermediate 1+ and 2+ intensities are considered. For highly heterogeneous tissues, the specimen is divided into zones, and each zone is scored separately and then combined into a single set of percentage values. The percentages of negative and positive cells for the different staining intensities are determined from each area and a median value is given to each zone. A final percentage value is given to the tissue for each staining intensity category: negative, 1+, 2+, and 3+. The sum of all staining intensities needs to be 100%. In one embodiment, the threshold number of cells that needs to be PD-L1 positive is at least about 100, at least about 125, at least about 150, at least about 175, or at least about 200 cells. In certain embodiments, the threshold number of cells that need to be PD-L1 positive is at least about 100 cells. [0180] Staining is also assessed in tumor-infiltrating inflammatory cells such as macrophages and lymphocytes. In most cases macrophages serve as an internal positive control since staining is observed in a large proportion of macrophages. While not required to stain with 3+ intensity, an absence of staining of macrophages should be taken into account to rule out any technical failure. Macrophages and lymphocytes are assessed for plasma membrane staining and only recorded for all samples as being positive or negative for each cell category. Staining is also characterized according to an outside/inside tumor immune cell designation. "Inside" means the immune cell is within the tumor tissue and/or on the boundaries of the tumor region without being physically intercalated among the tumor cells. "Outside" means that there is no physical association with the tumor, the immune cells being found in the periphery associated with connective or any associated adjacent tissue. [0181] In certain embodiments of these scoring methods, the samples are scored by two pathologists operating independently, and the scores are subsequently consolidated. In certain other embodiments, the identification of positive and negative cells is scored using appropriate software. [0182] A histoscore (also described as H-score) is used as a more quantitative measure of the IHC data. The histoscore is calculated as follows: Histoscore = [(% tumor x 1 (low intensity)) + (% tumor x 2 (medium intensity)) + (% tumor x 3 (high intensity)] [0183] To determine the histoscore, the pathologist estimates the percentage of stained cells in each intensity category within a specimen. Because expression of most biomarkers is heterogeneous the histoscore is a truer representation of the overall expression. The final histoscore range is 0 (no expression) to 300 (maximum expression). [0184] An alternative means of quantifying PD-L1 expression in a test tissue sample IHC is to determine the adjusted inflammation score (AIS) score defined as the density of inflammation multiplied by the percent PD-L1 expression by tumor-infiltrating inflammatory cells (Taube et al., "Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape," Sci. Transl. Med.4(127):127ra37 (2012)). II.D. Checkpoint Inhibitors [0185] The present disclosure is directed to methods for treating a human subject afflicted with a cancer comprising administering to the subject a checkpoint inhibitor. Any checkpoint inhibitor known in the art can be used in the methods disclosed herein. In some aspects, the checkpoint inhibitor is any reagent that blocks, inhibits, or reduces the activity of one or more checkpoint protein. In some aspects, the checkpoint protein is selected from the group selected from the group consisting of PD-1, CTLA-4, LAG3, TIGIT, TIM3, NKG2a, OX40, ICOS, CD137, KIR, TGFβ, IL-10, IL-8, IL-2, CD96, VISTA, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, and any combination thereof. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of PD-1. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CTLA-4. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of LAG3. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of TIGIT. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of TIM3. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of NKG2a. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of OX40. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of ICOS. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CD137. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of KIR. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of TGFβ. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of IL-10. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of IL-8. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of IL-2. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CD96. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of VISTA. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of B7-H4. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of Fas ligand. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CXCR4. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of mesothelin. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of CD27. In some aspects, the checkpoint inhibitor blocks, inhibits, or reduces the activity of GITR. [0186] Any inhibitor can be used in the methods disclosed herein. In some aspects, the inhibitor is a small molecule. In some aspects, the inhibitor is a protein. In some aspects, the inhibitor is an antibody or an antigen-binding portion thereof. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds PD-1. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CTLA-4. In some aspects, the checkpoint inhibitor is an antibody or antigen- binding portion thereof that specifically binds LAG3. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds TIGIT. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds TIM3. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds NKG2a. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds OX40. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds ICOS. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CD137. In some aspects, the checkpoint inhibitor is an antibody or antigen- binding portion thereof that specifically binds KIR. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds TGFβ. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds IL- 10. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds IL-8. In some aspects, the checkpoint inhibitor is an antibody or antigen- binding portion thereof that specifically binds IL-2. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CD96. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds VISTA. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds B7-H4. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds Fas ligand. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CXCR4. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds mesothelin. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds CD27. In some aspects, the checkpoint inhibitor is an antibody or antigen-binding portion thereof that specifically binds GITR. [0187] In some aspects, the subject is administered a monotherapy, e.g., an anti-PD-1 monotherapy, e.g., wherein the subject is not administered one or more additional anti-cancer agent. In some aspects, the subject is administered a combination therapy, e.g., wherein the subject is administered a first checkpoint inhibitor, e.g., an anti-PD-1 antibody, and one or more additional anti-cancer agents. 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- LAG-3 antibody. [0188] In other aspects of the present disclosure, an anti-PD-L1 antibody is substituted for the anti-PD-1 antibody. In certain aspects, the methods comprise administering an anti-PD-L1 antibody to a subject. In some aspects, the subject is administered an anti-PD-L1 monotherapy. In some aspects, the subject is administered a combination therapy comprising an anti-PD-L1 antibody and a second anti-cancer agent, e.g., an anti-CTLA-4 antibody. In some aspects, the subject is administered a combination therapy comprising an anti-PD-L1 antibody and a second anti-cancer agent, e.g., an anti-LAG-3 antibody. [0189] In some aspects, the antibody is a chimeric, humanized or human monoclonal antibody or a portion thereof. II.D.1. Anti-PD-1 Antibodies Useful for the Disclosure [0190] 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-γ 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 antigen- specific 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. [0191] 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. [0192] 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. 10:136 (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. 10:136 (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. 10:136 (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 10(2):183-203 (2018), and IBI308 (Innovent; see WO 2017/024465, WO 2017/025016, WO 2017/132825, and WO 2017/133540). [0193] 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). [0194] 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. [0195] 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). [0196] 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. [0197] 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. [0198] 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 antigen- binding 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 cross- competes with nivolumab for binding to human PD-1. [0199] 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. [0200] 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. [0201] 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. [0202] 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. [0203] 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. II.D.2. Anti-PD-L1 Antibodies Useful for the Disclosure [0204] In certain aspects, an anti-PD-L1 antibody is substituted for the anti-PD-1 antibody in any of the methods disclosed herein. Anti-PD-L1 antibodies that are known in the art can be used in the compositions and methods of the present disclosure. Examples of anti-PD-L1 antibodies useful in the compositions and methods of the present disclosure include the antibodies disclosed in US Patent No. 9,580,507. Anti-PD-L1 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-γ 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-L1 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. [0205] In certain aspects, the anti-PD-L1 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):TPS3113 (2018)), and CK-301 (Checkpoint Therapeutics; see Gorelik et al., AACR:Abstract 4606 (Apr 2016)). [0206] In certain aspects, the PD-L1 antibody is atezolizumab (TECENTRIQ®). Atezolizumab is a fully humanized IgG1 monoclonal anti-PD-L1 antibody. [0207] In certain aspects, the PD-L1 antibody is durvalumab (IMFINZI™). Durvalumab is a human IgG1 kappa monoclonal anti-PD-L1 antibody. [0208] In certain aspects, the PD-L1 antibody is avelumab (BAVENCIO®). Avelumab is a human IgG1 lambda monoclonal anti-PD-L1 antibody. [0209] Anti-PD-L1 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-L1 antibody disclosed herein, e.g., atezolizumab, durvalumab, and/or avelumab. In some aspects, the anti-PD-L1 antibody binds the same epitope as any of the anti-PD-L1 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 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., 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). [0210] 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. [0211] Anti-PD-L1 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. [0212] Anti-PD-L1 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-L1 "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-L1 antibody or antigen-binding portion thereof cross-competes with atezolizumab, durvalumab, and/or avelumab for binding to human PD-L1. [0213] The anti-PD-L1 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-L1 antibody is durvalumab. In other aspects, the anti-PD-L1 antibody is avelumab. In some aspects, the anti-PD-L1 antibody is atezolizumab. [0214] In some aspects, the anti-PD-L1 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. [0215] In some aspects, the anti-PD-L1 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-L1 antibody is administered at a dose of about 10 mg/kg body weight at about once every 2 weeks. [0216] In other aspects, the anti-PD-L1 antibody useful for the present disclosure is a flat dose. In some aspects, the anti-PD-L1 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-L1 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-L1 antibody is administered as a flat dose of about 1200 mg at about once every 3 weeks. In other aspects, the anti-PD-L1 antibody is administered as a flat dose of about 800 mg at about once every 2 weeks. In other aspects, the anti-PD-L1 antibody is administered as a flat dose of about 840 mg at about once every 2 weeks. [0217] 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. [0218] In some aspects, avelumab is administered as a flat dose of about 800 mg once about every 2 weeks. [0219] 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. II.D.3. Anti-CTLA-4 Antibodies [0220] 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. [0221] 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 (ka) of at least about 103, about 104, or about 105 m-1 s-1; (c) a kinetic disassociation constant (kd) 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. [0222] 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. [0223] In particular aspects, the CTLA-4 antibody is ipilimumab for use in the compositions and methods disclosed herein. Ipilimumab is a fully human, IgG1 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. [0224] In particular aspects, the CTLA-4 antibody is tremelimumab. [0225] In particular aspects, the CTLA-4 antibody is MK-1308. [0226] In particular aspects, the CTLA-4 antibody is AGEN-1884. [0227] 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 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., 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 cross- compete 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). [0228] 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 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. [0229] 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. [0230] 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 antigen- binding 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 antigen- binding portion thereof cross-competes with ipilimumab and/or tremelimumab for binding to human CTLA-4. [0231] 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 antigen- binding portion thereof is administered at a dose of 1 mg/kg body weight once every 6 weeks. [0232] 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 antigen- binding 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. [0233] 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.D.4. Anti-LAG-3 Antibodies [0234] As used herein a LAG-3 antagonist includes, but is not limited to, LAG-3 binding agents, e.g., a LAG-3 antibody, and soluble LAG-3 polypeptides, e.g., a fusion protein comprising the extracellular portion of LAG-3. [0235] In some aspects, the LAG-3 inhibitor is a soluble LAG-3 polypeptide, for example, a LAG-3-Fc fusion polypeptide capable of binding to MHC Class II. [0236] In some aspects, the LAG-3 antagonist comprises IMP321 (eftilagimod alpha). [0237] In some aspects, the LAG-3 antagonist is an anti-LAG-3 antibody or an antigen binding fragment thereof that specifically binds to LAG-3 ("anti-LAG-3 antibody"). [0238] Anti-LAG-3 antibodies (or VH/VL domains derived therefrom) suitable for use herein can be generated using methods well known in the art. Alternatively, art recognized anti- LAG-3 antibodies can be used. [0239] In some aspects, the anti-LAG-3 antibody is a chimeric, humanized, or human monoclonal antibody, or a portion thereof. In other aspects, the anti-LAG-3 antibody is a bispecific antibody or a multispecific antibody. [0240] In some aspects, the anti-LAG-3 antibody is relatlimab, e.g., BMS-986016 as described in PCT/US13/48999, the teachings of which are hereby incorporated by reference. [0241] In other aspects, the antibody has the heavy and light chain CDRs or variable regions of relatlimab. Accordingly, in one aspect, the antibody comprises CDR1, CDR2, and CDR3 domains of the VH region of relatlimab, and CDR1, CDR2 and CDR3 domains of the VL region of relatlimab. In another aspect, the antibody comprises VH and/or VL regions of relatlimab. [0242] In some aspects, the anti-LAG-3 antibody cross-competes with relatlimab for binding to human LAG-3. The ability of antibodies to cross-compete for binding to an antigen indicates that the 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., relatlimab, by virtue of their binding to the same epitope region. Cross- competing antibodies can be readily identified based on their ability to cross-compete in standard binding assays such as Biacore analysis, ELISA assays or flow cytometry (see, e.g., WO 2013/173223). [0243] In some aspects, the anti-LAG-3 antibody binds to the same epitope as relatlimab. [0244] In some aspects, the anti-LAG-3 antibody is a biosimilar of relatlimab. [0245] In some aspects, the anti- LAG-3 antibody is LAG-525, MK-4280, REGN3767, TSR-033, TSR-075, Sym022, FS-118, or any combination thereof. [0246] In some aspects, art recognized anti-LAG-3 antibodies can be used in the therapeutic methods of the disclosure. For example, the anti-human LAG-3 antibody described in US2011/0150892 A1, which is herein incorporated by reference, and referred to as monoclonal antibody 25F7 (also known as "25F7" and "LAG-3.1) can be used. Other art recognized anti- LAG-3 antibodies that can be used include IMP731 (H5L7BW) described in US 2011/007023, MK-4280 (28G-10) described in WO2016028672, REGN3767 described in Journal for ImmunoTherapy of Cancer, (2016) Vol.4, Supp. Supplement 1 Abstract Number: P195, BAP050 described in WO2017/019894, IMP-701 (LAG-525), aLAG3(0414), aLAG3(0416), Sym022, TSR-033, TSR-075, XmAb22841, MGD013, BI754111, FS118, P 13B02-30, AVA-017 and GSK2831781. These and other anti-LAG-3 antibodies useful in the claimed invention can be found in, for example: US 10,188,730, WO2016/028672, WO2017/106129, WO2017/062888, WO2009/044273, WO2018/069500, WO2016/126858, WO2014/179664, WO2016/200782, WO2015/200119, WO2017/019846, WO2017/198741, WO2017/220555, WO2017/220569, WO2018/071500, WO2017/015560, WO2017/025498, WO2017/087589, WO2017/087901, WO2018/083087, WO2017/149143, WO2017/219995, US2017/0260271, WO2017/086367, WO2017/086419, WO2018/034227, WO18/185046, WO18/185043, WO2018/217940, WO19/011306, WO2018/208868, and WO2014/140180. The contents of each of these references are incorporated by reference herein in their entirety. Antibodies that compete with any of the art-recognized antibodies for binding to LAG-3 also can be used. [0247] In some aspects, the anti-LAG-3antibody cross-competes with, binds to the same epitope as, or is a biosimilar of an anti-LAG-3 antibody that is described herein or that is known in the art. [0248] In certain aspects, an anti-LAG-3 antibody is used to determine LAG-3 expression. In some aspects, an anti-LAG-3 antibody is selected for its ability to bind to LAG-3 in formalin-fixed, paraffin-embedded (FFPE) tissue specimens. In other aspects, an anti-LAG-3 antibody is capable of binding to LAG-3 in frozen tissues. In further aspects, an anti-LAG-3 antibody is capable of distinguishing membrane bound, cytoplasmic, and/or soluble forms of LAG-3. [0249] In some aspects, an anti-LAG-3 antibody useful for assaying, detecting, and/or quantifying LAG-3 expression in accordance with the methods described herein is the 17B4 mouse IgG1 anti-human LAG-3 monoclonal antibody, or an antigen binding fragment thereof. See, e.g., J. Matsuzaki, et al.; PNAS 107, 7875 (2010). [0250] In some aspects, the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a dose ranging from about 0.1 mg/kg to about 10.0 mg/kg body weight once about every 1, 2, 3, 4, 5, 6, 7, or 8 weeks. In some aspects, the anti-LAG-3 antibody or antigen- binding portion thereof is administered at a dose of at least about 1 mg/kg, at least about 2 mg/kg, at least about 3 mg/kg, at least about 4 mg/kg, at least about 5 mg/kg, at least about 6 mg/kg, at least about 7 mg/kg, at least about 8 mg/kg, at least about 9 mg/kg, or at least about 10 mg/kg body weight about once every 1, 2, 3, 4, 5, 6, 7, or 8 weeks. [0251] In some aspects, the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose. In some aspects, the anti-LAG-3 antibody is administered at a flat dose of from about 20 mg to about 2000 mg. In one aspect, the anti-LAG-3 antibody or antigen- binding portion thereof is administered as a flat dose of at least about 80 mg or at least about 160 mg. In another aspect, the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose once about every 1, 2, 3, 4, 5, 6, 7, or 8 weeks. [0252] In some aspects, the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose. [0253] In some aspects, the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose of about 80 mg. [0254] In some aspects, the anti-LAG-3 antibody or antigen-binding portion thereof is administered at a flat dose of about 160 mg. II.D.5. Combination Therapies [0255] In certain aspects, the anti-PD-1 antibody, the anti-PD-L1 antibody, the anti-LAG- 3 antibody, and/or the anti-CTLA-4 antibody are administered at a therapeutically effective amount. In some aspects, the method comprises administering a therapeutically effective amount of anti-PD-1 antibody and an anti-CTLA-4 antibody. In some aspects, the method comprises administering a therapeutically effective amount of anti-PD-1 antibody and an anti-LAG-3 antibody. In other aspects, the method comprises administering a therapeutically effective amount of anti-PD-L1 antibody and an anti-CTLA-4 antibody. In other aspects, the method comprises administering a therapeutically effective amount of anti-PD-L1 antibody and an anti- LAG-3 antibody. Any anti-PD-1, anti-PD-L1, anti-LAG-3 antibody, or anti-CTLA-4 antibody disclosed herein can be used in the method. In certain aspects, the anti-PD-1 antibody comprises nivolumab. In some aspects, the anti-PD-1 antibody comprises pembrolizumab. In some aspects, the anti-PD-L1 antibody comprises atezolizumab. In some aspects, the anti-PD-L1 antibody comprises durvalumab. In some aspects, the anti-PD-L1 antibody comprises avelumab. In some aspects, the anti-CTLA-4 antibody comprises ipilimumab. In some aspects, the anti-CTLA-4 antibody comprises ipilimumab tremelimumab. [0256] In some aspects, the (a) anti-PD-1 antibody or the anti-PD-L1 antibody and the (b) anti-CTLA-4 antibody are each administered once about every 2 weeks, once about every 3 weeks, once about every 4 weeks, once about every 5 weeks, or once about every 6 weeks. In some aspects, the anti-PD-1 antibody or the anti-PD-L1 antibody is administered once about every 2 weeks, once about every 3 weeks or once about every 4 weeks, and the anti-CTLA-4 antibody is administered once about every 6 weeks. In some aspects, the anti-PD-1 antibody or anti-PD-L1 antibody is administered on the same day as the anti-CTLA-4 antibody. In some aspects, the anti-PD-1 antibody or the anti-PD-L1 antibody is administered on a different day than the anti-CTLA-4 antibody. [0257] In some aspects, the anti-CTLA-4 antibody is administered at a dose ranging from about 0.1 mg/kg to about 20.0 mg/kg body weight once about every 2, 3, 4, 5, 6, 7, or 8 weeks. In some aspects, the anti-CTLA-4 antibody is administered at a dose of about 0.1 mg/kg, about 0.3 mg/kg, about 0.6 mg/kg, about 0.9 mg/kg, about 1 mg/kg, about 3 mg/kg, about 6 mg/kg, about 9 mg/kg, about 10 mg/kg, about 12 mg/kg, about 15 mg/kg, about 18 mg/kg, or about 20 mg/kg. In certain aspects, the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 4 weeks. In some aspects, the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0258] In some aspects, the anti-CTLA-4 antibody is administered at a flat dose. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose ranging from at least about 40 mg to at least about 1600 mg. In some aspects, the anti-CTLA-4 antibody is administered at a flat dose of at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least about 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg, at least about 180 mg, at least about 190 mg, or at least about 200 mg. In some aspects, the CTLA-4 antibody is administered at a flat dose of at least about 220 mg, at least about 230 mg, at least about 240 mg, at least about 250 mg, at least about 260 mg, at least about 270 mg, at least about 280 mg, at least about 290 mg, at least about 300 mg, at least about 320 mg, at least about 360 mg, at least about 400 mg, at least about 440 mg, at least about 480 mg, at least about 520 mg, at least about 560 mg, or at least about 600 mg. In some aspects, the CTLA-4 antibody is administered at a flat dose of at least about 640 mg, at least about 720 mg, at least about 800 mg, at least about 880 mg, at least about 960 mg, at least about 1040 mg, at least about 1120 mg, at least about 1200 mg, at least about 1280 mg, at least about 1360 mg, at least about 1440 mg, or at least about 1600 mg. In some aspects, the anti-CTLA-4 antibody is administered in a flat dose at least once about every 2, 3, 4, 5, 6, 7, or 8 weeks. [0259] In certain aspects, the anti-PD-1 antibody is administered at a dose of about 2 mg/kg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a dose of about 3 mg/kg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti- PD-1 antibody is administered at a dose of about 6 mg/kg once about every 4 weeks and the anti- CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0260] In certain aspects, the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 240 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 480 mg once about every 4 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0261] In certain aspects, the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a flat dose of about 80 mg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 200 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 240 mg once about every 2 weeks and the anti- CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks. In some aspects, the anti-PD-1 antibody is administered at a flat dose of about 480 mg once about every 4 weeks and the anti-CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks. [0262] In certain aspects, the anti-PD-L1 antibody is administered at a dose of about 10 mg/kg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-L1 antibody is administered at a dose of about 15 mg/kg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0263] In certain aspects, the anti-PD-L1 antibody is administered at a flat dose of about 800 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. In some aspects, the anti-PD-L1 antibody is administered at a flat dose of about 1200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg once about every 6 weeks. [0264] In certain aspects, the anti-PD-L1 antibody is administered at a flat dose of about 800 mg once about every 2 weeks and the anti-CTLA-4 antibody is administered at a flat dose of about 80 mg once about every 6 weeks. In some aspects, the anti-PD-L1 antibody is administered at a flat dose of about 1200 mg once about every 3 weeks and the anti-CTLA-4 antibody is administered at a dose of about 80 mg once about every 6 weeks. [0265] In some aspects, the anti-PD-1 antibody, e.g., nivolumab, is administered at a dose of about 3 mg/kg and the anti-CTLA-4 antibody is administered at a dose of about 1 mg/kg on the same day, once about every 3 weeks for 4 doses, then the anti-PD-1 antibody, e.g., nivolumab, is administered at a flat dose of 240 mg once about every 2 weeks or 480 mg once about every 4 weeks. In some aspects, the anti-PD-1 antibody, e.g., nivolumab, is administered at a dose of about 1 mg/kg and the anti-CTLA-4 antibody is administered at a dose of about 3 mg/kg on the same day, once about every 3 weeks for 4 doses, then the anti-PD-1 antibody, e.g., nivolumab, is administered at a flat dose of 240 mg once about every 2 weeks or 480 mg once about every 4 weeks. II.D.6. Additional Anticancer Therapies [0266] In some aspects of the present disclosure, the methods disclosed herein further comprise administering an anti-PD-1 antibody (or an anti-PD-L1 antibody) and an additional anticancer therapy. In certain aspects, the method comprising administering an anti-PD-1 antibody (or an anti-PD-L1 antibody), an anti-CTLA-4 antibody, and an additional anticancer therapy The additional anticancer 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 additional anticancer therapy comprises a surgery, a radiation therapy, a chemotherapy, an immunotherapy, or any combination thereof. In some aspects, the additional anticancer therapy comprises a chemotherapy, including any chemotherapy disclosed herein. In some aspect, the additional anticancer therapy comprises an immunotherapy. In some aspects, the additional anticancer therapy comprises administration of an antibody or antigen-binding portion thereof that specifically binds TIGIT, TIM3, NKG2a, OX40, ICOS, MICA, CD137, KIR, TGFβ, IL-10, IL-8, B7-H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, or any combination thereof. II.E. Tumors [0267] 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, kidney cancer, head and neck cancer, colon cancer, 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 hepatocellular cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 gastroesophageal cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 melanoma, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 bladder cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 lung cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 kidney cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. 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 head and neck cancer, wherein the tumor has a high inflammatory signature score, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. In certain aspects, the tumor is derived from a colon 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, and wherein the tumor has a TMB status of at least about 10 mutations per megabase of genes examined. [0268] In certain aspects, the subject has received one, two, three, four, five or more prior cancer treatments. In other aspects, the subject is treatment-naïve. 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. [0269] 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-L1 antibody therapy. [0270] 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 platinum- based 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. [0271] In some aspects, the at least one prior therapy is selected from a therapy comprising administration of an anticancer 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. [0272] 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. II.F. Pharmaceutical Compositions and Dosages [0273] 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 co- formulation of an antibody with recombinant human hyaluronidase enzyme (rHuPH20), which removes traditional limitations on the volume of biologics 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, anti- oxidant, 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. [0274] In some aspects, the method comprises administering an anti-PD-1 antibody (or an anti-PD-L1 antibody) and an anti-CTLA-4 antibody, wherein the anti-PD-1 antibody (or the anti- PD-L1 antibody) is administered in a fixed dose with the anti-CTLA-4 antibody in a single composition. In some aspects, the anti-PD-1 antibody is administered in a fixed dose with the anti-CTLA-4 antibody. In some aspects, the anti-PD-L1 antibody is administered in a fixed dose with the anti-CTLA-4 antibody in a single composition. In some aspects, the ratio of the anti-PD- 1 antibody (or the anti-PD-L1 antibody) to the anti-CTLA-4 antibody is at least about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, about 1:10, about 1:15, about 1:20, about 1:30, about 1:40, about 1:50, about 1:60, about 1:70, about 1:80, about 1:90, about 1:100, about 1:120, about 1:140, about 1:160, about 1:180, about 1:200, about 200:1, about 180:1, about 160:1, about 140:1, about 120:1, about 100:1, about 90:1, about 80:1, about 70:1, about 60:1, about 50:1, about 40:1, about 30:1, about 20:1, about 15:1, about 10:1, about 9:1, about 8:1, about 7:1, about 6:1, about 5:1, about 4:1, about 3:1, or about 2:1. [0275] 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 toxicities reported in other trials of checkpoint inhibitors plus anti-angiogenic therapy (see, e.g., Johnson et al., 2013; Rini et al., 2011) support the selection of a nivolumab dose lower than 10 mg/kg. [0276] Treatment is continued as long as clinical benefit is observed or until unacceptable toxicity or disease progression occurs. Nevertheless, in certain aspects, the dosages of the anti- PD-1 antibody, the anti-PD-L1 antibody, and/or the anti-CTLA-4 antibody administered are significantly lower than the approved dosage, i.e., a subtherapeutic dosage, of the agent. The anti- PD-1 antibody, the anti-PD-L1 antibody, and/or the anti-CTLA-4 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. [0277] 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. [0278] 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 [0279] Also within the scope of the present disclosure are kits comprising (a) one or more checkpoint inhibitors described elsewhere herein, e.g., an anti-PD-1 antibody or an anti-PD-L1 antibody for therapeutic uses, wherein the kits are suitable for the subjects identified as described herein. 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 therapeutically effective dosage of a checkpoint inhibitor, e.g., 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-L1 antibody; and (b) instructions for using the anti-PD-1 antibody or the anti-PD-L1 antibody in the methods disclosed herein. This disclosure further provides a kit for treating a subject identified as a suitable candidate for a checkpoint inhibitor therapy as described herein, the kit comprising: (a) a therapeutically effective dosage of a checkpoint inhibitor, e.g., 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-L1 antibody; and (b) instructions for using the checkpoint inhibitor, e.g., the anti- PD-1 antibody or the anti-PD-L1 antibody, in the methods disclosed herein. In some aspects, this disclosure provides a kit for treating a subject identified as a suitable candidate for a checkpoint inhibitor therapy as described herein, the kit comprising: (a) a therapeutically effective dosage of a checkpoint inhibitor, e.g., 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-L1 antibody; and (b) instructions for using the checkpoint inhibitor, e.g., the anti-PD-1 antibody or the anti-PD-L1 antibody, in the methods disclosed herein. [0280] 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. [0281] 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. [0282] In some aspects, the kit further includes an inflammatory gene panel assay disclosed herein. In some aspects, the kit further includes instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having a high inflammatory signature score, according to the methods disclosed herein. In other aspects, the kit further includes an anti-CTLA-4 antibody and instructions to administer (a) the anti-PD-1 antibody or the anti-PD-L1 antibody and (b) the anti-CTLA-4 antibody to a subject identified as having a high inflammatory signature score, according to the methods disclosed herein. [0283] In some aspects, the kit further includes a comprehensive genomic profiling assay disclosed herein. In some aspects, the kit includes a FOUNDATIONONE® CDX™ genomic profiling assay. In some aspects, the kit further includes instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein. In other aspects, the kit further includes an anti-CTLA-4 antibody and instructions to administer (a) the anti-PD-1 antibody or the anti-PD-L1 antibody and (b) the anti- CTLA-4 antibody to a subject identified as having a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein. [0284] In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a comprehensive genomic profiling assay disclosed herein, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a high inflammatory signature score and (b) a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein. In other aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an anti-CTLA-4 antibody, (c) an inflammatory gene panel assay disclosed herein, (d) a comprehensive genomic profiling assay disclosed herein, and (e) instructions to administer (a) the anti-PD-1 antibody or the anti-PD-L1 antibody and (b) the anti-CTLA-4 antibody to a subject identified as having (a) a high inflammatory signature score and (b) a high TMB status, e.g., a TMB status of at least about 10 mutations/Mb of genome sequenced, according to the methods disclosed herein. [0285] In some aspects, the kit further comprises an anti-PD-L1 antibody. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) positive tumor PD-L1 expression, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 1%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD- L1 expression of at least about 5%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 10%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 15%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 20%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti- PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD- L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 25%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 30%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 35%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 40%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti- PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD- L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 45%, as disclosed herein. In some aspects, the kit comprises (a) an anti-PD-1 antibody or an anti-PD-L1 antibody, (b) an inflammatory gene panel assay disclosed herein, (c) a diagnostic anti-PD-L1 antibody, and (d) instructions to administer the anti-PD-1 antibody or the anti-PD-L1 antibody to a subject identified as having (a) a low signature score and (b) tumor PD-L1 expression of at least about 50%, as disclosed herein. IV. Devices [0286] Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, (b) a PD-L1 expression pattern in a biological sample, and (c) a tumor mutation burden (TMB) status of a biological sample. Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, and (b) a PD-L1 expression pattern in a biological sample. Certain aspects of the present disclosure are directed to a device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, and (b) a tumor mutation burden (TMB) status of a biological sample. [0287] In some aspects, the device further comprises a means for detecting a level of expression of the transcript. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample comprises a first biological probe, wherein the first biological probe specifically interacts with a transcript of at least one gene in the panel of genes. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe, a third biological probe, a fourth biological probe, a fifth biological probe, a sixth biological probe, a seventh biological probe, an eight biological probe, a ninth biological probe, or a tenth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a third biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a fourth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a fifth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a sixth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a seventh biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises an eight biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises a ninth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample further comprises or a tenth biological probe. In some aspects, the means for detecting a level of expression of a panel of genes in the biological sample comprises more than ten biological probes. [0288] In some aspects, the first biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a first gene of the at least 5 genes. In some aspects, the second biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a second gene of the at least 5 genes. In some aspects, the third biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a third gene of the at least 5 genes. In some aspects, the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes. In some aspects, the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes. In some aspects, the fifth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fifth gene of the at least 5 genes. [0289] In some aspects, the transcript comprises an mRNA. In some aspects, the transcript comprises a cDNA. [0290] All of the references cited above, as well as all references cited herein, are incorporated herein by reference in their entireties. [0291] The following examples are offered by way of illustration and not by way of limitation. EXAMPLES Example 1 [0292] Infiltration of CD8+ T cells is a marker of inflammation in the tumor microenvironment (TME) and has been associated with clinical benefit from immune oncology (I-O) therapy in patients with cancer. Programmed death ligand 1 (PD-L1) expression, determined using approved immunohistochemistry (IHC) assays, is associated with response to I- O therapy. Methodological standardization and heterogeneous PD-L1 expression present challenges for assessment of PD-L1 expression by IHC. [0293] The predictive value of tumor mutational burden (TMB) as a biomarker for response to I-O therapy is ongoing across multiple tumor types. However, clinical validation may be hindered by assay variation, tissue heterogeneity, and variation across different tumor types. Accordingly, additional biomarkers are needed to improve selection of patients more likely to respond to I-O therapy. [0294] Simultaneous evaluation of multiple RNA transcripts by gene expression profiling (GEP) has previously been employed to derive gene expression signatures as potential biomarkers of response in patients receiving I-O therapy across multiple tumor types. [0295] A parenchymal inflammatory signature score (parenchymal signature) that correlates with CD8 expression by IHC can accurately assess inflammation in the TME. The parenchymal signature has been analytically validated using an investigational use only (IUO) GEP-based inflammation panel across multiple tumor types, including urothelial carcinoma (UC). [0296] The association of the parenchymal phenotype signature with clinical responses to nivolumab, using tumor tissue samples from patients with UC enrolled in a phase 2 clinical trial was retrospectively investigated. This was completed by comparing the performance of the parenchymal signature with other potential biomarkers of response (PD-L1 expression and TMB) in predicting clinical response to nivolumab in patients with UC. Methods [0297] Patients [0298] Two hundred seventy patients with platinum-resistant metastatic UC and evaluable tumor PD-L1 expression were treated with nivolumab therapy as part of the phase 2 clinical trial (NCT02387996) trial, as described in Sharma P, et al. Lancet Oncol 2017;18:312– 322. The minimum follow-up time for the current analysis was about three years, with responses determined by blinded, independent review committee assessments. Objective response (OR) was measured according to Response Evaluation Criteria in Solid Tumors (RECIST) v1.1. The associations of biomarkers with responses were evaluated by comparing patients with complete or partial response (CR + PR) with patients with stable, progressive, or nonevaluable disease (SD + PD + NE). [0299] Biomarker analysis [0300] Gene expression profiling was performed using the IUO 95-gene inflammation panel to derive the parenchymal signature scores. CD8 immunohistochemistry (IHC) was performed using an automated commercial proprietary assay (Dako mouse clone C8/144B; Agilent Technologies Co). Expression of PD-L1 was measured on tumor cells using the Dako PD-L1 IHC 28-8 pharmDx assay. Tumor mutational burden (TMB) was determined by whole exome sequencing (WES), and performed as previously described (Galsky M, et al. Ann Oncol 2017;28 (suppl 5), Abstract 848PD). The TMB was defined as the total number of missense somatic mutations per tumor. [0301] Statistical analysis [0302] For the statistical analysis, Pearson’s correlation was used to compare parenchymal signature scores with CD8 IHC. Cox proportional-hazards regression was used to evaluate the dependence of progression-free survival (PFS) or overall survival (OS) on parenchymal signature, PDL1, and TMB scores. Logistic regression was used to assess the dependence of OR on parenchymal signature, PDL1, and TMB scores. [0303] The statistical models included linear effects of each biomarker and the multiplicative interaction between them. Likelihood-ratio tests (two-sided) were used to test overall biomarker and interaction effects. No formal correction for multiple hypothesis testing was done. Kaplan–Meier plots based on categorization of the biomarker scores were used to illustrate associations with PFS or OS. All data analyses were performed with R 3.4.1 for Linux. Results [0304] As shown in Table 16, below, PD-L1 was evaluable in 270 patients with UC, GEP was evaluable in 205 patients (76%), and GEP + WES was evaluable in 113 patients (42%). Baseline characteristics, OR, PFS, and OS were similar between all treated patients and the biomarker cohorts. Table 16:
Figure imgf000099_0002
[0305] Additionally, higher median parenchymal signature scores were observed in patients with CR + PR than in patients with SD + PD + NE (P < 0.01) (FIG. 1). In patients with high parenchymal signature scores, longer PFS (P < 0.01) and OS (P < 0.001) were observed (FIGs. 2A and 2B). The median survival of PFS and OS are provided in Tables 17 and 18, below. Table 17: Median Survival – PFS
Figure imgf000099_0001
Table 18: Median Survival – OS
Figure imgf000100_0001
[0306] In patients evaluable for GEP with advanced UC (n = 203), CD8 signature scores correlated with CD8 IHC (r = 0.77). The parenchymal signature did not appear to correlate with TMB and tumor cell PD-L1 expression. Poor correlation (r = 0.31) was observed between parenchymal signature scores and tumor cell PD-L1 expression in the GEP cohort (FIG. 3A). Poor correlation (r = 0.01) was also observed between parenchymal signature and TMB scores in the GEP + WES cohort (FIG. 3B). However, strong association between parenchymal signature score and PFS (P < 0.05) and OS (P < 0.01) was maintained regardless of PD-L1 expression levels (FIGs.4A-4B). The strong association between parenchymal signature score and PFS (P = 0.05) and OS (P < 0.01) was maintained regardless of TMB levels (FIGs. 5A-5E). The TMB cutoffs are shown in FIG.5C. [0307] This example demonstrates that the parenchymal signature was associated with clinical responses to I-O treatment in patients with advanced UC. As shown, parenchymal signature scores did not correlate with other potential biomarkers of response (PD-L1 expression and TMB), and associations of the parenchymal signature with survival were independent of PD- L1 expression and TMB. [0308] These data suggest that the parenchymal signature can be used alone and/or in combination with other relevant biomarkers such as PD-L1 expression or TMB to identify patients more likely to benefit from I-O therapies.

Claims

WHAT IS CLAIMED IS: 1. A method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.
2. A method of treating a tumor in a human subject in need thereof, comprising: (a) determining whether the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined; and (b) administering an effective amount of a checkpoint inhibitor to the subject.
3. A method of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy, comprising any two or more of the following: (i) determining whether the subject has a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) detecting a level of PD-L1 expression in tumor cells of the tumor; and (iii) measuring a TMB status of a biological sample obtained from the subject.
4. The method of claim 3, further comprising administering to the subject a checkpoint inhibitor if the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression, and (iii) the subject exhibits a TMB status of at least about 10 mutations per megabase of genes examined.
5. A checkpoint inhibitor for treating a tumor in a human subject in need thereof, wherein the subject meets at least two of the following conditions: (i) the subject is identified as having a parenchymal inflammatory phenotype, wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject; (ii) at least 1% of tumor cells in the tumor are positive for PD-L1 expression; and (iii) the subject exhibits a tumor mutation burden (TMB) status of at least about 10 mutations per megabase of genes examined.
6. A combination of assays for identifying a tumor in a human subject in need thereof, wherein the combination of assays comprises at least two of the following: (i) the subject is to be determined for having a parenchymal inflammatory phenotype; wherein the parenchymal inflammatory phenotype is determined by measuring the expression of a panel of genes comprising at least 5 genes in a sample obtained from the subject (ii) a level of PD-L1 expression in tumor cells of the tumor is to be detected; and (iii) the subject is to be measured for a TMB status of a biological sample obtained from the subject.
7. The method of any one or claims 1 to 4, the checkpoint inhibitor of claim 5, or the combination of assays of claim 6, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA-4, Pan- KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA- DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3.
8. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least about 5 additional genes, at least about 10 additional genes, at least about 15 additional genes, at least about 20 additional genes, at least about 25 additional genes, at least about 30 additional genes, at least about 35 additional genes, at least about 40 additional genes, at least about 45 additional genes, at least about 50 additional genes, at least about 55 additional genes, at least about 60 additional genes, at least about 65 additional genes, at least about 70 additional genes, at least about 75 additional genes, at least about 80 additional genes, at least about 85 additional genes, at least about 90 additional genes, at least about 95 additional genes, at least about 100 additional genes, at least about 110 additional genes, at least about 120 additional genes, at least about 130 additional genes, at least about 140 additional genes, at least about 150 additional genes, at least about 160 additional genes, at least about 170 additional genes, at least about 180 additional genes, at least about 190 additional genes, or at least about 200 additional genes.
9. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein at least one of the additional genes is selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA- DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS- L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof.
10. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least about 63 genes.
11. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least one housekeeping gene.
12. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 housekeeping genes.
13. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the housekeeping genes are selected from the group consisting of ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof.
14. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least one control gene.
15. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 control genes.
16. The method, checkpoint inhibitor, or the combination of assays of claim 14 or 15, wherein the control genes are selected from the group consisting of ANT1, 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.
17. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises at least about 95 genes.
18. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3.
19. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ.
20. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin - 9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA- DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR- Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ.
21. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ.
22. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein a parenchymal inflammatory phenotype is characterized by a low signature score, wherein the signature score is determined by measuring the composite expression of the genes in the gene panel in a tumor sample form the subject, and comparing the composite expression of the genes in the gene panel to the average composite expression of the panel of genes in tumor samples obtained from a population of subjects afflicted with the tumor.
23. The method, checkpoint inhibitor, or the combination of assays of claim 22, wherein the average composite signature score is determined by measuring the composite expression of the panel of genes in tumor samples obtained from the population of subjects.
24. The method, checkpoint inhibitor, or the combination of assays of of 22 or 21, wherein the low signature score is characterized by a signature score of less than about 50, less than about 45, less than about 40, less than about 35, less than about 30, less than about 25, less than about 20, less than about 15, less than about 10, or less than about 5; wherein a signature score of 100 indicates that the subject has no parenchymal inflammatory phenotype; and wherein a signature score of 1 indicates that a subject has a high parenchymal phenotype.
25. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the TMB status is determined by sequencing nucleic acids in the tumor and identifying a genomic alteration in the sequenced nucleic acids.
26. The method, checkpoint inhibitor, or the combination of assays of claim 25, wherein the genomic alteration comprises one or more somatic mutations.
27. The method, checkpoint inhibitor, or the combination of assays of claim 25 or 26, wherein the genomic alteration comprises one or more nonsynonymous mutations.
28. The method, checkpoint inhibitor, or the combination of assays of any one of claims 25 to 27, wherein the genomic alteration comprises one or more missense mutations.
29. The method, checkpoint inhibitor, or the combination of assays of any one of claims 25 to 28, wherein the genomic alteration comprises one or more alterations selected from the group consisting of a base pair substitution, a base pair insertion, a base pair deletion, a copy number alteration (CNAs), a gene rearrangement, and any combination thereof.
30. The method, checkpoint inhibitor, or the combination of assays of any of the preceding claims, wherein the TMB status of the tumor comprises at least 10 mutations, at least about 11 mutations, at least about 12 mutations, at least about 13 mutations, at least about 14 mutations, at least about 15 mutations, at least about 16 mutations, at least about 17 mutations, at least about 18 mutations, at least about 19 mutations, at least about 20 mutations, at least about 21 mutations, at least about 22 mutations, at least about 23 mutations, at least about 24 mutations, at least about 25 mutations, at least about 26 mutations, at least about 27 mutations, at least about 28 mutations, at least about 29 mutations, or at least about 30 mutations per megabase of genome examined as measured by a FOUNDATIONONE® CDX™ assay.
31. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein PD-L1 expression is determined using an IHC assay.
32. The method, checkpoint inhibitor, or the combination of assays of of claim 31, wherein the IHC assay is an automated IHC assay.
33. The method, checkpoint inhibitor, or the combination of assays of any one of claims 22 to 32, wherein the tumor sample is a tumor tissue biopsy.
34. The method, checkpoint inhibitor, or the combination of assays of any one of claims 23 to 33, wherein the tumor sample is a formalin-fixed, paraffin-embedded tumor tissue or a fresh-frozen tumor tissue.
35. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the expression of the genes in the panel of genes is determined by measuring the level of mRNA, the level of a protein encoded by the gene, or both.
36. The method, checkpoint inhibitor, or the combination of assays of claim 35, wherein the level of mRNA is determined using reverse transcriptase PCR.
37. The method, checkpoint inhibitor, or the combination of assays of claim 35 or 36, wherein the level of the protein encoded by the gene is determined using an IHC assay.
38. The method, checkpoint inhibitor, or the combination of assays of claim 37, wherein the IHC assay is an automated IHC assay.
39. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the checkpoint inhibitor comprises a polypeptide, a small molecule, a polynucleotide, or any combination thereof.
40. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the checkpoint inhibitor comprises an antibody or an antigen-binding fragment thereof.
41. The method, checkpoint inhibitor, or the combination of assays of claim 40, wherein the antibody or antigen-binding fragment thereof specifically binds a target selected from PD-1 ("anti-PD-1 antibody"), PD-L1 ("anti-PD-L1 antibody"), CTLA-4, LAG3, TIGIT, TIM3, NKG2a, OX40, ICOS, CD137, KIR, TGFβ, IL-10, IL-8, IL-2, CD96, VISTA, B7- H4, Fas ligand, CXCR4, mesothelin, CD27, GITR, and any combination thereof.
42. The method, checkpoint inhibitor, or the combination of assays of claim 40 or 41, wherein the antibody is a chimeric, humanized or human monoclonal antibody or a portion thereof.
43. The method, checkpoint inhibitor, or the combination of assays of claim 41 or 42, wherein the anti-PD-1 antibody cross-competes with nivolumab for binding to human PD-1.
44. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 43, wherein the anti-PD-1 antibody binds to the same epitope as nivolumab.
45. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 44, wherein the anti-PD-1 antibody comprises a heavy chain constant region which is of a human IgG1 or IgG4 isotype.
46. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 45, wherein the anti-PD-1 antibody is nivolumab.
47. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 45, wherein the anti-PD-1 antibody is pembrolizumab.
48. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 47, wherein the anti-PD-1 antibody is administered at a dose ranging from at least about 0.1 mg/kg to at least about 10.0 mg/kg body weight once about every 1, 2 or 3 weeks.
49. The method, checkpoint inhibitor, or the combination of assays of claim 48, wherein the anti-PD-1 antibody is administered at a dose of at least about 3 mg/kg body weight once about every 2 weeks.
50. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 49, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose.
51. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 47 and 50, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of at least about 200, at least about 220, at least about 240, at least about 260, at least about 280, at least about 300, at least about 320, at least about 340, at least about 360, at least about 380, at least about 400, at least about 420, at least about 440, at least about 460, at least about 480, at least about 500 or at least about 550 mg.
52. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 47, 50, and 51, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 240 mg.
53. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 47, 50, and 51, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 480 mg.
54. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 47, and 50 to 53, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose about once every 1, 2, 3 or 4 weeks.
55. The method, checkpoint inhibitor, or the combination of assays of claims 41 to 47, 50, 51, and 54 wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose or about 240 mg once about every two weeks.
56. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 47, 50, and 51, wherein the anti-PD-1 antibody or antigen-binding portion thereof is administered at a flat dose of about 480 mg once about every four weeks.
57. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 57, wherein the anti-PD-1 antibody is administered for as long as clinical benefit is observed or until unmanageable toxicity or disease progression occurs.
58. The method, checkpoint inhibitor, or the combination of assays of any one of claims 40 to 57, wherein the antibody is formulated for intravenous administration.
59. The method, checkpoint inhibitor, or the combination of assays of any one of claims 40 to 48, wherein the antibody is administered at a subtherapeutic dose.
60. The method, checkpoint inhibitor, or the combination of assays of any one of claims 41 to 59, further comprising administering an antibody or an antigen binding fragment thereof that binds specifically to CTLA-4 ("an anti-CTLA-4 antibody").
61. The method, checkpoint inhibitor, or the combination of assays of claim 60, wherein the anti-CTLA-4 antibody cross-competes with ipilimumab or tremelimumab for binding to human CTLA-4.
62. The method, checkpoint inhibitor, or the combination of assays of claim 60 or 61, wherein the anti-CTLA-4 antibody binds to the same epitope as ipilimumab or tremelimumab.
63. The method, checkpoint inhibitor, or the combination of assays of any one of claims 60 to 62, wherein the anti-CTLA-4 antibody is ipilimumab.
64. The method, checkpoint inhibitor, or the combination of assays of any one of claims 60 to 62, wherein the anti-CTLA-4 antibody is tremelimumab.
65. The method, checkpoint inhibitor, or the combination of assays of any one of claims 60 to 64, wherein the anti-CTLA-4 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.
66. The method, checkpoint inhibitor, or the combination of assays of any one of claims 60 to 65, wherein the anti-CTLA-4 antibody is administered at a dose of 1 mg/kg body weight once every 6 weeks.
67. The method, checkpoint inhibitor, or the combination of assays of any one of claims 60 to 65, wherein the anti-CTLA-4 antibody is administered at a dose of 1 mg/kg body weight once every 4 weeks.
68. The method, checkpoint inhibitor, or the combination of assays of any one of claims 60 to 64, wherein the anti-CTLA-4 antibody is administered at a flat dose.
69. The method, checkpoint inhibitor, or the combination of assays of claim 68, wherein the anti-CTLA-4 antibody is administered at a flat dose of at least about 40 mg, at least about 50 mg, at least about 60 mg, at least about 70 mg, at least about 80 mg, at least about 90 mg, at least about 100 mg, at least about 110 mg, at least about 120 mg, at least about 130 mg, at least about 140 mg, at least about 150 mg, at least about 160 mg, at least about 170 mg,at least about 180 mg, at least about 190 mg, or at least about 200 mg.
70. The method, checkpoint inhibitor, or the combination of assays of claim 68 or 69, wherein the anti-CTLA-4 antibody is administered as a flat dose about once every 2, 3, 4, 5, 6, 7, or 8 weeks.
71. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is derived from a cancer selected from the group consisting of hepatocellular cancer, gastroesophageal cancer, melanoma, bladder cancer, lung cancer, kidney cancer, head and neck cancer, colon cancer, and any combination thereof.
72. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is derived from a melanoma.
73. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is derived from a small cell carcinoma of the head and neck.
74. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is relapsed.
75. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is refractory.
76. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is refractory following at least one prior therapy comprising administration of at least one anticancer agent.
77. The method, checkpoint inhibitor, or the combination of assays of claim 76, wherein the at least one anticancer agent comprises a standard of care therapy.
78. The method, checkpoint inhibitor, or the combination of assays of claim 76 or 77, wherein the at least one anticancer agent comprises an immunotherapy.
79. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is locally advanced.
80. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the tumor is metastatic.
81. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the administering treats the tumor.
82. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the administering reduces the size of the tumor.
83. The method, checkpoint inhibitor, or the combination of assays of claim 82, wherein the size of the tumor is reduced by at least about 10%, about 20%, about 30%, about 40%, or about 50% compared to the tumor size prior to the administration.
84. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the subject exhibits progression-free survival of at least about one month, at least about 2 months, at least about 3 months, at least about 4 months, at least about 5 months, at least about 6 months, at least about 7 months, at least about 8 months, at least about 9 months, at least about 10 months, at least about 11 months, at least about one year, at least about eighteen months, at least about two years, at least about three years, at least about four years, or at least about five years after the initial administration.
85. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the subject exhibits stable disease after the administration.
86. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the subject exhibits a partial response after the administration.
87. The method, checkpoint inhibitor, or the combination of assays of any preceding claim, wherein the subject exhibits a complete response after the administration.
88. A device comprising: (i) a means for loading a biological sample; and (ii) a means for detecting: (a) a level of expression of a panel of genes in the biological sample, (b) a PD-L1 expression pattern in a biological sample, and (c) a tumor mutation burden (TMB) status of a biological sample.
89. The device of claim 88, wherein the panel of genes comprises at least 5 genes, wherein the at least 5 genes comprise: (i) at least one gene selected from the group consisting of TDO2, PD1, CTLA-4, Pan- KIR-L, Pan-KIR-S, ICOS-L, CCR2, CD73, CSF1R, GITR, GITRL, IL8, CXCR1, CD80, CD86, Galectin -9, OX40, OX40L, STING, TIM3, NKG2D, PVR, EP4, CXCR2, Nectin-2, and KIR-Liri; and (ii) at least one gene selected from the group consisting of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA- DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3.
90. The device of claim 88 or 89, wherein the means for detecting a level of expression of a panel of genes in the biological sample comprises a first biological probe, wherein the first biological probe specifically interacts with a transcript of at least one gene in the panel of genes.
91. The device of claim 90, further comprising a means for detecting a level of expression of the transcript.
92. The device of claim 90 or 91, wherein the transcript comprises an mRNA or a cDNA.
93. The device of any one of claims 90 to 92, wherein the means for detecting a level of expression of a panel of genes in the biological sample further comprises a second biological probe, a third biological probe, a fourth biological probe, and a fifth biological probe.
94. The device of claim 93, wherein the first biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a first gene of the at least 5 genes; wherein the second biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a second gene of the at least 5 genes; wherein the third biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a third gene of the at least 5 genes; wherein the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes; and wherein the fourth biological probe is capable of specifically binding to a nucleotide sequence within a transcript of a fourth gene of the at least 5 genes.
95. The device of claim 94, wherein the transcript of the first gene, the transcript of the second gene, the transcript of the third gene, and/or the transcript of the fourth gene comprises an mRNA or a cDNA.
96. The device of any one of claims 88 to 95, wherein the biological sample comprises a tumor sample from a human subject.
97. The device of claim 96, wherein the tumor sample is a tumor tissue biopsy.
98. The device of claim 96 or 97, wherein the tumor sample is a formalin-fixed, paraffin- embedded tumor tissue or a fresh-frozen tumor tissue.
99. The device of any one of claims 88 to 98, wherein the panel of genes comprises 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 110, at least about 120, at least about 130, at least about 140, at least about 150, at least about 160, at least about 170, at least about 180, at least about 190, or at least about 110 additional genes.
100. The device of claim 99, wherein at least one of the additional genes are selected from CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA- DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS- L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, and any combination thereof.
101. The device of any one of claims 88 to 100, wherein the panel of genes comprises at least about 63 genes.
102. The device of any one of claims 88 to 101, wherein the panel of genes comprises at least one housekeeping gene.
103. The device of any one of claims 88 to 102, wherein the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 housekeeping genes.
104. The device of claim 102 or 103, wherein the housekeeping genes are selected from the group consisting of ACTB, ATP5F1, DDX5, EEF1G, GAPDH, NCL, OAZ1, PPIA, RPL38, RPL6, RPS7, SLC25A3, SOD1, YWHAZ, and any combination thereof.
105. The device of any one of claims 88 to 104, wherein the panel of genes comprises at least one control gene.
106. The device of any one of claims 88 to 105, wherein the panel of genes comprises at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 16, at least 17, at least 18, at least 19, at least, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, or at least 50 control genes.
107. The device of claim 105 or 106, wherein the control genes are selected from the group consisting of ANT1, 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.
108. The device of any one of claims 88 to 107, wherein the panel of genes comprises at least about 95 genes.
109. The device of any one of claims 88 to 108, wherein the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA- DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, and TIM3.
110. The device of any one of claims 88 to 109, wherein the panel of genes comprises CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA- DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ.
111. The device of any one of claims 88 to 110, wherein the panel of genes consists essentially of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA-DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ.
112. The device of any one of claims 88 to 110, wherein the panel of genes consists of CCL2, CCL3, CCL4, CCL5, CCR2, CCR5, CD27, CD274, CD276, CD28, CD3, CD73, CD80, CD86, CD8A, CMKLR1, CSF1R, CTLA-4, CXCL10, CXCL11, CXCL9, CXCR1, CXCR2, CXCR6, EP4, Galectin -9, GITR, GITRL, GZMA, GZMK, HLA-DMA, HLA-DMB, HLA- DOA, HLA-DOB, HLA-DQA1, HLA-DRA, HLA-DRB1, HLA-E, ICOS, ICOS-L, IDO1, IFNG, IL8, IRF1, KIR-Liri, LAG3, Nectin-2, NKG2D, NKG7, OX40, OX40L, Pan-KIR-L, Pan-KIR-S, PD1, PDCD1LG2, PRF1, PSMB10, PVR, STAT1, STING, TDO2, TIGIT, TIM3, ANT1, 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, and YWHAZ.
113. The device of any one of claims 88 to 112, wherein the means for detecting a PD-L1 expression pattern in a biological sample comprises means for conducting an IHC assay.
114. The device of claim 113, wherein the IHC assay is an automated IHC assay.
115. The device of any one of claims 88 to 113, wherein the means for detecting a tumor mutation burden (TMB) status of a biological sample comprises an automated DNA sequencer.
116. The device of claim 115, wherein the automated DNA sequencer is capable of whole exome sequencing (WES).
117. A method of treating a tumor in a human subject in need thereof, comprising administering an effective amount of a checkpoint inhibitor to the subject, wherein the subject is identified as having a tumor suitable for a checkpoint inhibitor therapy, wherein the subject is identified as suitable by obtaining a biological sample from the subject and applying the biological sample to the device of any one of claims 88 to 116.
118. A method of identifying a subject suitable for an immunotherapy, comprising obtaining a biological sample from the subject and applying the biological sample to the device of any one of claims 88 to 117.
119. The method of claim 118, further comprising administering an immunotherapy to the subject.
PCT/US2020/059174 2019-11-06 2020-11-05 Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy WO2021092221A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201962931744P 2019-11-06 2019-11-06
US62/931,744 2019-11-06

Publications (1)

Publication Number Publication Date
WO2021092221A1 true WO2021092221A1 (en) 2021-05-14

Family

ID=73646517

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/059174 WO2021092221A1 (en) 2019-11-06 2020-11-05 Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy

Country Status (1)

Country Link
WO (1) WO2021092221A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023154895A1 (en) * 2022-02-11 2023-08-17 Foundation Medicine, Inc. Use of tumor mutational burden as a predictive biomarker for immune checkpoint inhibitor versus chemotherapy effectiveness in cancer treatment
US11919953B2 (en) 2020-07-15 2024-03-05 Amgen Inc. TIGIT and CD112R blockade

Citations (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
WO2007113648A2 (en) 2006-04-05 2007-10-11 Pfizer Products Inc. Ctla4 antibody combination therapy
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
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
US20110007023A1 (en) 2009-07-09 2011-01-13 Sony Ericsson Mobile Communications Ab Display device, touch screen device comprising the display device, mobile device and method for sensing a force on a display device
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
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
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
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies 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
WO2014140180A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
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
US20150079109A1 (en) 2013-09-13 2015-03-19 Beigene, Ltd. Anti-PD1 Antibodies and their Use as Therapeutics and Diagnostics
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
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
WO2016106159A1 (en) 2014-12-22 2016-06-30 Enumeral Biomedical Holding, Inc. Anti-pd-1 antibodies
WO2016126858A2 (en) 2015-02-03 2016-08-11 Anaptysbio, Inc. Antibodies directed against lymphocyte activation gene 3 (lag-3)
WO2016149201A2 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
WO2016191751A1 (en) * 2015-05-28 2016-12-01 Bristol-Myers Squibb Company Treatment of pd-l1 positive lung cancer using an anti-pd-1 antibody
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
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
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
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
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
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
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
WO2017062888A1 (en) 2015-10-09 2017-04-13 Regeneron Pharmaceuticals, Inc. Anti-lag3 antibodies and uses thereof
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
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
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
WO2017087589A2 (en) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Pd1 and/or lag3 binders
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
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
WO2017149143A1 (en) 2016-03-04 2017-09-08 Agency For Science, Technology And Research Anti-lag-3 antibodies
US20170260271A1 (en) 2014-05-13 2017-09-14 Chugai Seiyaku Kabushiki Kaisha T Cell-Redirected Antigen-Binding Molecule For Cells Having Immunosuppression Function
WO2017198741A1 (en) 2016-05-18 2017-11-23 Boehringer Ingelheim International Gmbh Anti pd-1 and anti-lag3 antibodies for cancer treatment
WO2017220555A1 (en) 2016-06-20 2017-12-28 F-Star Beta Limited Lag -3 binding members
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
WO2017220569A1 (en) 2016-06-20 2017-12-28 F-Star Delta Limited Binding molecules binding pd-l1 and lag-3
WO2018034227A1 (en) 2016-08-15 2018-02-22 国立大学法人北海道大学 Anti-lag-3 antibody
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
WO2018185043A1 (en) 2017-04-05 2018-10-11 F. Hoffmann-La Roche Ag Bispecific antibodies specifically binding to pd1 and lag3
WO2018185046A1 (en) 2017-04-05 2018-10-11 F. Hoffmann-La Roche Ag Anti-lag3 antibodies
WO2018208868A1 (en) 2017-05-10 2018-11-15 Smet Pharmaceutical Inc Human monoclonal antibodies against lag3 and uses thereof
WO2018217940A2 (en) 2017-05-24 2018-11-29 Sutro Biopharma, Inc. Pd-1/lag3 bi-specific antibodies, compositions thereof, and methods of making and using the same
WO2019011306A1 (en) 2017-07-13 2019-01-17 Nanjing Leads Biolabs Co., Ltd. Antibodies binding lag-3 and uses thereof
WO2019051542A1 (en) * 2017-09-12 2019-03-21 The University Of Sydney Predicting responses to immunotherapy
WO2019075468A1 (en) * 2017-10-15 2019-04-18 Bristol-Myers Squibb Company Methods of treating tumor
WO2019191676A1 (en) * 2018-03-30 2019-10-03 Bristol-Myers Squibb Company Methods of treating tumor
WO2020198676A1 (en) * 2019-03-28 2020-10-01 Bristol-Myers Squibb Company Methods of treating tumor

Patent Citations (90)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
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
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
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
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)
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007113648A2 (en) 2006-04-05 2007-10-11 Pfizer Products Inc. Ctla4 antibody combination therapy
WO2008156712A1 (en) 2007-06-18 2008-12-24 N. V. Organon Antibodies to human programmed death receptor pd-1
US8900587B2 (en) 2007-06-18 2014-12-02 Merck Sharp & Dohme Corp. 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
WO2009044273A2 (en) 2007-10-05 2009-04-09 Immutep Use of recombinant lag-3 or the derivatives thereof for eliciting monocyte immune response
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US20110150892A1 (en) 2008-08-11 2011-06-23 Medarex, Inc. Human antibodies that bind lymphocyte activation gene-3 (lag-3) and uses thereof
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
US20110007023A1 (en) 2009-07-09 2011-01-13 Sony Ericsson Mobile Communications Ab Display device, touch screen device comprising the display device, mobile device and method for sensing a force on a display device
WO2011066389A1 (en) 2009-11-24 2011-06-03 Medimmmune, Limited Targeted binding agents against b7-h1
WO2012122444A1 (en) 2011-03-10 2012-09-13 Provectus Pharmaceuticals, Inc. Combination of local and systemic immunomodulative therapies for enhanced treatment of cancer
WO2012145493A1 (en) 2011-04-20 2012-10-26 Amplimmune, Inc. Antibodies and other molecules that bind b7-h1 and pd-1
WO2013079174A1 (en) 2011-11-28 2013-06-06 Merck Patent Gmbh Anti-pd-l1 antibodies 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
WO2014140180A1 (en) 2013-03-15 2014-09-18 Glaxosmithkline Intellectual Property Development Limited Anti-lag-3 binding proteins
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
US20160272708A1 (en) 2013-06-26 2016-09-22 Shanghai Junshi Biosciences Inc. Anti-pd-1 antibody and use thereof
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
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
US20170260271A1 (en) 2014-05-13 2017-09-14 Chugai Seiyaku Kabushiki Kaisha T Cell-Redirected Antigen-Binding Molecule For Cells Having Immunosuppression Function
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
US10188730B2 (en) 2014-08-19 2019-01-29 Merck Sharp & Dohme Corp. Anti-LAG3 antibodies and antigen-binding fragments
WO2016028672A1 (en) 2014-08-19 2016-02-25 Merck Sharp & Dohme Corp. Anti-lag3 antibodies and antigen-binding fragments
WO2016106159A1 (en) 2014-12-22 2016-06-30 Enumeral Biomedical Holding, Inc. Anti-pd-1 antibodies
WO2016126858A2 (en) 2015-02-03 2016-08-11 Anaptysbio, Inc. Antibodies directed against lymphocyte activation gene 3 (lag-3)
WO2016149201A2 (en) 2015-03-13 2016-09-22 Cytomx Therapeutics, Inc. Anti-pdl1 antibodies, activatable anti-pdl1 antibodies, and methods of use thereof
WO2016191751A1 (en) * 2015-05-28 2016-12-01 Bristol-Myers Squibb Company Treatment of pd-l1 positive lung cancer using an anti-pd-1 antibody
WO2016196237A1 (en) 2015-05-29 2016-12-08 Agenus Inc. Anti-ctla-4 antibodies and methods of use thereof
WO2016200782A1 (en) 2015-06-08 2016-12-15 Macrogenics, Inc. Lag-3-binding molecules and methods of use thereof
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
WO2017019894A1 (en) 2015-07-29 2017-02-02 Novartis Ag Combination therapies comprising antibody molecules to lag-3
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
WO2017025016A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017024465A1 (en) 2015-08-10 2017-02-16 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 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
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
WO2017062888A1 (en) 2015-10-09 2017-04-13 Regeneron Pharmaceuticals, Inc. Anti-lag3 antibodies and uses thereof
WO2017086419A1 (en) 2015-11-18 2017-05-26 中外製薬株式会社 Method for enhancing humoral immune response
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
WO2017087589A2 (en) 2015-11-18 2017-05-26 Merck Sharp & Dohme Corp. Pd1 and/or lag3 binders
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
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
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
WO2017132827A1 (en) 2016-02-02 2017-08-10 Innovent Biologics (Suzhou) Co., Ltd. Pd-1 antibodies
WO2017149143A1 (en) 2016-03-04 2017-09-08 Agency For Science, Technology And Research Anti-lag-3 antibodies
WO2017198741A1 (en) 2016-05-18 2017-11-23 Boehringer Ingelheim International Gmbh Anti pd-1 and anti-lag3 antibodies for cancer treatment
WO2017220569A1 (en) 2016-06-20 2017-12-28 F-Star Delta Limited Binding molecules binding pd-l1 and lag-3
WO2017220555A1 (en) 2016-06-20 2017-12-28 F-Star Beta Limited Lag -3 binding members
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
WO2018034227A1 (en) 2016-08-15 2018-02-22 国立大学法人北海道大学 Anti-lag-3 antibody
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
WO2018185046A1 (en) 2017-04-05 2018-10-11 F. Hoffmann-La Roche Ag Anti-lag3 antibodies
WO2018185043A1 (en) 2017-04-05 2018-10-11 F. Hoffmann-La Roche Ag Bispecific antibodies specifically binding to pd1 and lag3
WO2018208868A1 (en) 2017-05-10 2018-11-15 Smet Pharmaceutical Inc Human monoclonal antibodies against lag3 and uses thereof
WO2018217940A2 (en) 2017-05-24 2018-11-29 Sutro Biopharma, Inc. Pd-1/lag3 bi-specific antibodies, compositions thereof, and methods of making and using the same
WO2019011306A1 (en) 2017-07-13 2019-01-17 Nanjing Leads Biolabs Co., Ltd. Antibodies binding lag-3 and uses thereof
WO2019051542A1 (en) * 2017-09-12 2019-03-21 The University Of Sydney Predicting responses to immunotherapy
WO2019075468A1 (en) * 2017-10-15 2019-04-18 Bristol-Myers Squibb Company Methods of treating tumor
WO2019191676A1 (en) * 2018-03-30 2019-10-03 Bristol-Myers Squibb Company Methods of treating tumor
WO2020198676A1 (en) * 2019-03-28 2020-10-01 Bristol-Myers Squibb Company Methods of treating tumor

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
"GenBank", Database accession no. AAB59385
"Oxford Dictionary of Biochemistry And Molecular Biology", 2000, OXFORD UNIVERSITY PRESS
"The Dictionary of Cell and Molecular Biology", 1999, ACADEMIC PRESS
BIRD ET AL., SCIENCE, vol. 242, 1988, pages 423 - 426
CARL MORRISON ET AL: "Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden", JOURNAL FOR IMMUNOTHERAPY OF CANCER, BIOMED CENTRAL LTD, LONDON, UK, vol. 6, no. 1, 9 May 2018 (2018-05-09), pages 1 - 12, XP021256268, DOI: 10.1186/S40425-018-0344-8 *
CLIN CANCER RES, vol. 21, 2015, pages 1258 - 1266
DESAI ET AL., JCO, vol. 36, 2018
DIANE DAMOTTE ET AL: "The tumor inflammation signature (TIS) is associated with anti-PD-1 treatment benefit in the CERTIM pan-cancer cohort", JOURNAL OF TRANSLATIONAL MEDICINE, vol. 17, no. 1, 4 November 2019 (2019-11-04), XP055727871, DOI: 10.1186/s12967-019-2100-3 *
GALSKY M ET AL., ANN ONCOL, vol. 28, 2017
GORELIK ET AL., AACR, April 2016 (2016-04-01)
HERBST ET AL., J CLIN ONCOL, vol. 31, 2013
HUSTON ET AL., PROC. NATL. ACAD. SCI. USA, vol. 85, 1988, pages 5879 - 5883
J. MATSUZAKI ET AL., PNAS, vol. 107, 2010, pages 7875
JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 4, 2016
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
MCCABEWU: "Positive progress in immunoPET—not just a coincidence", CANCER BIOTHER. RADIOPHARM., vol. 25, no. 3, 2010, pages 253 - 61, XP009150571, DOI: 10.1089/cbr.2010.0776
OLAFSEN ET AL.: "ImmunoPET imaging of B-cell lymphoma using 124I-anti-CD20 scFv dimers (diabodies", PROTEIN ENG. DES. SEL., vol. 23, no. 4, 2010, pages 243 - 9
PADMANEE SHARMA ET AL: "Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial", LANCET ONCOL, 25 January 2017 (2017-01-25), pages 312 - 322, XP055682224, Retrieved from the Internet <URL:https://www.sciencedirect.com/science/article/pii/S1470204517300657?via%3Dihub> [retrieved on 20200402], DOI: 10.1016/S1470-2045(17)30065-7 *
PETERS ET AL., AACR, 1 April 2017 (2017-04-01)
RAZVAN CRISTESCU ET AL: "Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy", SCIENCE, VOL. 362, N. 6411, 11 October 2018 (2018-10-11), pages 1 - 11, XP055731458, Retrieved from the Internet <URL:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6718162/pdf/nihms-1038273.pdf> [retrieved on 20200917], DOI: 10.1126/science.aar3593 *
RIBAS, UPDATE CANCER THER., vol. 2, no. 3, 2007, pages 133 - 39
SHARMA P ET AL., LANCET ONCOL, vol. 18, 2017, pages 312 - 322
SINGAL ET AL., ASCO, 1 June 2017 (2017-06-01)
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
TAUBE ET AL.: "Colocalization of inflammatory response with B7-hl expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape", SCI. TRANSL. MED., vol. 4, no. 127, 2012, pages 127 - 37, XP055370851, DOI: 10.1126/scitranslmed.3003689
VOGELSTEIN ET AL., SCIENCE, vol. 339, 2013, pages 1546 - 1558
WANG ET AL., CANCER IMMUNOL RES., vol. 2, no. 9, 2014, pages 846 - 56
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
YAP ET AL., SCI TRANL MED, vol. 4, 2012, pages 1 - 5
ZHANG ET AL., CELL DISCOV, vol. 7, March 2017 (2017-03-01), pages 3
ZHU JASON ET AL: "Evaluation of tumor microenvironment and biomarkers of immune checkpoint inhibitor (ICI) response in metastatic renal cell carcinoma (mRCC).", JOURNAL OF CLINICAL ONCOLOGY, vol. 37, no. 8_suppl, 10 March 2019 (2019-03-10), US, pages 63 - 63, XP055773659, ISSN: 0732-183X, DOI: 10.1200/JCO.2019.37.8_suppl.63 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11919953B2 (en) 2020-07-15 2024-03-05 Amgen Inc. TIGIT and CD112R blockade
WO2023154895A1 (en) * 2022-02-11 2023-08-17 Foundation Medicine, Inc. Use of tumor mutational burden as a predictive biomarker for immune checkpoint inhibitor versus chemotherapy effectiveness in cancer treatment

Similar Documents

Publication Publication Date Title
JP7458188B2 (en) How to treat tumors
US11919957B2 (en) Methods of treating tumor
US20230279114A1 (en) Methods of treating a tumor using an anti-pd-1 antibody
US20210032344A1 (en) Methods of treating tumor
US20210380693A1 (en) Methods of treating tumor
WO2021092221A1 (en) Methods of identifying a subject with a tumor suitable for a checkpoint inhibitor therapy
US20220195046A1 (en) Methods of treating tumor
EP4288090A1 (en) Methods and biomarkers in cancer
WO2023196987A1 (en) Methods of treating tumor
EA046134B1 (en) METHODS FOR TREATING 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: 20816764

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: 20816764

Country of ref document: EP

Kind code of ref document: A1