METHODS FOR SUBTYPING OF HEAD AND NECK SQUAMOUS CELL CARCINOMA

Abstract
Methods and compositions are provided for determining a subtype of head and neck squamous cell carcinoma (HNSCC) of an individual by detecting the expression level of at least one classifier biomarker selected from a group of gene signatures for HNSCC. Also provided herein are methods and compositions for determining the response of an individual with a HNSCC subtype to a therapy such as immunotherapy.
Description
FIELD OF THE INVENTION

The present invention relates to methods for determining a squamous cell carcinoma subtype of a head and neck sample and for predicting the response to a treatment for a patient inflicted with specific subtypes of head and neck cancer.


STATEMENT REGARDING SEQUENCE LISTING

The Sequence Listing associated with this application is provided in text format in lieu of a paper copy, and is hereby incorporated by reference into the specification. The name of the text file containing the Sequence Listing is GNCN_011_03WO_SeqList_ST25.txt. The text file is ˜674 KB, and was created on Aug. 2, 2018, and is being submitted electronically via EFS-Web.


BACKGROUND OF THE INVENTION

Head and Neck Squamous Cell Carcinoma (HNSCC) is comprised of cancers arising from the oral cavity, oropharynx, nasopharynx, hypopharynx, and larynx and are responsible for approximately 3% of all malignancies (NCI HNSCC https://www.cancer.gov/types/head-and-neck/hp accessed 6-7-17). The most significant predisposing factors include heavy smoking and/or alcohol use, and more recently an increasing proportion of HNSCC tumors are caused by Human Papilloma Virus (HPV) Infection. In the United States, it is projected that in 2015, there were approximately 60,000 new cases and 12,000 deaths of HNSCCC (see Siegel R L, Miller K D, Jemal A. Cancer Statistics, 2015. CA Cancer J Clin. 2015; 65: 5-29). HNSCC has been traditionally managed with surgery, radiation therapy, and/or chemotherapy such that early stage tumors are often managed with a single treatment modality while advanced stage tumors require multimodality therapy. Risk stratification and treatment decisions vary by anatomic site, stage at presentation, histologic characteristics of the tumor, and patient factors.


Recent advances in cancer genomics have led to an increased understanding of mutational and gene expression profiles in HNSCC. HNSCC subtypes, as defined by underlying genomic features, have shown varied cell of origin, tumor drivers, proliferation, immune responses, and prognosis (Lawrence M S, Sougnez C, Lichtenstein L, Cibulskis K, Lander E, Gabriel S B, et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015; 517: 576-582; Von Walter, Yin X, Wilkerson M D, Cabanski C R, Zhao N, Du Y, Ang M K, Hayward M C, Salazar A H, Hoadley K A, Fritchie K, Sailey C J, Weissler M C, Shockley W W, Zanation A M, Hackman T, Thorne L B, Funkhouser W D, Muldrew K L, Olshan A F, Randell S H, Wright F A, Shores C G, Hayes D N. (2013). Molecular Subtypes in Head and Neck Cancer Exhibit Distinct Patterns of Chromosomal Gain and Loss of Canonical Cancer Genes. PLoS One, 8(2):e56823; Keck M K, Zuo Z, Khattri a., Stricker T P, Brown C D, Imanguli M, et al. Integrative Analysis of Head and Neck Cancer Identifies Two Biologically Distinct HPV and Three Non-HPV Subtypes. Clin Cancer Res. 2014; 21: 870-881). Currently, HNSCC tumors can be categorized into one of 4 subtypes (Atypical (AT), Mesenchymal (MS), Classical (CL), Basal (BA)). Additionally, while traditionally associated with tobacco and alcohol use, an increased number of incident oropharyngeal cancers are caused by human papillomavirus (HPV).


Accordingly, there has been a growing interest in studies of HPV associated HNSCC tumors. With the exception of the use of P16 immunohistochemistry as a marker of HPV infection in oropharyngeal tumors, the molecular characteristics of HPV-associated HNSCC have largely not been incorporated into risk stratification, drug response stratification, nor clinical management decisions (chemotherapy, etc).


Cancer immunosurveillance is the principle that the immune system can identify precancerous and cancerous cells and kill these cells before they become clinically relevant, which has been demonstrated in immunodeficient mouse models. Innate and adaptive immune responses can work together to either promote or inhibit cancer growth, and evasion of immune destruction is an emerging hallmark of cancer. Deficiencies in tumor antigen expression and presentation on antigen presenting cells (APCs), infiltration of immunosuppressive cells and cytokines, and ineffective T-cell activation can lead to immunosuppression at the tumor site. Advances in the understanding of cancer and the immune system have led to effective therapies that activate antitumor responses, even in tumors that have highly developed methods of immune evasion. However the high immunosuppressive effects caused by some types of tumors limit the beneficial effects of these advances due to a delicate balance between immunoactivation and immunosuppression in a patient. Accordingly, new methods are needed to further define populations that might be likely to respond to immunotherapy.


The present invention addresses these and other needs in the field for an efficient method for improved HNSCC tumor classification that could inform prognosis, drug response and patient management based on underlying genomic and biologic tumor characteristics. The diagnostic method includes evaluation of gene expression subtypes followed by HPV gene expression and application of an algorithm for categorization of HNSCC tumors into one of 5 subtypes (Atypical (AT), Mesenchymal (MS), Classical (CL), Basal (BA), and HPV “Atypical-like”).


SUMMARY OF THE INVENTION

In one aspect, provided herein is a method for determining a head and neck squamous cell carcinoma (HNSCC) subtype of a head and neck tissue sample obtained from a patient, the method comprising detecting an expression level of at least one classifier biomarker of Table 1 or Table 3, wherein the detection of the expression level of the classifier biomarker specifically identifies a basal (BA), mesenchymal (MS), atypical (AT) or classical (CL) HNSCC subtype. In some cases, the method further comprises comparing the detected levels of expression of the at least one classifier biomarkers of Table 1 or Table 3 to the expression of the at least one classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the sample as BA, MS, AT or CL subtype based on the results of the comparing step. In some cases, the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm. In some cases, the expression level of the classifier biomarker is detected at the nucleic acid level. In some cases, the nucleic acid level is RNA or cDNA. In some cases, the detecting an expression level comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques. In some cases, the expression level is detected by performing qRT-PCR. In some cases, the detection of the expression level comprises using at least one pair of oligonucleotide primers specific for at least one classifier biomarker of Table 1 or Table 3. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the at least one classifier biomarker comprises a plurality of classifier biomarkers. In some cases, the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1. In some cases, the at least one classifier biomarker comprises all the classifier biomarkers of Table 1. In some cases, the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 5 classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3. In some cases, the at least one classifier biomarker comprises all the classifier biomarkers of Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In another aspect, provided herein is a method for determining a HNSCC subtype of a head and neck tissue sample obtained from a patient comprising detecting an expression level of at least one nucleic acid molecule that encodes a classifier biomarker having a specific expression pattern in head and neck cancer cells, wherein the classifier biomarker is selected from the group consisting of the classifier genes set forth in Table 1 or Table 3, the method comprising: (a) isolating nucleic acid material from a head and neck tissue sample from a patient; (b) mixing the nucleic acid material with oligonucleotides that are substantially complementary to portions of nucleic acid molecule of the classifier biomarker; and (c) detecting expression of the classifier biomarker. In some cases, the method further comprises comparing the detected levels of expression of the at least one classifier biomarkers of Table 1 or Table 3 to the expression of the at least one classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the sample as BA, MS, AT or CL subtype based on the results of the comparing step. In some cases, the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm. In some cases, the detecting the expression level comprises performing qRT-PCR or any hybridization-based gene assays. In some cases, the expression level is detected by performing qRT-PCR. In some cases, the detection of the expression level comprises using at least one pair of oligonucleotide primers specific for at least one classifier biomarker of Table 1 or Table 3. In some cases, the method further comprises predicting the response to a therapy for treating a subtype of HNSCC based on the detected expression level of the classifier biomarker. In some cases, the therapy is radiotherapy, surgical intervention, chemotherapy, angiogenesis inhibitors and/or immunotherapy. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the at least one nucleic acid molecule that encodes a classifier biomarker comprises a plurality of nucleic acid molecules that encode a plurality of classifier biomarkers. In some cases, the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1. In some cases, the at least one classifier biomarker comprises all the classifier biomarkers of Table 1. In some cases, the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 5 classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3. In some cases, the at least one classifier biomarker comprises all the classifier biomarkers of Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In still another aspect, provided herein is a method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method comprising measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay. In some cases, the head neck tissue sample was previously diagnosed as being squamous cell carcinoma. In some cases, the previous diagnosis was by histological examination. In some cases, the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques. In some cases, the expression level is detected by performing qRT-PCR. In some cases, the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In yet another aspect, provided herein is a method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method consisting essentially of measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay. In some cases, the head and neck tissue sample was previously diagnosed as being squamous cell carcinoma. In some cases, the previous diagnosis was by histological examination. In some cases, the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques. In some cases, the expression level is detected by performing qRT-PCR. In some cases, the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In still yet another aspect, provided herein is a method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method consisting of measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay. In some cases, the head and neck tissue sample was previously diagnosed as being squamous cell carcinoma. In some cases, the previous diagnosis was by histological examination. In some cases, the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques. In some cases, the expression level is detected by performing qRT-PCR. In some cases, the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In one aspect, provided herein is a method of determining whether a HNSCC patient is likely to respond to immunotherapy, the method comprising: determining the HNSCC subtype of a head and neck tissue sample from the patient, wherein the HNSCC subtype is selected from the group consisting of basal, mesenchymal, atypical and classical; and based on the subtype, assessing whether the patient is likely to respond to immunotherapy. In some cases, the immunotherapy comprises checkpoint inhibitor therapy. In some cases, the checkpoint inhibitor targets PD-1 or PD-L1. In some cases, the checkpoint inhibitor targets CTLA-4. In some cases, the checkpoint inhibitor is Pembrolizumab, Nivolumab or an antigen fragment binding fragment thereof. In some cases, the checkpoint inhibitor is Ipilimumab or an antigen binding fragment thereof. In some cases, the patient is initially determined to have HNSCC via a histological analysis of a sample. In some cases, the patient's HNSCC molecular subtype is selected from basal, mesenchymal, atypical or classical and is determined via a histological analysis of a sample obtained from the patient. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the determining the HNSCC subtype comprises determining expression levels of a plurality of classifier biomarkers. In some cases, the determining the expression levels of the plurality of classifier biomarkers is at a nucleic acid level by performing RNA sequencing, reverse transcriptase polymerase chain reaction (RT-PCR) or hybridization based analyses. In some cases, the plurality of classifier biomarkers for determining the HNSCC subtype is selected from a publically available HNSCC dataset. In some cases, the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset. In some cases, the plurality of classifier biomarkers for determining the HNSCC subtype is selected from Table 1 or Table 3. In some cases, the RT-PCR is quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR). In some cases, the RT-PCR is performed with primers specific to the plurality of classifier biomarkers of Table 1 or Table 3. In some cases, the method further comprises comparing the detected levels of expression of the plurality of classifier biomarkers of Table 1 or Table 3 to the expression of the plurality of classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the first sample as BA, MS, AT or CL based on the results of the comparing step. In some cases, the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm. In some cases, the plurality of the classifier biomarkers comprise each of the classifier biomarkers set forth in Table 1 or Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In another aspect, provided herein is a method for selecting a HNSCC patient for immunotherapy, the method comprising, determining a HNSCC subtype of a head and neck tissue sample from the patient, based on the subtype; and selecting the patient for immunotherapy. In some cases, the immunotherapy comprises checkpoint inhibitor therapy. In some cases, the checkpoint inhibitor targets PD-1 or PD-L1. In some cases, the checkpoint inhibitor targets CTLA-4. In some cases, the checkpoint inhibitor is Pembrolizumab, Nivolumab or an antigen fragment binding fragment thereof. In some cases, the checkpoint inhibitor is Ipilimumab or an antigen binding fragment thereof. In some cases, the patient is initially determined to have HNSCC via a histological analysis of a sample. In some cases, the patient's HNSCC molecular subtype is selected from basal, mesenchymal, atypical or classical and is determined via a histological analysis of a sample obtained from the patient. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the determining the HNSCC subtype comprises determining expression levels of a plurality of classifier biomarkers. In some cases, the determining the expression levels of the plurality of classifier biomarkers is at a nucleic acid level by performing RNA sequencing, reverse transcriptase polymerase chain reaction (RT-PCR) or hybridization based analyses. In some cases, the plurality of classifier biomarkers for determining the HNSCC subtype is selected from a publically available HNSCC dataset. In some cases, the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset. In some cases, the plurality of classifier biomarkers for determining the HNSCC subtype is selected from Table 1 or Table 3. In some cases, the RT-PCR is quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR). In some cases, the RT-PCR is performed with primers specific to the plurality of classifier biomarkers of Table 1 or Table 3. In some cases, the method further comprises comparing the detected levels of expression of the plurality of classifier biomarkers of Table 1 or Table 3 to the expression of the plurality of classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the first sample as BA, MS, AT or CL based on the results of the comparing step. In some cases, the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm. In some cases, the plurality of the classifier biomarkers comprise each of the classifier biomarkers set forth in Table 1 or Table 3. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


In yet another aspect, provided herein is a method of treating HNSCC in a subject, the method comprising: measuring the expression level of at least one biomarker nucleic acid in a HNSCC sample obtained from the subject, wherein the at least one biomarker nucleic acid is selected from a set of biomarkers listed in Table 1 or Table 3, wherein the presence, absence and/or level of the at least one biomarker indicates a subtype of the HNSCC; and administering an immunotherapeutic agent based on the subtype of the HNSCC. In some cases, the head and neck sample is a HNSCC sample. In some cases, the at least one biomarker nucleic acid selected from the set of biomarkers comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1. In some cases, the at least one biomarker nucleic acid selected from the set of biomarkers comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3. In some cases, the head and neck tissue sample was previously diagnosed as HNSCC. In some cases, the previous diagnosis was by histological examination. In some cases, the method further comprises measuring the expression of at least one biomarker from an additional set of biomarkers. In some cases, the additional set of biomarkers comprise gene expression signatures of Innate Immune Cells (IIC), Adaptive Immune Cells (AIC), one or more individual immune biomarkers, one or more interferon (IFN) genes, one or more major histocompatibility complex, class II (MHCII) genes or a combination thereof. In some cases, the additional set of biomarkers comprises genes selected from Tables 6A, 6B, 7, 8, 9, or a combination thereof. In some cases, the gene expression signatures of AICs are selected from Table 6A. In some cases, the gene expression signature of IICs are selected from Table 6B. In some cases, the one or more individual immune biomarkers are selected from Table 7. In some cases, one or more IFN genes are selected from Table 8. In some cases, the one or more MHCII genes are selected from Table 9. In some cases, the measuring the expression level is conducted using an amplification, hybridization and/or sequencing assay. In some cases, the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques. In some cases, the expression level is detected by performing qRT-PCR. In some cases, the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient. In some cases, the bodily fluid is blood or fractions thereof, urine, saliva, or sputum. In some cases, the subject's HNSCC subtype is selected from basal, mesenchymal, atypical or classical. In some cases, the at least one biomarker nucleic acid is a plurality of biomarker nucleic acids, wherein the plurality of biomarker nucleic acids comprises at least one biomarker nucleic acid listed in Table 1 or Table 3 in combination with one or more biomarker nucleic acids from a publically available HNSCC dataset, wherein the presence, absence and/or level of the plurality of biomarker nucleic acids indicates a subtype of the HNSCC. In some cases, the at least one biomarker nucleic acid is a plurality of biomarker nucleic acids, wherein the plurality of biomarker nucleic acids comprises all of the biomarker nucleic acids listed in Table 1 or Table 3 in combination with one or more biomarker nucleic acids from a publically available HNSCC dataset, wherein the presence, absence and/or level of the plurality of biomarker nucleic acids indicates a subtype of the HNSCC. In some cases, the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset. In some cases, the method further comprises determining the HPV status of the patient. In some cases, the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient. In some cases, the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 illustrates five-fold cross validation curves using a Clanc 50:50 high;low approach on the TCGA dataset (n=520) to guide the selection of the number of genes per subtype to include in the signature for HNSCC subtyping provided herein.



FIG. 2A illustrates survival curves comparing HPV positive atypical vs. HPV positive non-Atypical tumors from TCGA dataset that were subtyped using the 144 gene set (Table 1) showing that HPV positive samples that do not belong to the atypical gene expression subtype or “atypical-like” subtype demonstrate a worse survival and may be more similar to smoking induced non-HPV HNSCC tumors. FIG. 2B illustrates the distribution of sample correlation with atypical centroid and silhouette among non-atypical HPV+ samples. Coxph agreed with logrank test (p=0.039). When adjusted for stage p=0.15.



FIG. 3 illustrates the distribution of sample correlation with atypical centroid and silhouette among non-atypical HPV+ samples.



FIG. 4A illustrates survival curves comparing HPV positive atypical to HPV positive non-Atypical tumors in the TCGA dataset with and without adjustment by correlation and silhouette score using the 840 gene gold standard. FIG. 4B illustrates that among non-atypical HPV+ tumors, 3 were atypical-like as measured by correlation with atypical centroid and silhouette. FIG. 4C illustrates survival differences strengthened with adjustment of atypical-like HPV samples using correlation and silhouette. Silhouette* compares distance to the winner centroid vs distance to the atypical centroid. Values near zero mean the sample was almost called atypical. Coxph analysis results agree with logrank test. When adjusted for stage (I-III vs IV) in part FIG. 4C, the survival-group association p-value goes from p=0.026 to p=0.11. HPV+ patients have a lot of missing stage data (28/71 missing).



FIG. 5 illustrates heatmap of immune cells and 30 immune markers across defined HNSCC subtypes (the 144 gene signature plus HPV gene expression) in the TCGA dataset as described in Example 1. Tcm=central memory T cells, Tem=Effector Memory T cells, Th1=Type 1 T helper cells, Th2=Type 2 T helper cells, TFH=T follicular helper cells, Th17=T helper 17 cells, Treg=Tregulatory cells, Tgd=Gamma Delta Tcells.



FIG. 6 illustrates boxplots of immune cells and immune markers across defined HNSCC subtypes (144 gene signature plus HPV gene expression) in the TCGA dataset. Mutation burden is also included at the end of the immune box plots.



FIG. 7 illustrates a pairwise correlation matrix of immune cells and immune markers examined in the TCGA dataset



FIG. 8 illustrates association strength (adjusted R-squared from linear model) between PDL1 (low/high) and individual immune markers versus subtype (using the 144 gene signature with HPV group) and immune markers. PDL1 predictive strength of Tcell expression across the 5 subtypes.



FIG. 9 illustrates Tcell expression pattern in various datasets using the 144 gene signature. Evaluations included the fifth subtype, HPV Atypical-like in datasets where gene expression of HPV was available.



FIG. 10 illustrates TCGA immune marker-survival hazard ratios and nominal 95% confidence intervals, within subtype (adjusting for stage using coxph) and overall (adjusting for stage and subtype). Immune markers with at least one association p<0.05 are shown. 144-gene subtyper described in Example 1 was used.



FIG. 11 illustrates the TCGA datasets, and the other datasets (samples, age, smoking, anatomic site, tumor, node, metastasis (TNM) stage and HPV by gene expression.) used to develop and validate the GeneCentric HNSCC subtyping.



FIG. 12 illustrates the agreement between the reduced gene signature of 144 genes (36 for each of the 4 subtypes) and the gold standard 840 gene signature in the training (TCGA) as well as multiple testing datasets (Keck, von Walter and Wichman) is provided.



FIG. 13 illustrates the evaluation of HPV status versus gold standard subtype and versus 144 gene subtype in TCGA and Keck datasets, using gene expression defined HPV status. Whole genome or E6 gene expression was used in the TCGA dataset and HPV E6 expression was used in the Keck dataset based on available HPV expression data.



FIG. 14 illustrates the agreement of the reduced gene signature of 80 genes (20 for each subtype) with the gold standard 840 gene signature in the training dataset (TCGA) as well as multiple testing datasets Von Walter, Keck, and Wichman is provided.



FIG. 15 illustrates the gold standard subtype agreement with the 80 gene signature and 144 gene signature overall, and by subtype. Gold standard subtype agreement with the 80 gene signature and 144 gene signature overall, and by subtype.



FIG. 16 provides stage information for the TCGA dataset, and the other datasets used to develop and validate the GeneCentric HNSCC subtyping.



FIG. 17 illustrates survival curves comparing basal, mesenchymal, atypical, classical, and HPV positive (“Atypical-like”) tumors from the TCGA dataset that were subtyped using the 144 gene set (Table 1).





DETAILED DESCRIPTION OF THE INVENTION
Overview

The present invention provides kits, compositions and methods for identifying or diagnosing head and neck squamous cell carcinoma or cancer (HNSCC). That is, the methods can be useful for molecularly defining subsets of HNSCC cancer. The kits, compositions and methods can be performed to detect HNSCC in patients that are HPV negative or HPV positive. HPV status of the patient can be determined by detecting expression of HPV related genes and/or protein as described herein. The methods provide a classification of HNSCC that can be prognostic and predictive for therapeutic response. The therapeutic response can include chemotherapy, immunotherapy, surgical intervention and radiotherapy. The methods can be also provide a prognosis with regards to nodal metastasis and overall survival for HNSCC patients according to their HNSCC subtype (e.g., AT, HPV+ AT-like, MS, CL and BA).


While a useful term for epidemiologic purposes, “Head and Neck Squamous Cell Carcinoma” can refer to cancers arising from the oral cavity, oropharynx, nasopharynx, hypopharynx, and larynx. Subtypes of these types of cancer as defined by underlying genomic features can have varied cell of origin, tumor drivers, proliferation, immune responses, and prognosis.


“Determining a HNSCC subtype” can include, for example, diagnosing or detecting the presence and type of HNSCC, monitoring the progression of the disease, and identifying or detecting cells or samples that are indicative of subtypes.


In one embodiment, HNSCC status is assessed through the evaluation of expression patterns, or profiles, of a plurality of classifier genes or biomarkers in one or more subject samples alone or in combination with assessing HPV status. For the purpose of discussion, the term subject, or subject sample, refers to an individual regardless of health and/or disease status. A subject can be a subject, a study participant, a control subject, a screening subject, or any other class of individual from whom a sample is obtained and assessed in the context of the invention. Accordingly, a subject can be diagnosed with HNSCC (including subtypes, or grades thereof), can present with one or more symptoms of HNSCC, or a predisposing factor, such as a family (genetic) or medical history (medical) factor, for HNSCC, can be undergoing treatment or therapy for HNSCC, or the like. Alternatively, a subject can be healthy with respect to any of the aforementioned factors or criteria. It will be appreciated that the term “healthy” as used herein, is relative to HNSCC status, as the term “healthy” cannot be defined to correspond to any absolute evaluation or status. Thus, an individual defined as healthy with reference to any specified disease or disease criterion, can in fact be diagnosed with any other one or more diseases, or exhibit any other one or more disease criterion, including one or more other cancers.


As used herein, an “expression profile” or a “biomarker profile” or “gene signature” comprises one or more values corresponding to a measurement of the relative abundance, level, presence, or absence of expression of a discriminative or classifier gene or biomarker. An expression profile can be derived from a subject prior to or subsequent to a diagnosis of HNSCC, can be derived from a biological sample collected from a subject at one or more time points prior to or following treatment or therapy, can be derived from a biological sample collected from a subject at one or more time points during which there is no treatment or therapy (e.g., to monitor progression of disease or to assess development of disease in a subject diagnosed with or at risk for HNSCC), or can be collected from a healthy subject. The term subject can be used interchangeably with patient. The patient can be a human patient. The one or more biomarkers of the biomarker profiles provided herein are selected from one or more biomarkers of Table 1 or 3.


As used herein, the term “determining an expression level” or “determining an expression profile” or “detecting an expression level” or “detecting an expression profile” as used in reference to a biomarker or classifier means the application of a biomarker specific reagent such as a probe, primer or antibody and/or a method to a sample, for example a sample of the subject or patient and/or a control sample, for ascertaining or measuring quantitatively, semi-quantitatively or qualitatively the amount of a biomarker or biomarkers, for example the amount of biomarker polypeptide or mRNA (or cDNA derived therefrom). For example, a level of a biomarker can be determined by a number of methods including for example immunoassays including for example immunohistochemistry, ELISA, Western blot, immunoprecipation and the like, where a biomarker detection agent such as an antibody for example, a labeled antibody, specifically binds the biomarker and permits for example relative or absolute ascertaining of the amount of polypeptide biomarker, hybridization and PCR protocols where a probe or primer or primer set are used to ascertain the amount of nucleic acid biomarker, including for example probe based and amplification based methods including for example microarray analysis, RT-PCR such as quantitative RT-PCR (qRT-PCR), serial analysis of gene expression (SAGE), Northern Blot, digital molecular barcoding technology, for example Nanostring Counter Analysis, and TaqMan quantitative PCR assays. Other methods of mRNA detection and quantification can be applied, such as mRNA in situ hybridization in formalin-fixed, paraffin-embedded (FFPE) tissue samples or cells. This technology is currently offered by the QuantiGene ViewRNA (Affymetrix), which uses probe sets for each mRNA that bind specifically to an amplification system to amplify the hybridization signals; these amplified signals can be visualized using a standard fluorescence microscope or imaging system. This system for example can detect and measure transcript levels in heterogeneous samples; for example, if a sample has normal and tumor cells present in the same tissue section. As mentioned, TaqMan probe-based gene expression analysis (PCR-based) can also be used for measuring gene expression levels in tissue samples, and this technology has been shown to be useful for measuring mRNA levels in FFPE samples. In brief, TaqMan probe-based assays utilize a probe that hybridizes specifically to the mRNA target. This probe contains a quencher dye and a reporter dye (fluorescent molecule) attached to each end, and fluorescence is emitted only when specific hybridization to the mRNA target occurs. During the amplification step, the exonuclease activity of the polymerase enzyme causes the quencher and the reporter dyes to be detached from the probe, and fluorescence emission can occur. This fluorescence emission is recorded and signals are measured by a detection system; these signal intensities are used to calculate the abundance of a given transcript (gene expression) in a sample.


In one embodiment, the “expression profile” or a “biomarker profile” or “gene signature” associated with the gene cassettes or classifier genes described herein (e.g., Tables 1 and 3) can be useful for distinguishing between normal and tumor samples. In another embodiment, the tumor samples are Head and Neck Squamous Cell Carcinoma (HNSCC). In another embodiment, HNSCC can be further classified as atypical (AT), basal (BA), classical (CL) and mesenchymal (MS) based upon an expression profile determined using the methods provided herein. In still another embodiment, the expression of HPV genes is determined in the HNSCC sample in order to ascertain the HPV status. The HPV status can be determined prior to, in parallel or after classifying the subtype of HNSCC using the gene signatures presented herein. Expression profiles using the classifier genes disclosed herein (e.g., Table 1 or Table 3) can provide valuable molecular tools for specifically identifying HNSCC subtypes, and for evaluating therapeutic efficacy in treating HNSCC. Accordingly, the invention provides methods for screening and classifying a subject for molecular HNSCC subtypes and methods for monitoring efficacy of certain therapeutic treatments for HNSCC.


In some instances, a single classifier gene provided herein is capable of identifying subtypes of HNSCC with a predictive success of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%.


In some instances, a single classifier gene as provided herein is capable of determining HNSCC subtypes with a sensitivity or specificity of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%.


The present invention also encompasses a system capable of distinguishing various subtypes of HNSCC not detectable using current methods. This system can be capable of processing a large number of subjects and subject variables such as expression profiles and other diagnostic criteria. The methods described herein can also be used for “pharmacometabonomics,” in analogy to pharmacogenomics, e.g., predictive of response to therapy. In this embodiment, subjects could be divided into “responders” and “nonresponders” using the expression profile as evidence of “response,” and features of the expression profile could then be used to target future subjects who would likely respond to a particular therapeutic course.


The expression profile can be used in combination with other diagnostic methods including histochemical, immunohistochemical, cytologic, immunocytologic, and visual diagnostic methods including histologic or morphometric evaluation of head and neck tissue.


In various embodiments of the present invention, the expression profile derived from a subject is compared to a reference expression profile. A “reference expression profile” or “control expression profile” can be a profile derived from the subject prior to treatment or therapy; can be a profile produced from the subject sample at a particular time point (usually prior to or following treatment or therapy, but can also include a particular time point prior to or following diagnosis of HNSCC); or can be derived from a healthy individual or a pooled reference from healthy individuals. A reference expression profile can be generic for HNSCC or can be specific to different subtypes of HNSCC. The HNSCC reference expression profile can be from the oral cavity, oropharynx, nasopharynx, hypopharynx, larynx or any combination thereof.


The reference expression profile can be compared to a test expression profile. A “test expression profile” can be derived from the same subject as the reference expression profile except at a subsequent time point (e.g., one or more days, weeks or months following collection of the reference expression profile) or can be derived from a different subject. In summary, any test expression profile of a subject can be compared to a previously collected profile from a subject that has a AT, MS, BL or CL HNSCC subtype. The previously collected profile can be HPV positive or negative.


The classifier biomarkers of the invention can include nucleic acids (RNA, cDNA, and DNA) and proteins, and variants and fragments thereof. Such biomarkers can include DNA comprising the entire or partial sequence of the nucleic acid sequence encoding the biomarker, or the complement of such a sequence. The biomarkers described herein can include RNA comprising the entire or partial sequence of any of the nucleic acid sequences of interest, or their non-natural cDNA products, obtained synthetically in vitro in a reverse transcription reaction. The biomarker nucleic acids can also include any expression product or portion thereof of the nucleic acid sequences of interest. A biomarker protein can be a protein encoded by or corresponding to a DNA biomarker of the invention. A biomarker protein can comprise the entire or partial amino acid sequence of any of the biomarker proteins or polypeptides. The biomarker nucleic acid can be extracted from a cell or can be cell free or extracted from an extracellular vesicular entity such as an exosome.


A “classifier biomarker” or “biomarker” or “classifier gene” can be any gene or protein whose level of expression in a tissue or cell is altered compared to that of a normal or healthy cell or tissue. For example, a “classifier biomarker” or “biomarker” or “classifier gene” can be any gene or protein whose level of expression in a tissue or cell is altered in a specific HNSCC subtype. The detection of the biomarkers of the invention can permit the determination of the specific subtype. The “classifier biomarker” or “biomarker” or “classifier gene” may be one that is up-regulated (e.g. expression is increased) or down-regulated (e.g. expression is decreased) relative to a reference or control as provided herein. The reference or control can be any reference or control as provided herein. In some embodiments, the expression values of genes that are up-regulated or down-regulated in a particular subtype of HNSCC can be pooled into one gene cassette. The overall expression level in each gene cassette is referred to herein as the “expression profile” and is used to classify a test sample according to the subtype of HNSCC. However, it is understood that independent evaluation of expression for each of the genes disclosed herein can be used to classify tumor subtypes without the need to group up-regulated and down-regulated genes into one or more gene cassettes. In some cases, as shown in Table 2, a total of 144 biomarkers can be used for HNSCC subtype determination. For each HNSCC subtype, 18 of the 36 biomarkers can be negatively correlated genes while 18 can be positively correlated genes which can be selected as the gene signature of a specific HNSCC subtype. In some cases, as shown in Table 4, a total of 80 biomarkers can be used for HNSCC subtype determination. For each HNSCC subtype, 10 of the 20 biomarkers can be negatively correlated genes while 10 can be positively correlated genes which can be selected as the gene signature of a specific HNSCC subtype.


The classifier biomarkers of the invention include any gene or protein that is selectively expressed in HNSCC, as defined herein above. Sample biomarker genes are listed in Tables 1-4, below. In Table 2 or Table 4, the first column of the table represents the biomarker list selected for distinguishing atypical (AT). The second column of the table represents the biomarker list selected for distinguishing Mesenchymal (MS). The third column of the table represents the biomarker list selected for distinguishing classical (CL). The last column of the table represents the biomarker list selected for distinguishing basal (BA).


The relative gene expression levels as represented by the tsat as described herein of the classifier biomarkers for HNSCC subtyping are shown in Table 1. In one embodiment, the gene expression levels (i.e., T-statistics) of the classifier biomarkers for HNSCC subtyping are shown in Table 1. In one embodiment, all 144 genes of Table 1 can be used to classify the subtypes of HNSCC. In one embodiment, the first 36 genes are the selected gene signature biomarkers for Basal (BA), with gene numbers 1-18 up-regulated and gene numbers 19-36 down-regulated compared to a non-BA sample. In another embodiment, gene numbers 37-72 are the selected gene signature biomarkers specific for Mesenchymal (MS), with gene numbers 37-55 up-regulated and gene numbers 56-72 down-regulated compared to a non-MS sample. In yet another embodiment, gene numbers 73-108 are the selected gene signature biomarkers specific for Atypical (AT), with gene numbers 73-90 up-regulated and gene numbers 91-108 down-regulated compared to a non-AT sample. In still another embodiment, gene numbers 109-144 are the selected gene signature biomarkers specific for Classical (CL), with gene numbers 109-126 up-regulated and gene numbers 127-144 down-regulated compared to a non-CL sample.









TABLE 1







Gene Centroids of 144 Classifier Biomarkers for the Head & Neck Squamous Cell Carcinoma (HNSCC) Subtypes




















GenBank
SEQ


Gene

Atypical
Mesenchymal
Classical
Basal
Accession
ID


Symbol
Gene Name
(AT)
(MS)
(CL)
(BL)
Number*
NO:

















ABCC1
ATP binding cassette
−2.082462349
−6.056787955
14.59888581
−4.393196801
NM_004996
1



subfamily C member 1


ABCC5
ATP binding cassette
6.735673492
−6.863599358
13.37272019
−11.24755644
NM_005688
2



subfamily C member 5


ACTN1
actinin alpha 1
−15.80447651
9.780116364
−2.540362155
8.160936953
NM_001130004
3


ACTR1A
ARP1 actin related protein
−7.43029722
5.851180405
−10.40965677
10.4118965
NM_005736
4



1 homolog A


ADCY10
adenylate cyclase 10
−0.716636613
−1.221986421
12.76526279
−8.904177745
NM_018417
5


AKR1C1
aldo-keto reductase
−1.10933351
−6.291183618
12.40184171
−3.263102998
NM_001353
6



family 1 member C1


APBB2
amyloid beta precursor
−15.01146625
12.44403993
−3.898903276
5.930295847
NM_004307
7



protein binding family B



member 2


APOL3
apolipoprotein L3
2.286291323
5.440122405
−11.35875857
2.052558416
NM_014349
8


AQP3
aquaporin 3
3.750713938
−5.715868054
−11.04600183
11.23366375
NM_004925
9


ATP13A4
ATPase 13A4
12.01816284
−12.34716568
1.995296458
−1.467805824
NM_032279
10


ATP6V1D
ATPase H+ transporting
−7.52217377
−0.516445247
−7.608420636
14.34606418
NM_015994
11



V1 subunit D


C16orf57
U6 snRNA biogenesis
−13.73713236
5.333139172
−2.658346322
10.56578203
NM_024598
12



phosphodiesterase 1


C6orf168
failed axon connections
6.916821415
−3.918221166
12.70167928
−13.73240719
NM_032511
13



homolog


CAB39
calcium binding protein
−5.824036851
0.858140491
−9.792409733
13.18131087
NM_016289
14



39


CABYR
calcium binding tyrosine
−0.879225603
−0.139933807
13.33042096
−10.27836496
NM_012189
15



phosphorylation



regulated


CALD1
caldesmon 1
−13.57155338
14.00571674
−0.997301758
0.533651105
NM_033138
16


CASP4
caspase 4
−6.233121399
1.302626733
−11.00541654
14.1766376
NM_001225
17


CAV1
caveolin 1
−13.83491768
7.418519883
−3.94809826
9.71830341
NM_001753
18


CD276
CD276 molecule
−13.57242716
10.95369333
1.01404026
1.810966974
NM_001024736
19


CD74
CD74 molecule
5.84959342
7.771329118
−10.50803194
−4.449565802
NM_001025159
20


CDSN
comeodesmosin
−10.6365512
−1.493069449
−4.906096366
16.077773
NM_001264
21


CEACAM5
carcinoembryonic antigen
11.35567594
−11.26562383
0.422805332
−0.540569935
NM_004363
22



related cell adhesion



molecule 5


CHPT1
choline
9.816276839
2.262831082
4.328822728
−15.5331284
NM_020244
23



phosphotransferase 1


CHST7
carbohydrate
1.127395223
−1.274663652
13.63700234
−11.40544572
NM_019886
24



sulfotransferase 7


CIITA
class II major
7.849953146
5.449974972
−11.0739908
−3.674106625
NM_001286402
25



histocompatibility



complex transactivator


CLCN2
chloride voltage-gated
1.869955841
−3.034672968
12.48578206
−9.444551535
NM_004366
26



channel 2


CMTM3
CKLF like MARVEL
−6.94786756
14.99532869
−1.596971364
−6.444079867
NM_144601
27



transmembrane domain



containing 3


COCH
cochlin
7.245471303
−2.225717996
11.06208646
−14.32016052
NM_001135058
28


COL6A1
collagen type VI alpha 1
−10.61945956
15.09999138
0.023929886
−4.304047285
NM_001848
29



chain


COL6A2
collagen type VI alpha 2
−9.816813759
15.40019653
−0.059414969
−5.31650074
NM_001849
30



chain


CREB3L4
cAMP responsive
12.35426247
−1.036014582
4.170449139
−14.67779961
NM_130898
31



element binding protein 3



like 4


CSNK1A1
casein kinase 1 alpha 1
−4.157095083
−4.379943874
−6.408502727
13.78896776
NM_001025105
32


CSTA
cystatin A
7.6349338
−13.92125741
−4.634101181
9.992427797
NM_005213
33


CSTB
cystatin B
5.441142897
−10.92489787
−5.097693335
9.619831396
NM_000100
34


CYP26A1
cytochrome P450 family
5.568513158
−4.690925538
14.07156336
−12.8104492
NM_000783
35



26 subfamily A member 1


CYP4B1
cytochrome P450 family
14.45700653
−7.05416422
−4.703606141
−3.36503105
NM_001099772
36



4 subfamily B member 1


DHRS1
dehydrogenase/reductase
−0.193215811
−6.493438067
−9.311117789
14.40626466
NM_001136050
37



1


DSG1
desmoglein 1
−7.614095504
−3.451410234
−3.737838114
14.0254529
NM_001942
38


ELF3
E74 like ETS
13.68731232
−13.60589315
4.343709261
−3.869532938
NM_004433
39



transcription factor 3


EPCAM
epithelial cell adhesion
4.265932752
−6.736175943
13.73306161
−9.245971416
NM_002354
40



molecule


EPGN
epithelial mitogen
−5.618779265
−3.300485871
−6.577764094
14.31742891
NM_001270989
41


EYA2
EYA transcriptional
15.04545577
−2.190930112
1.591800862
−14.01747204
NM_005244
42



coactivator and



phosphatase 2


F2RL1
F2R like trypsin receptor
−13.80547895
2.765020504
1.370242141
9.730255475
NM_005242
43



1


FAM171A1
family with sequence
9.195667146
2.45871035
3.837122752
−14.69736658
NM_001010924
44



similarity 171 member



A1


FAM3B
family with sequence
23.49686554
−10.23769328
1.726344969
−14.59685293
NM_058186
45



similarity 3 member B


FAM40A
striatin interacting protein
0.072030977
−1.193725622
−11.40053476
10.74128539
NM_033088
46



1


FBLIM1
filamin binding LIM
−13.8444937
3.484519683
−5.129731741
14.56682766
NM_017556
47



protein 1


FGD2
FYVE, RhoGEF and PH
6.075397444
5.847611036
−10.21140803
−3.045464114
NM_173558
48



domain containing 2


FKBP9
FK506 binding protein 9
−13.43106274
7.315547198
4.438931756
2.324123909
NM_007270
49


FN1
fibronectin 1
−10.72721251
14.68092168
1.901951252
−5.378217455
NM_212482
50


FOXA1
forkhead box A1
17.64133976
−11.07111965
6.438446491
−12.00758435
NM_004496
51


FSTL3
follistatin like 3
−15.00524247
9.744960025
−2.21225958
7.130950667
NM_005860
52


FUT6
fucosyltransferase 6
16.26859049
−12.17798039
−2.993362909
−1.594985757
NM_000150
53


GALNT6
polypeptide N-acetyl
−13.07407145
2.051509658
−3.432309832
13.77063593
NM_007210
54



galactosaminyltransferase



6


GCNT2
glucosaminyl (N-acetyl)
11.00945937
−5.005886796
12.29522358
−16.35521859
NM_145649
55



transferase 2, I-branching



enzyme (I blood group)


GLS2
glutaminase 2
15.18663893
−8.147146843
8.499138056
−14.18880539
NM_013267
56


GPR110
G-protein coupled
12.7116071
−11.65119722
−3.963069483
2.212488501
AY140952
57



receptor GPR110


GPRC5B
G protein-coupled
5.833385675
5.048036921
4.465010876
−14.44626312
NM_016235
58



receptor class C group 5



member B


GPX8
glutathione peroxidase 8
−10.3612256
14.94672566
0.015638923
−4.401628144
NM_001008397
59



(putative)


GRHL3
grainyhead like
7.100258671
−14.0190496
−0.688942122
7.275695546
NM_021180
60



transcription factor 3


GSDMA
gasdermin A
−9.15951728
−1.729489137
−5.968036589
15.75333543
NM_178171
61


HEY1
hes related family bHLH
0.159448658
3.534415289
12.27328877
−13.99193179
NM_012258
62



transcription factor with



YRPW motif 1


HLA-DRA
HLA-DRA
5.635788952
7.540528354
−10.23102672
−4.248292697
KY497357
63


HLF
HLF, PAR bZIP
16.77558527
−10.04027801
4.574705233
−10.58434418
NM_002126
64



transcription factor


IKZF2
IKAROS family zinc
14.98317462
−6.074474772
−3.858000118
−5.554553773
NM_016260
65



finger 2


IL4R
interleukin 4 receptor
0.217776544
0.457253449
−13.81453534
11.02939058
NM_000418
66


INHBA
inhibin beta A subunit
−14.79489264
8.427676625
−0.696922701
6.927158767
NM_002192
67


KIAA1609
KIAA1609
−9.8340117
2.292555067
−8.206314175
14.38614729
AB046829
68


KLF5
Kruppel like factor 5
4.562842302
−13.00491807
3.89026837
4.908510892
NM_001730
69


LEPRE1
prolyl 3-hydroxylase 1
−11.83578491
16.04587546
0.555964732
−4.480170241
NM_022356
70


LMO4
LIM domain only 4
14.78188317
−4.286105968
2.544496006
−12.51954481
NM_006769
71


LOC643008
small integral membrane
12.65412229
−11.504798
−3.985040113
2.144789085
XM_017024943
72



protein 5


LRIG1
leucine rich repeats and
6.317083957
2.548224024
6.751703803
−14.41779751
NM_015541
73



immunoglobulin like



domains 1


LTBP3
latent transforming
3.235488698
7.629013681
6.180250436
−15.85948808
NM_001130144
74



growth factor beta binding



protein 3


MAL2
mal, T-cell differentiation
6.413419457
−11.71334434
−0.431651323
5.483862841
NM_052886
75



protein 2


MAP7D1
MAP7 domain containing
−8.910849678
3.124678656
−10.08235276
14.25296762
NM_018067
76



1


MEIS1
Meis homeobox 1
17.32037654
−5.648950707
−2.757291231
−9.201660614
NM_002398
77


MMP1
matrix metallopeptidase 1
−13.26866191
6.822304749
−2.002155679
8.096135704
NM_002421
78


MOBKL2B
MOB kinase activator 3B
−2.913703778
4.221634948
−11.2899732
8.301609003
NM_024761
79


MPPED1
metallophosphoesterase
0.75117262
−3.628778766
12.92063032
−8.131564761
NM_001044370
80



domain containing 1


MRAP2
melanocortin 2 receptor
8.821566644
−4.975238292
12.75983052
−14.62483234
NM_138409
81



accessory protein 2


MUC20
mucin 20, cell surface
15.44317871
−10.50413061
2.271620266
−6.871349665
NM_001282506
82



associated


MUC4
mucin 4, cell surface
15.96069019
−8.652630447
2.582388666
−9.450568232
NM_018406
83



associated


MYB
MYB proto-oncogene,
15.0257076
−5.385618207
5.6522876
−14.3163554
NM_001130173
84



transcription factor


NNMT
nicotinamide N-
−10.09900408
15.95020532
−3.274573382
−2.854813073
NM_006169
85



methyltransferase


NSUN7
NOP2/Sun RNA
16.08489314
−4.722807369
6.335643561
−16.58396549
NM_024677
86



methyltransferase family



member 7


NTRK2
neurotrophic receptor
10.23093315
−6.376330063
12.76453966
−14.64867156
NM_006180
87



tyrosine kinase 2


OLFML2B
olfactomedin like 2B
−6.80300319
15.66852662
−0.83099608
−7.891812186
NM_001297713
88


OLFML3
olfactomedin like 3
−6.546734956
16.36834609
−3.370305787
−6.678215459
NM_020190
89


P4HTM
prolyl 4-hydroxylase,
11.8274774
2.672495061
5.253671456
−18.69507373
NM_177939
90



transmembrane


PAQR5
progestin and adipoQ
−10.21974925
−0.676386205
−2.846883938
13.12800887
NM_001104554
91



receptor family member 5


PATZ1
POZ/BTB and AT hook
8.493850484
2.966225133
7.456620531
−17.56473037
NM_014323
92



containing zinc finger 1


PBX1
PBX homeobox 1
13.2718934
−1.583812326
4.352097293
−15.20010123
NM_002585
93


PCOLCE
procollagen C-
−8.027597076
16.0810449
0.683124494
−8.370334
NM_002593
94



endopeptidase enhancer


PHLDB1
pleckstrin homology like
−11.25408443
15.15862534
−1.738679371
−2.245099413
NM_015157
95



domain family B member



1


PIR
pirin
7.490180987
−9.475330645
13.12744434
−9.233843088
NM_003662
96


PKP3
plakophilin 3
−5.164891557
−4.425600449
−5.236735217
13.83389626
NM_007183
97


PLAC8
placenta specific 8
16.72602757
−4.212412233
1.082753487
−13.26806322
NM_001130716
98


PLD2
phospholipase D2
−4.518197205
−5.092960256
−7.348969116
15.63604839
NM_002663
99


PMP22
peripheral myelin protein
−7.194357544
14.68864942
1.396604647
−8.43537032
NM_000304
100



22


PPARD
peroxisome proliferator
−7.071754506
−3.884764304
−4.912078226
14.90821141
NM_006238
101



activated receptor delta


PPL
periplakin
6.261655734
−12.16995239
−1.756388798
7.199844946
NM_002705
102


PRKX
protein kinase, X-linked
8.851366302
−8.998241561
13.1773563
−11.08142743
NM_005044
103


PRSS27
protease, serine 27
9.329134265
−10.80925456
−5.741713479
6.225164659
NM_031948
104


PTH1R
parathyroid hormone 1
−6.722598413
14.82555669
−0.472460255
−7.45169274
NM_000316
105



receptor


PTRF
caveolae associated
−13.14332726
9.468944832
−4.638965523
7.621207859
NM_012232
106



protein 1


RAB25
RAB25, member RAS
5.219572333
−11.52664115
−0.156829226
6.244155734
NM_020387
107



oncogene family


RAB6B
RAB6B, member RAS
−1.603161781
0.110021996
15.94132967
−12.01912932
NM_016577
108



oncogene family


RCN3
reticulocalbin 3
−8.161767875
14.46620853
1.654069744
−7.483978219
NM_020650
109


RFTN1
raftlin, lipid raft linker 1
−4.573623438
9.589915176
−10.67106662
4.172768882
NM_015150
110


RGS20
regulator of G protein
−12.52290526
2.810238619
−4.159943794
13.1034325
NM_170587
111



signaling 20


RIMKLA
ribosomal modification
7.013215205
−3.910313002
13.32678642
−14.36397185
NM_173642
112



protein rimK like family



member A


SCARA3
scavenger receptor class
8.486922621
2.228910754
4.488976365
−14.32712171
NM_016240
113



A member 3


SCNN1A
sodium channel epithelial
9.314590424
−10.85586757
4.480301787
−2.364983519
NM_001038
114



1 alpha subunit


SERPINE1
serpin family E member 1
−14.50591442
8.635050854
1.480793338
4.597552116
NM_000602
115


SERPINH1
serpin family H member
−14.74205489
14.20433013
0.878953362
−0.095802953
NM_001207014
116



1


SETMAR
SET domain and mariner
12.83440944
0.233932813
2.138469148
−14.67025168
NM_006515
117



transposase fusion gene


SFXN3
sideroflexin 3
−13.93133999
11.56847653
−6.74034327
8.126085326
NM_030971
118


SGEF
SH3-containing guanine
15.50488845
−8.827171693
14.7491026
−19.12717961
AY552599
119



nucleotide exchange



factor


SH2D4A
SH2 domain containing
14.91939392
−10.45712779
−1.285782521
−3.391377233
NM_022071
120



4A


SH2D5
SH2 domain containing 5
−13.29966495
1.411309304
−2.686017395
13.9859309
NM_001103161
121


SLAMF7
SLAM family member 7
4.242417997
2.960164251
−10.72277915
2.009246005
NM_021181
122


SLC16A14
solute carrier family 16
8.453385598
−5.352426361
12.76874135
−13.90188986
NM_152527
123



member 14


SLC31A2
solute carrier family 31
−8.009411845
6.173856422
−11.1271606
11.27415259
NM_001860
124



member 2


SLC9A3R1
SLC9A3 regulator 1
9.971190685
−12.54044249
4.659221949
−1.518674303
NM_004252
125


SNAI2
snail family
−14.55807791
7.947052877
0.055367787
6.526518445
NM_003068
126



transcriptional repressor 2


SPARC
secreted protein acidic and
−9.442299973
14.5249952
0.460899979
−5.27130454
NM_003118
127



cysteine rich


SPINK5
serine peptidase inhibitor,
8.887143269
−10.88801305
−5.380297697
6.431225145
NM_001127698
128



Kazal type 5


TAGLN
transgelin
−8.774403137
14.78574024
1.128839097
−6.748363792
NM_001001522
129


TBXA2R
thromboxane A2 receptor
−7.041663263
14.43062303
−1.279759745
−6.069091489
NM_001060
130


TGFB3
transforming growth
−5.513659516
14.73938538
−3.22553509
−6.227831211
NM_003239
131



factor beta 3


TGFBI
transforming growth
−15.54040671
11.61576363
−3.305840607
6.757370024
NM_000358
132



factor beta induced


TJP3
tight junction protein 3
17.32770506
−9.778563744
−1.776083891
−6.00908976
NM_001267560
133


TMEM51
transmembrane protein
2.637524229
3.608309483
−13.856265
5.607931398
NM_001136216
134



51


TMPRSS11A
transmembrane protease,
14.96390452
−13.07259679
1.124526863
−2.922374221
NM_182606
135



serine 11A


TMPRSS11B
transmembrane protease,
16.17986512
−11.62554287
−1.63009884
−3.200386118
NM_182502
136



serine 11B


TMPRSS2
transmembrane protease,
19.36438158
−10.34381575
2.078270847
−10.72366129
NM_001135099
137



serine 2


TTC9
tetratricopeptide repeat
11.89329997
−12.38268834
−1.756842725
1.864848764
NM_015351
138



domain 9


TXNRD1
thioredoxin reductase 1
−0.999104468
−3.250337096
14.50623299
−8.119912193
NM_182729
139


UBA7
ubiquitin like modifier
5.929616343
4.581359308
−11.05993504
−0.948212309
NM_003335
140



activating enzyme 7


VAMP3
vesicle associated
−3.668365448
1.73966571
−10.63156161
10.90939129
NM_004781
141



membrane protein 3


VAV3
vav guanine nucleotide
4.420409601
−0.330479172
−10.38204488
4.757063432
NM_006113
142



exchange factor 3


VEGFC
vascular endothelial
−13.74904145
10.29391086
−8.204386968
10.42939104
NM_005429
143



growth factor C


ZDHHC2
zinc finger DHHC-type
9.037395105
0.904115321
11.9082815
−19.85433707
NM_016353
144



containing 2





*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.













TABLE 2







Classifier Biomarkers Selected for


AT, MS, BA and CL HNSCC Subtypes












Atypical
Mesenchymal
Classical
Basal


Number
(AT)
(MS)
(CL)
(BA)














1
ACTN1
ATP13A4
ABCC1
ATP6V1D


2
APBB2
CEACAM5
ABCC5
CAB39


3
C16orf57
CMTM3
ACTR1A
CDSN


4
CALD1
COL6A1
ADCY10
CHPT1


5
CAV1
COL6A2
AKR1C1
COCH


6
CD276
CSTA
APOL3
CREB3L4


7
CYP4B1
CSTB
AQP3
CSNK1A1


8
EYA2
ELF3
C6orf168
DHRS1


9
F2RL1
FN1
CABYR
DSG1


10
FAM3B
GPR110
CASP4
EPGN


11
FKBP9
GPX8
CD74
FAM171A1


12
FOXA1
GRHL3
CHST7
FBLIM1


13
FSTL3
KLF5
CIITA
GALNT6


14
FUT6
LEPRE1
CLCN2
GCNT2


15
GLS2
LOC643008
CYP26A1
GPRC5B


16
HLF
MAL2
EPCAM
GSDMA


17
IKZF2
NNMT
FAM40A
KIAA1609


18
INHBA
OLFML2B
FGD2
LRIG1


19
LMO4
OLFML3
HEY1
LTBP3


20
MEIS1
PCOLCE
HLA-DRA
MAP7D1


21
MMP1
PHLDB1
IL4R
MRAP2


22
MUC20
PMP22
MOBKL2B
NSUN7


23
MUC4
PPL
MPPED1
NTRK2


24
MYB
PRSS27
PIR
P4HTM


25
PLAC8
PTH1R
PRKX
PAQR5


26
PTRF
RAB25
RAB6B
PATZ1


27
SERPINE1
RCN3
RFTN1
PBX1


28
SERPINH1
SCNN1A
RIMKLA
PKP3


29
SFXN3
SLC9A3R1
SLAMF7
PLD2


30
SH2D4A
SPARC
SLC16A14
PPARD


31
SNAI2
SPINK5
SLC31A2
RGS20


32
TGFBI
TAGLN
TMEM51
SCARA3


33
TJP3
TBXA2R
TXNRD1
SETMAR


34
TMPRSS11B
TGFB3
UBA7
SGEF


35
TMPRSS2
TMPRSS11A
VAMP3
SH2D5


36
VEGFC
TTC9
VAV3
ZDHHC2









The relative gene expression levels as represented by the tsat as described herein of the classifier biomarkers for HNSCC subtyping are shown in Table 3. In one embodiment, the gene expression levels (i.e., T-statistics) of the classifier biomarkers for HNSCC subtyping are shown in Table 3. In one embodiment, all 80 genes of Table 3 can be used to classify the subtypes of HNSCC. In one embodiment, the first 20 genes are the selected gene signature biomarkers for Basal (BA), with gene numbers 1-10 up-regulated and gene numbers 11-20 down-regulated compared to a non-BA sample. In another embodiment, gene numbers 21-40 are the selected gene signature biomarkers specific for Mesenchymal (MS), with gene numbers 21-30 up-regulated and gene numbers 31-40 down-regulated compared to a non-MS sample. In yet another embodiment, gene numbers 41-60 are the selected gene signature biomarkers specific for Atypical (AT), with gene numbers 41-50 up-regulated and gene numbers 51-60 down-regulated compared to a non-AT sample. In still another embodiment, gene numbers 61-80 are the selected gene signature biomarkers specific for Classical (CL), with gene numbers 61-70 up-regulated and gene numbers 71-80 down-regulated compared to a non-CL sample.









TABLE 3







Gene Centroids of 80 Classifier Biomarkers for the Head & Neck Squamous Cell Carcinoma (HNSCC) Subtypes




















GenBank
SEQ


Gene

Atypical
Mesenchymal
Classical
Basal
Accession
ID


Symbol
Gene Name
(AT)
(MS)
(CL)
(BL)
Number*
NO:

















ABCC1
ATP binding
−2.082462
−6.056787955
14.59889
−4.3932
NM_004996
1



cassette



subfamily C



member 1


ABCC5
ATP binding
6.7356735
−6.863599358
13.37272
−11.2476
NM_005688
2



cassette



subfamily C



member 5


ACTN1
actinin alpha 1
−15.80448
9.780116364
−2.54036
8.160937
NM_001130004
3


APBB2
amyloid beta
−15.01147
12.44403993
−3.8989
5.930296
NM_004307
7



precursor protein



binding family B



member 2


APOL3
apolipoprotein
2.2862913
5.440122405
−11.3588
2.052558
NM_014349
8



L3


AQP3
aquaporin 3
3.7507139
−5.715868054
−11.046
11.23366
NM_004925
9


ATP13A4
ATPase 13A4
12.018163
−12.34716568
1.995296
−1.46781
NM_032279
10


ATP6V1D
ATPase H+
−7.522174
−0.516445247
−7.60842
14.34606
NM_015994
11



transporting V1



subunit D


CABYR
calcium binding
−0.879226
−0.139933807
13.33042
−10.2784
NM_012189
15



tyrosine



phosphorylation



regulated


CASP4
caspase 4
−6.233121
1.302626733
−11.0054
14.17664
NM_001225
17


CAV1
caveolin 1
−13.83492
7.418519883
−3.9481
9.718303
NM_001753
18


CDSN
corneodesmosin
−10.63655
−1.493069449
−4.9061
16.07777
NM_001264
21


CHPT1
choline
9.8162768
2.262831082
4.328823
−15.5331
NM_020244
23



phosphotransferase 1


CHST7
carbohydrate
1.1273952
−1.274663652
13.637
−11.4054
NM_019886
24



sulfotransferase



7


CIITA
class II major
7.8499531
5.449974972
−11.074
−3.67411
NM_001286402
25



histocompatibility complex



transactivator


CMTM3
CKLF like
−6.947868
14.99532869
−1.59697
−6.44408
NM_144601
27



MARVEL



transmembrane



domain



containing 3


COL6A1
collagen type VI
−10.61946
15.09999138
0.02393
−4.30405
NM_001848
29



alpha 1 chain


COL6A2
collagen type VI
−9.816814
15.40019653
−0.05941
−5.3165
NM_001849
30



alpha 2 chain


CSTA
cystatin A
7.6349338
−13.92125741
−4.6341
9.992428
NM_005213
33


CYP26A1
ytochrome P450
5.5685132
−4.690925538
14.07156
−12.8104
NM_000783
35



family 26



subfamily A



member 1


DHRS1
dehydrogenase/reductase 1
−0.193216
−6.493438067
−9.31112
14.40626
NM_001136050
37


ELF3
E74 like ETS
13.687312
−13.60589315
4.343709
−3.86953
NM_004433
39



transcription



factor 3


EPCAM
epithelial cell
4.2659328
−6.736175943
13.73306
−9.24597
NM_002354
40



adhesion



molecule


EPGN
epithelial
−5.618779
−3.300485871
−6.57776
14.31743
NM_001270989
41



mitogen


FAM171A1
family with
9.1956671
2.45871035
3.837123
−14.6974
NM_001010924
44



sequence



similarity 171



member A1


FAM3B
family with
23.496866
−10.23769328
1.726345
−14.5969
NM_058186
45



sequence



similarity 3



member B


FAM40A
striatin
0.072031
−1.193725622
−11.4005
10.74129
NM_033088
46



interacting



protein 1


FBLIM1
filamin binding
−13.84449
3.484519683
−5.12973
14.56683
NM_017556
47



LIM protein 1


FOXA1
forkhead box A1
17.64134
−11.07111965
6.438446
−12.0076
NM_004496
51


FSTL3
follistatin like 3
−15.00524
9.744960025
−2.21226
7.130951
NM_005860
52


FUT6
fucosyltransferase 6
16.26859
−12.17798039
−2.99336
−1.59499
NM_000150
53


GCNT2
glucosaminyl
11.009459
−5.005886796
12.29522
−16.3552
NM_145649
55



(N-acetyl)



transferase 2,I-



branching



enzyme (I blood



group)


GPX8
glutathione
−10.36123
14.94672566
0.015639
−4.40163
NM_001008397
59



peroxidase 8



(putative)


GRHL3
grainyhead like
7.1002587
−14.0190496
−0.68894
7.275696
NM_021180
60



transcription



factor 3


GSDMA
gasdermin A
−9.159517
−1.729489137
−5.96804
15.75334
NM_178171
61


HLF
HLF, PAR bZIP
16.775585
−10.04027801
4.574705
−10.5843
NM_002126
64



transcription



factor


IL4R
interleukin 4
0.2177765
0.457253449
−13.8145
11.02939
NM_000418
66



receptor


INHBA
inhibin beta A
−14.79489
8.427676625
−0.69692
6.927159
NM_ 002192
67



subunit


KIAA1609
KIAA1609
−9.834012
2.292555067
−8.20631
14.38615
AB046829
68


KLF5
Kruppel like
4.5628423
−13.00491807
3.890268
4.908511
NM_001730
69



factor 5


LEPRE1
prolyl 3-
−11.83578
16.04587546
0.555965
−4.48017
NM_022356
70



hydroxylase 1


LTBP3
latent
3.2354887
7.629013681
6.18025
−15.8595
NM_001130144
74



transforming



growth factor



beta binding



protein 3


MAL2
mal, T-cell
6.4134195
−11.71334434
−0.43165
5.483863
NM_052886
75



differentiation



protein 2


MAP7D1
MAP7 domain
−8.91085
3.124678656
−10.0824
14.25297
NM_018067
76



containing 1


MEIS1
Meis homeobox
17.320377
−5.648950707
−2.75729
−9.20166
NM_002398
77



1


MOBKL2B
MOB kinase
−2.913704
4.221634948
−11.29
8.301609
NM_024761
79



activator 3B


MUC4
mucin 4, cell
15.96069
−8.652630447
2.582389
−9.45057
NM_018406
83



surface



associated


NNMT
nicotinamide N-
−10.099
15.95020532
−3.27457
−2.85481
NM_024677
86



methyltransferase


NSUN7
NOP2/Sun RNA
16.084893
−4.722807369
6.335644
−16.584
NM_006180
87



methyltransferase



family member 7


OLFML2B
olfactomedin
−6.803003
15.66852662
−0.831
−7.89181
NM_001297713
88



like 2B


OLFML3
olfactomedin like
−6.546735
16.36834609
−3.37031
−6.67822
NM_020190
89



3


P4HTM
prolyl 4-
11.827477
2.672495061
5.253671
−18.6951
NM_177939
90



hydroxylase,



transmembrane


PATZ1
POZ/BTB and
8.4938505
2.966225133
7.456621
−17.5647
NM_014323
92



AT hook



containing zinc



finger 1


PBX1
PBX homeobox
13.271893
−1.583812326
4.352097
−15.2001
NM_002585
93



1


PCOLCE
procollagen C-
−8.027597
16.0810449
0.683124
−8.37033
NM_002593
94



endopeptidase



enhancer


PHLDB1
pleckstrin
−11.25408
15.15862534
−1.73868
−2.2451
NM_015157
95



homology like



domain family B



member 1


PLAC8
placenta specific
16.726028
−4.212412233
1.082753
−13.2681
NM_001130716
98



8


PLD2
phospholipase
−4.518197
−5.092960256
−7.34897
15.63605
NM_002663
99



D2


PPARD
peroxisome
−7.071755
−3.884764304
−4.91208
14.90821
NM_006238
101



proliferator



activated



receptor delta


PPL
periplakin
6.2616557
−12.16995239
−1.75639
7.199845
NM_002705
102


PRKX
protein kinase,
8.8513663
−8.998241561
13.17736
−11.0814
NM_005044
103



X-linked


RAB6B
RAB6B,
−1.603162
0.110021996
15.94133
−12.0191
NM_016577
108



member RAS



oncogene family


RIMKLA
ribosomal
7.0132152
−3.910313002
13.32679
−14.364
NM_173642
112



modification



protein rimK like



family member A


SERPINE1
serpin family E
−14.50591
8.635050854
1.480793
4.597552
NM_000602
115



member 1


SERPINH1
serpin family H
−14.74205
14.20433013
0.878953
−0.0958
NM_001207014
116



member 1


SFXN3
sideroflexin 3
−13.93134
11.56847653
−6.74034
8.126085
NM_030971
118


SGEF
SH3-containing
15.504888
−8.827171693
14.7491
−19.1272
AY552599
119



guanine



nucleotide



exchange factor


SLC31A2
solute carrier
−8.009412
6.173856422
−11.1272
11.27415
NM_001860
124



family 31



member 2


SLC9A3R1
SLC9A3
9.9711907
−12.54044249
4.659222
−1.51867
NM_004252
125



regulator 1


SNAI2
snail family
−14.55808
7.947052877
0.055368
6.526518
NM_003068
126



transcriptional



repressor 2


TGFBI
transforming
−15.54041
11.61576363
−3.30584
6.75737
NM_000358
132



growth factor



beta induced


TJP3
tight junction
17.327705
−9.778563744
−1.77608
−6.00909
NM_001267560
133



protein 3


TMEM51
transmembrane
2.6375242
3.608309483
−13.8563
5.607931
NM_001136216
134



protein 51


TMPRSS11A
transmembrane
14.963905
−13.07259679
1.124527
−2.92237
NM_182606
135



protease, serine



11A


TMPRSS11B
transmembrane
16.179865
−11.62554287
−1.6301
−3.20039
NM_182502
136



protease, serine



11B


TMPRSS2
transmembrane
19.364382
−10.34381575
2.078271
−10.7237
NM_001135099
137



protease, serine 2


TTC9
tetratricopeptide
11.8933
−12.38268834
−1.75684
1.864849
NM_015351
138



repeat domain 9


TXNRD1
thioredoxin
−0.999104
−3.250337096
14.50623
−8.11991
NM_182729
139



reductase 1


UBA7
ubiquitin like
5.9296163
4.581359308
−11.0599
−0.94821
NM_003335
140



modifier



activating



enzyme 7


ZDHHC2
zinc finger
9.0373951
0.904115321
11.90828
−19.8543
NM_016353
144



DHHC-type



containing 2





*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.













TABLE 4







Classifier Biomarkers Selected for


AT, MS, BL and CL HNSCC Subtypes












Atypical
Mesenchymal
Classical
Basal


Number
(AT)
(MS)
(CL)
(BA)














1
ACTN1
ATP13A4
ABCC1
ATP6V1D


2
APBB2
CMTM3
ABCC5
CDSN


3
CAV1
COL6A1
APOL3
CHPT1


4
FAM3B
COL6A2
AQP3
DHRS1


5
FOXA1
CSTA
CABYR
EPGN


6
FSTL3
ELF3
CASP4
FAM171A1


7
FUT6
GPX8
CHST7
FBLIM1


8
HLF
GRHL3
CIITA
GCNT2


9
INHBA
KLF5
CYP26A1
GSDMA


10
MEIS1
LEPRE1
EPCAM
KIAA1609


11
MUC4
MAL2
FAM40A
LTBP3


12
PLAC8
NNMT
IL4R
MAP7D1


13
SERPINE1
OLFML2B
MOBKL2B
NSUN7


14
SERPINH1
OLFML3
PRKX
P4HTM


15
SFXN3
PCOLCE
RAB6B
PATZ1


16
SNAI2
PHLDB1
RIMKLA
PBX1


17
TGFBI
PPL
SLC31A2
PLD2


18
TJP3
SLC9A3R1
TMEM51
PPARD


19
TMPRSS11B
TMPRSS11A
TXNRD1
SGEF


20
TMPRSS2
TTC9
UBA7
ZDHHC2









Diagnostic Uses

In one embodiment, the methods and compositions provided herein allow for the differentiation of the four subtypes of HNSCC: (1) Basal (BA); (2) Mesenchymal (MS); (3) Atypical (AT); and (4) Classical (CL), with fewer genes needed than the molecular HNSCC subtyping methods known in the art.


In general, the methods provided herein are used to classify HNSCC sample as a particular HNSCC subtype (e.g. subtype of HNSCC). In one embodiment, the method comprises measuring, detecting or determining an expression level of at least one of the classifier biomarkers of any publically available HNSCC expression dataset. In one embodiment, the method comprises detecting or determining an expression level of at least one of the classifier biomarkers of Table 1 or Table 3 in a HNSCC sample obtained from a patient or a subject. The HNSCC sample for the detection or differentiation methods described herein can be a sample previously determined or diagnosed as squamous cell carcinoma (SCC) sample. The previous diagnosis can be based on a histological analysis. The histological analysis can be performed by one or more pathologists.


In one embodiment, the measuring or detecting step is at the nucleic acid level by performing RNA-seq, a reverse transcriptase polymerase chain reaction (RT-PCR) or a hybridization assay with oligonucleotides that are substantially complementary to portions of cDNA molecules of the at least one classifier biomarker (such as the classifier biomarkers of Table 1 or Table 3) under conditions suitable for RNA-seq, RT-PCR or hybridization and obtaining expression levels of the at least one classifier biomarkers based on the detecting step. The expression levels of the at least one of the classifier biomarkers are then compared to reference expression levels of the at least one of the classifier biomarker (such as the classifier biomarkers of Table 1 or Table 3) from at least one sample training set. The at least one sample training set can comprise, (i) expression levels of the at least one biomarker from a sample that overexpresses the at least one biomarker, (ii) expression levels from a reference BA, MS, AT or CL sample, or (iii) expression levels from SCC free head and neck sample, and classifying the head and neck tissue sample as a BA, MS, AT or CL subtype. The head and neck cancer sample can then be classified as a BA, MS, AT or CL subtype of squamous cell carcinoma based on the results of the comparing step. In one embodiment, the comparing step can comprise applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the head and neck tissue or cancer sample and the expression data from the at least one training set(s); and classifying the head and neck tissue or cancer sample as a BA, MS, AT or CL sample subtype based on the results of the statistical algorithm.


In one embodiment, the method comprises probing the levels of at least one of the classifier biomarkers provided herein, such as the classifier biomarkers of Table 1 or 3 at the nucleic acid level, in a head and neck cancer sample obtained from the patient. The head and neck cancer sample can be a sample previously determined or diagnosed as a squamous cell carcinoma (SCC or SQ) sample. The previous diagnosis can be based on a histological analysis. The histological analysis can be performed by one or more pathologists. The probing step, in one embodiment, comprises mixing the sample with one or more oligonucleotides that are substantially complementary to portions of cDNA molecules of the at least one classifier biomarkers provided herein, such as the classifier biomarkers of Table 1 or 3 under conditions suitable for hybridization of the one or more oligonucleotides to their complements or substantial complements; detecting whether hybridization occurs between the one or more oligonucleotides to their complements or substantial complements; and obtaining hybridization values of the at least one classifier biomarkers based on the detecting step. The hybridization values of the at least one classifier biomarkers are then compared to reference hybridization value(s) from at least one sample training set. For example, the at least one sample training set comprises hybridization values from a reference BA SCC, MS SCC AT SCC, and/or CL SCC sample. The head and neck cancer sample is classified, for example, as BA, MS, AT or CL based on the results of the comparing step.


The head and neck tissue sample can be any sample isolated from a human subject or patient. For example, in one embodiment, the analysis is performed on head and neck biopsies that are embedded in paraffin wax. In one embodiment, the sample can be a fresh frozen head and neck tissue sample. In another embodiment, the sample can be a bodily fluid obtained from the patient. The bodily fluid can be blood or fractions thereof (i.e., serum, plasma), urine, saliva, sputum or cerebrospinal fluid (CSF). The sample can contain cellular as well as extracellular sources of nucleic acid for use in the methods provided herein. The extracellular sources can be cell-free DNA and/or exosomes. In one embodiment, the sample can be a cell pellet or a wash. This aspect of the invention provides a means to improve current diagnostics by accurately identifying the major histological types, even from small biopsies. The methods of the invention, including the RT-PCR methods, are sensitive, precise and have multi-analyte capability for use with paraffin embedded samples. See, for example, Cronin et al. (2004) Am. J Pathol. 164(1):35-42, herein incorporated by reference.


Formalin fixation and tissue embedding in paraffin wax is a universal approach for tissue processing prior to light microscopic evaluation. A major advantage afforded by formalin-fixed paraffin-embedded (FFPE) specimens is the preservation of cellular and architectural morphologic detail in tissue sections. (Fox et al. (1985) J Histochem Cytochem 33:845-853). The standard buffered formalin fixative in which biopsy specimens are processed is typically an aqueous solution containing 37% formaldehyde and 10-15% methyl alcohol. Formaldehyde is a highly reactive dipolar compound that results in the formation of protein-nucleic acid and protein-protein crosslinks in vitro (Clark et al. (1986) J Histochem Cytochem 34:1509-1512; McGhee and von Hippel (1975) Biochemistry 14:1281-1296, each incorporated by reference herein).


In one embodiment, the sample used herein is obtained from an individual, and comprises formalin-fixed paraffin-embedded (FFPE) tissue. However, other tissue and sample types are amenable for use herein. In one embodiment, the other tissue and sample types can be fresh frozen tissue, wash fluids, or cell pellets, or the like. In one embodiment, the sample can be a bodily fluid obtained from the individual. The bodily fluid can be blood or fractions thereof (e.g., serum, plasma), urine, sputum, saliva or cerebrospinal fluid (CSF). A biomarker nucleic acid as provided herein can be extracted from a cell or can be cell free or extracted from an extracellular vesicular entity such as an exosome.


Methods are known in the art for the isolation of RNA from FFPE tissue. In one embodiment, total RNA can be isolated from FFPE tissues as described by Bibikova et al. (2004) American Journal of Pathology 165:1799-1807, herein incorporated by reference. Likewise, the High Pure RNA Paraffin Kit (Roche) can be used. Paraffin is removed by xylene extraction followed by ethanol wash. RNA can be isolated from sectioned tissue blocks using the MasterPure Purification kit (Epicenter, Madison, Wis.); a DNase I treatment step is included. RNA can be extracted from frozen samples using Trizol reagent according to the supplier's instructions (Invitrogen Life Technologies, Carlsbad, Calif.). Samples with measurable residual genomic DNA can be resubjected to DNasel treatment and assayed for DNA contamination. All purification, DNase treatment, and other steps can be performed according to the manufacturer's protocol. After total RNA isolation, samples can be stored at −80° C. until use.


General methods for mRNA extraction are well known in the art and are disclosed in standard textbooks of molecular biology, including Ausubel et al., ed., Current Protocols in Molecular Biology, John Wiley & Sons, New York 1987-1999. Methods for RNA extraction from paraffin embedded tissues are disclosed, for example, in Rupp and Locker (Lab Invest. 56:A67, 1987) and De Andres et al. (Biotechniques 18:42-44, 1995). In particular, RNA isolation can be performed using a purification kit, a buffer set and protease from commercial manufacturers, such as Qiagen (Valencia, Calif.), according to the manufacturer's instructions. For example, total RNA from cells in culture can be isolated using Qiagen RNeasy mini-columns. Other commercially available RNA isolation kits include MasterPure™. Complete DNA and RNA Purification Kit (Epicentre, Madison, Wis.) and Paraffin Block RNA Isolation Kit (Ambion, Austin, Tex.). Total RNA from tissue samples can be isolated, for example, using RNA Stat-60 (Tel-Test, Friendswood, Tex.). RNA prepared from a tumor can be isolated, for example, by cesium chloride density gradient centrifugation. Additionally, large numbers of tissue samples can readily be processed using techniques well known to those of skill in the art, such as, for example, the single-step RNA isolation process of Chomczynski (U.S. Pat. No. 4,843,155, incorporated by reference in its entirety for all purposes).


In one embodiment, a sample comprises cells harvested from a head and neck tissue sample, for example, a squamous cell carcinoma sample. Cells can be harvested from a biological sample using standard techniques known in the art. For example, in one embodiment, cells are harvested by centrifuging a cell sample and resuspending the pelleted cells. The cells can be resuspended in a buffered solution such as phosphate-buffered saline (PBS). After centrifuging the cell suspension to obtain a cell pellet, the cells can be lysed to extract nucleic acid, e.g, messenger RNA. All samples obtained from a subject, including those subjected to any sort of further processing, are considered to be obtained from the subject.


The sample, in one embodiment, is further processed before the detection of the biomarker levels of the combination of biomarkers set forth herein. For example, mRNA in a cell or tissue sample can be separated from other components of the sample. The sample can be concentrated and/or purified to isolate mRNA in its non-natural state, as the mRNA is not in its natural environment. For example, studies have indicated that the higher order structure of mRNA in vivo differs from the in vitro structure of the same sequence (see, e.g., Rouskin et al. (2014). Nature 505, pp. 701-705, incorporated herein in its entirety for all purposes).


mRNA from the sample in one embodiment, is hybridized to a synthetic DNA probe, which in some embodiments, includes a detection moiety (e.g., detectable label, capture sequence, barcode reporting sequence). Accordingly, in these embodiments, a non-natural mRNA-cDNA complex is ultimately made and used for detection of the biomarker. In another embodiment, mRNA from the sample is directly labeled with a detectable label, e.g., a fluorophore. In a further embodiment, the non-natural labeled-mRNA molecule is hybridized to a cDNA probe and the complex is detected.


In one embodiment, once the mRNA is obtained from a sample, it is converted to complementary DNA (cDNA) prior to the hybridization reaction or is used in a hybridization reaction together with one or more cDNA probes. cDNA does not exist in vivo and therefore is a non-natural molecule. Furthermore, cDNA-mRNA hybrids are synthetic and do not exist in vivo. Besides cDNA not existing in vivo, cDNA is necessarily different than mRNA, as it includes deoxyribonucleic acid and not ribonucleic acid. The cDNA is then amplified, for example, by the polymerase chain reaction (PCR) or other amplification method known to those of ordinary skill in the art. For example, other amplification methods that may be employed include the ligase chain reaction (LCR) (Wu and Wallace, Genomics, 4:560 (1989), Landegren et al., Science, 241:1077 (1988), incorporated by reference in its entirety for all purposes, transcription amplification (Kwoh et al., Proc. Natl. Acad. Sci. USA, 86:1173 (1989), incorporated by reference in its entirety for all purposes), self-sustained sequence replication (Guatelli et al., Proc. Nat. Acad. Sci. USA, 87:1874 (1990), incorporated by reference in its entirety for all purposes), incorporated by reference in its entirety for all purposes, and nucleic acid based sequence amplification (NASBA). Guidelines for selecting primers for PCR amplification are known to those of ordinary skill in the art. See, e.g., McPherson et al., PCR Basics: From Background to Bench, Springer-Verlag, 2000, incorporated by reference in its entirety for all purposes. The product of this amplification reaction, i.e., amplified cDNA is also necessarily a non-natural product. First, as mentioned above, cDNA is a non-natural molecule. Second, in the case of PCR, the amplification process serves to create hundreds of millions of cDNA copies for every individual cDNA molecule of starting material. The numbers of copies generated are far removed from the number of copies of mRNA that are present in vivo.


In one embodiment, cDNA is amplified with primers that introduce an additional DNA sequence (e.g., adapter, reporter, capture sequence or moiety, barcode) onto the fragments (e.g., with the use of adapter-specific primers), or mRNA or cDNA biomarker sequences are hybridized directly to a cDNA probe comprising the additional sequence (e.g., adapter, reporter, capture sequence or moiety, barcode). Amplification and/or hybridization of mRNA to a cDNA probe therefore serves to create non-natural double stranded molecules from the non-natural single stranded cDNA, or the mRNA, by introducing additional sequences and forming non-natural hybrids. Further, as known to those of ordinary skill in the art, amplification procedures have error rates associated with them. Therefore, amplification introduces further modifications into the cDNA molecules. In one embodiment, during amplification with the adapter-specific primers, a detectable label, e.g., a fluorophore, is added to single strand cDNA molecules. Amplification therefore also serves to create DNA complexes that do not occur in nature, at least because (i) cDNA does not exist in vivo, (i) adapter sequences are added to the ends of cDNA molecules to make DNA sequences that do not exist in vivo, (ii) the error rate associated with amplification further creates DNA sequences that do not exist in vivo, (iii) the disparate structure of the cDNA molecules as compared to what exists in nature, and (iv) the chemical addition of a detectable label to the cDNA molecules.


In some embodiments, the expression of a biomarker of interest is detected at the nucleic acid level via detection of non-natural cDNA molecules.


In some embodiments, the method for head and neck cancer SCC subtyping includes detecting expression levels of a classifier biomarker set in a sample obtained from a subject. The method can further comprise detecting expression levels of said classifier biomarker set in one or more control or reference samples. The one or more control or reference samples can be selected from a normal or HNSCC-free sample, a HNSCC AT sample, a HNSCC HPV+ AT-like sample, a HNSCC BA sample, a HNSCC MS sample, a HNSCC CL sample or any combination thereof. In some embodiments, the detecting includes all of the classifier biomarkers of Table 1 or Table 3 at the nucleic acid level or protein level. In some embodiments, the detecting includes all of the classifier biomarkers of Table 1 at the nucleic acid level or protein level. In another embodiment, a single or a subset or a plurality of the classifier biomarkers of Table 1 are detected, for example, from about 18 to about 36. For example, in one embodiment, from about 9 to about 18, from about 18 to about 36, from about 36 to about 72, from about 72 to about 108, from about 108 to about 144 of the biomarkers in Table 1 are detected in a method to determine the Head and Neck cancer SQ subtype. In another embodiment, each of the biomarkers from Table 1 is detected in a method to determine the Head and Neck cancer subtype. In another embodiment, 36 of the biomarkers from Table 1 are selected as the gene signatures for a specific Head and Neck cancer SQ subtype. In some embodiments, the detecting includes all of the classifier biomarkers of Table 3 at the nucleic acid level or protein level. In another embodiment, a single or a subset or a plurality of the classifier biomarkers of Table 3 are detected, for example, from about 10 to about 20. For example, in one embodiment, from about 5 to about 10, from about 10 to about 20, from about 20 to about 40, from about 40 to about 60, from about 60 to about 80 of the biomarkers in Table 3 are detected in a method to determine the Head and Neck cancer SQ subtype. In another embodiment, each of the biomarkers from Table 3 is detected in a method to determine the Head and Neck cancer subtype. In another embodiment, 20 of the biomarkers from Table 3 are selected as the gene signatures for a specific Head and Neck cancer SQ subtype. The detecting can be performed by any suitable technique including, but not limited to, RNA-seq, a reverse transcriptase polymerase chain reaction (RT-PCR), a microarray hybridization assay, or another hybridization assay, e.g., a NanoString assay for example, with primers and/or probes specific to the classifier biomarkers, and/or the like. In some cases, the primers useful for the amplification methods (e.g., RT-PCR or qRT-PCR) are any forward and reverse primers suitable for binding to a classifier gene provided herein, such as the classifier biomarkers listed in Table 1 or Table 3.


The biomarkers described herein include RNA comprising the entire or partial sequence of any of the nucleic acid sequences of interest, or their non-natural cDNA product, obtained synthetically in vitro in a reverse transcription reaction. The term “fragment” is intended to refer to a portion of the polynucleotide that generally comprise at least 10, 15, 20, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 550, 600, 650, 700, 800, 900, 1,000, 1,200, or 1,500 contiguous nucleotides, or up to the number of nucleotides present in a full-length biomarker polynucleotide disclosed herein. A fragment of a biomarker polynucleotide will generally encode at least 15, 25, 30, 50, 100, 150, 200, or 250 contiguous amino acids, or up to the total number of amino acids present in a full-length biomarker protein of the invention.


In some embodiments, overexpression, such as of an RNA transcript or its expression product, is determined by normalization to the level of reference RNA transcripts or their expression products, which can be all measured transcripts (or their products) in the sample or a particular reference set of RNA transcripts (or their non-natural cDNA products). Normalization is performed to correct for or normalize away both differences in the amount of RNA or cDNA assayed and variability in the quality of the RNA or cDNA used. Therefore, an assay typically measures and incorporates the expression of certain normalizing genes, including well known housekeeping genes, such as, for example, GAPDH and/or β-Actin. Alternatively, normalization can be based on the mean or median signal of all of the assayed biomarkers or a large subset thereof (global normalization approach).


Isolated mRNA can be used in hybridization or amplification assays that include, but are not limited to, Southern or Northern analyses, PCR analyses and probe arrays, NanoString Assays. One method for the detection of mRNA levels involves contacting the isolated mRNA or synthesized cDNA with a nucleic acid molecule (probe) that can hybridize to the mRNA encoded by the gene being detected. The nucleic acid probe can be, for example, a cDNA, or a portion thereof, such as an oligonucleotide of at least 7, 15, 30, 50, 100, 250, or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to the non-natural cDNA or mRNA biomarker of the present invention.


As explained above, in one embodiment, once the mRNA is obtained from a sample, it is converted to complementary DNA (cDNA) in a hybridization reaction. Conversion of the mRNA to cDNA can be performed with oligonucleotides or primers comprising sequence that is complementary to a portion of a specific mRNA. Conversion of the mRNA to cDNA can be performed with oligonucleotides or primers comprising random sequence. Conversion of the mRNA to cDNA can be performed with oligonucleotides or primers comprising sequence that is complementary to the poly(A) tail of an mRNA. cDNA does not exist in vivo and therefore is a non-natural molecule. In a further embodiment, the cDNA is then amplified, for example, by the polymerase chain reaction (PCR) or other amplification method known to those of ordinary skill in the art. PCR can be performed with the forward and/or reverse primers comprising sequence complementary to at least a portion of a classifier gene provided herein, such as the classifier biomarkers in Table 1 or Table 3. The product of this amplification reaction, i.e., amplified cDNA is necessarily a non-natural product. As mentioned above, cDNA is a non-natural molecule. Second, in the case of PCR, the amplification process serves to create hundreds of millions of cDNA copies for every individual cDNA molecule of starting material. The number of copies generated is far removed from the number of copies of mRNA that are present in vivo.


In one embodiment, cDNA is amplified with primers that introduce an additional DNA sequence (adapter sequence) onto the fragments (with the use of adapter-specific primers). The adaptor sequence can be a tail, wherein the tail sequence is not complementary to the cDNA. For example, the forward and/or reverse primers comprising sequence complementary to at least a portion of a classifier gene provided herein, such as the classifier biomarkers from Table 1 or Table 3 can comprise tail sequence. Amplification therefore serves to create non-natural double stranded molecules from the non-natural single stranded cDNA, by introducing barcode, adapter and/or reporter sequences onto the already non-natural cDNA. In one embodiment, during amplification with the adapter-specific primers, a detectable label, e.g., a fluorophore, is added to single strand cDNA molecules. Amplification therefore also serves to create DNA complexes that do not occur in nature, at least because (i) cDNA does not exist in vivo, (ii) adapter sequences are added to the ends of cDNA molecules to make DNA sequences that do not exist in vivo, (iii) the error rate associated with amplification further creates DNA sequences that do not exist in vivo, (iv) the disparate structure of the cDNA molecules as compared to what exists in nature, and (v) the chemical addition of a detectable label to the cDNA molecules.


In one embodiment, the synthesized cDNA (for example, amplified cDNA) is immobilized on a solid surface via hybridization with a probe, e.g., via a microarray. In another embodiment, cDNA products are detected via real-time polymerase chain reaction (PCR) via the introduction of fluorescent probes that hybridize with the cDNA products. For example, in one embodiment, biomarker detection is assessed by quantitative fluorogenic RT-PCR (e.g., with TaqMan® probes). For PCR analysis, well known methods are available in the art for the determination of primer sequences for use in the analysis.


Biomarkers provided herein in one embodiment, are detected via a hybridization reaction that employs a capture probe and/or a reporter probe. For example, the hybridization probe is a probe derivatized to a solid surface such as a bead, glass or silicon substrate. In another embodiment, the capture probe is present in solution and mixed with the patient's sample, followed by attachment of the hybridization product to a surface, e.g., via a biotin-avidin interaction (e.g., where biotin is a part of the capture probe and avidin is on the surface). The hybridization assay, in one embodiment, employs both a capture probe and a reporter probe. The reporter probe can hybridize to either the capture probe or the biomarker nucleic acid. Reporter probes e.g., are then counted and detected to determine the level of biomarker(s) in the sample. The capture and/or reporter probe, in one embodiment contain a detectable label, and/or a group that allows functionalization to a surface.


For example, the nCounter gene analysis system (see, e.g., Geiss et al. (2008) Nat. Biotechnol. 26, pp. 317-325, incorporated by reference in its entirety for all purposes, is amenable for use with the methods provided herein.


Hybridization assays described in U.S. Pat. Nos. 7,473,767 and 8,492,094, the disclosures of which are incorporated by reference in their entireties for all purposes, are amenable for use with the methods provided herein, i.e., to detect the biomarkers and biomarker combinations described herein.


Biomarker levels may be monitored using a membrane blot (such as used in hybridization analysis such as Northern, Southern, dot, and the like), or microwells, sample tubes, gels, beads, or fibers (or any solid support comprising bound nucleic acids). See, for example, U.S. Pat. Nos. 5,770,722, 5,874,219, 5,744,305, 5,677,195 and 5,445,934, each incorporated by reference in their entireties.


In one embodiment, microarrays are used to detect biomarker levels. Microarrays are particularly well suited for this purpose because of the reproducibility between different experiments. DNA microarrays provide one method for the simultaneous measurement of the expression levels of large numbers of genes. Each array consists of a reproducible pattern of capture probes attached to a solid support. Labeled RNA or DNA is hybridized to complementary probes on the array and then detected by laser scanning hybridization intensities for each probe on the array are determined and converted to a quantitative value representing relative gene expression levels. See, for example, U.S. Pat. Nos. 6,040,138, 5,800,992 and 6,020,135, 6,033,860, and 6,344,316, each incorporated by reference in their entireties. High-density oligonucleotide arrays are particularly useful for determining the gene expression profile for a large number of RNAs in a sample.


Techniques for the synthesis of these arrays using mechanical synthesis methods are described in, for example, U.S. Pat. No. 5,384,261. Although a planar array surface is generally used, the array can be fabricated on a surface of virtually any shape or even a multiplicity of surfaces. Arrays can be nucleic acids (or peptides) on beads, gels, polymeric surfaces, fibers (such as fiber optics), glass, or any other appropriate substrate. See, for example, U.S. Pat. Nos. 5,770,358, 5,789,162, 5,708,153, 6,040,193 and 5,800,992, each incorporated by reference in their entireties. Arrays can be packaged in such a manner as to allow for diagnostics or other manipulation of an all-inclusive device. See, for example, U.S. Pat. Nos. 5,856,174 and 5,922,591, each incorporated by reference in their entireties.


Serial analysis of gene expression (SAGE) in one embodiment is employed in the methods described herein. SAGE is a method that allows the simultaneous and quantitative analysis of a large number of gene transcripts, without the need of providing an individual hybridization probe for each transcript. First, a short sequence tag (about 10-14 bp) is generated that contains sufficient information to uniquely identify a transcript, provided that the tag is obtained from a unique position within each transcript. Then, many transcripts are linked together to form long serial molecules, that can be sequenced, revealing the identity of the multiple tags simultaneously. The expression pattern of any population of transcripts can be quantitatively evaluated by determining the abundance of individual tags, and identifying the gene corresponding to each tag. See, Velculescu et al. Science 270:484-87, 1995; Cell 88:243-51, 1997, incorporated by reference in its entirety.


An additional method of biomarker level analysis at the nucleic acid level is the use of a sequencing method, for example, RNAseq, next generation sequencing, and massively parallel signature sequencing (MPSS), as described by Brenner et al. (Nat. Biotech. 18:630-34, 2000, incorporated by reference in its entirety). This is a sequencing approach that combines non-gel-based signature sequencing with in vitro cloning of millions of templates on separate 5 μm diameter microbeads. First, a microbead library of DNA templates is constructed by in vitro cloning. This is followed by the assembly of a planar array of the template-containing microbeads in a flow cell at a high density (typically greater than 3.0×106 microbeads/cm2). The free ends of the cloned templates on each microbead are analyzed simultaneously, using a fluorescence-based signature sequencing method that does not require DNA fragment separation. This method has been shown to simultaneously and accurately provide, in a single operation, hundreds of thousands of gene signature sequences from a yeast cDNA library.


Another method of biomarker level analysis at the nucleic acid level is the use of an amplification method such as, for example, RT-PCR or quantitative RT-PCR (qRT-PCR). Methods for determining the level of biomarker mRNA in a sample may involve the process of nucleic acid amplification, e.g., by RT-PCR (the experimental embodiment set forth in Mullis, 1987, U.S. Pat. No. 4,683,202), ligase chain reaction (Barany (1991) Proc. Natl. Acad. Sci. USA 88:189-193), self-sustained sequence replication (Guatelli et al. (1990) Proc. Natl. Acad. Sci. USA 87:1874-1878), transcriptional amplification system (Kwoh et al. (1989) Proc. Natl. Acad. Sci. USA 86:1173-1177), Q-Beta Replicase (Lizardi et al. (1988) Bio/Technology 6:1197), rolling circle replication (Lizardi et al., U.S. Pat. No. 5,854,033) or any other nucleic acid amplification method, followed by the detection of the amplified molecules using techniques well known to those of skill in the art. Numerous different PCR or qRT-PCR protocols are known in the art and can be directly applied or adapted for use using the presently described compositions for the detection and/or quantification of expression of discriminative genes in a sample. See, for example, Fan et al. (2004) Genome Res. 14:878-885, herein incorporated by reference. Generally, in PCR, a target polynucleotide sequence is amplified by reaction with at least one oligonucleotide primer or pair of oligonucleotide primers. The primer(s) hybridize to a complementary region of the target nucleic acid and a DNA polymerase extends the primer(s) to amplify the target sequence. Under conditions sufficient to provide polymerase-based nucleic acid amplification products, a nucleic acid fragment of one size dominates the reaction products (the target polynucleotide sequence which is the amplification product). The amplification cycle is repeated to increase the concentration of the single target polynucleotide sequence. The reaction can be performed in any thermocycler commonly used for PCR.


Quantitative RT-PCR (qRT-PCR) (also referred as real-time RT-PCR) is preferred under some circumstances because it provides not only a quantitative measurement, but also reduced time and contamination. As used herein, “quantitative PCR” (or “real time qRT-PCR”) refers to the direct monitoring of the progress of a PCR amplification as it is occurring without the need for repeated sampling of the reaction products. In quantitative PCR, the reaction products may be monitored via a signaling mechanism (e.g., fluorescence) as they are generated and are tracked after the signal rises above a background level but before the reaction reaches a plateau. The number of cycles required to achieve a detectable or “threshold” level of fluorescence varies directly with the concentration of amplifiable targets at the beginning of the PCR process, enabling a measure of signal intensity to provide a measure of the amount of target nucleic acid in a sample in real time. A DNA binding dye (e.g., SYBR green) or a labeled probe can be used to detect the extension product generated by PCR amplification. Any probe format utilizing a labeled probe comprising the sequences of the invention may be used.


Immunohistochemistry methods are also suitable for detecting the levels of the biomarkers of the present invention. Samples can be frozen for later preparation or immediately placed in a fixative solution. Tissue samples can be fixed by treatment with a reagent, such as formalin, gluteraldehyde, methanol, or the like and embedded in paraffin. Methods for preparing slides for immunohistochemical analysis from formalin-fixed, paraffin-embedded tissue samples are well known in the art.


In one embodiment, the levels of the biomarkers provided herein, such as the classifier biomarkers of Table 1 (or subsets thereof, for example 18 to 36, 36 to 54, 54 to 72, 72 to 90, 90 to 108, 108 to 126, or 126 to 144 biomarkers), are normalized against the expression levels of all RNA transcripts or their non-natural cDNA expression products, or protein products in the sample, or of a reference set of RNA transcripts or a reference set of their non-natural cDNA expression products, or a reference set of their protein products in the sample. In one embodiment, the levels of the biomarkers provided herein, such as the classifier biomarkers of Table 3 (or subsets thereof, for example 10 to 20, 20 to 30, 30 to 40, 40 to 50, 50 to 60, 60 to 70, or 70 to 80 biomarkers), are normalized against the expression levels of all RNA transcripts or their non-natural cDNA expression products, or protein products in the sample, or of a reference set of RNA transcripts or a reference set of their non-natural cDNA expression products, or a reference set of their protein products in the sample.


In one embodiment, HNSCC subtypes can be evaluated using levels of protein expression of one or more of the classifier genes provided herein, such as the classifier biomarkers listed in Table 1 or Table 3. The level of protein expression can be measured using an immunological detection method. Immunological detection methods which can be used herein include, but are not limited to, competitive and non-competitive assay systems using techniques such as Western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, protein A immunoassays, and the like. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. I, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety).


In one embodiment, antibodies specific for biomarker proteins are utilized to detect the expression of a biomarker protein in a body sample. The method comprises obtaining a body sample from a patient or a subject, contacting the body sample with at least one antibody directed to a biomarker that is selectively expressed in Head and Neck cancer cells, and detecting antibody binding to determine if the biomarker is expressed in the patient sample. In one aspect of the present invention provided is an immunocytochemistry technique for diagnosing Head and Neck cancer subtypes. One of skill in the art will recognize that the immunocytochemistry method described herein below may be performed manually or in an automated fashion.


As provided throughout, the methods set forth herein provide a method for determining the Head and Neck cancer SCC subtype of a patient. Once the biomarker levels are determined, for example by measuring non-natural cDNA biomarker levels or non-natural mRNA-cDNA biomarker complexes, the biomarker levels are compared to reference values or a reference sample as provided herein, for example with the use of statistical methods or direct comparison of detected levels, to make a determination of the Head and Neck cancer molecular SCC subtype. Based on the comparison, the patient's Head and Neck cancer sample is SCC classified, e.g., as BA, MS, AT or CL.


In one embodiment, expression level values of the at least one classifier biomarkers provided herein, such as the classifier biomarkers of Table 1 are compared to reference expression level value(s) from at least one sample training set, wherein the at least one sample training set comprises expression level values from a reference sample(s). In a further embodiment, the at least one sample training set comprises expression level values of the at least one classifier biomarkers provided herein, such as the classifier biomarkers of Table 1 or Table 3 from a HNSCC BA, HNSCC MS, HNSCC AT, HNSCC CL, or HNSCC-free sample or a combination thereof.


In a separate embodiment, hybridization values of the at least one classifier biomarkers provided herein, such as the classifier biomarkers of Table 1 or Table 3 are compared to reference hybridization value(s) from at least one sample training set, wherein the at least one sample training set comprises hybridization values from a reference sample(s). In a further embodiment, the at least one sample training set comprises hybridization values of the at least one classifier biomarkers provided herein, such as the classifier biomarkers of Table 1 or Table 3 from a HNSCC BA, HNSCC MS, HNSCC AT, HNSCC CL, or HNSCC-free sample, or a combination thereof. Methods for comparing detected levels of biomarkers to reference values and/or reference samples are provided herein. Based on this comparison, in one embodiment a correlation between the biomarker levels obtained from the subject's sample and the reference values is obtained. An assessment of the Head and Neck cancer SCC subtype is then made.


Various statistical methods can be used to aid in the comparison of the biomarker levels obtained from the patient and reference biomarker levels, for example, from at least one sample training set.


In one embodiment, a supervised pattern recognition method is employed. Examples of supervised pattern recognition methods can include, but are not limited to, the nearest centroid methods (Dabney (2005) Bioinformatics 21(22):4148-4154 and Tibshirani et al. (2002) Proc. Natl. Acad. Sci. USA 99(10):6576-6572); soft independent modeling of class analysis (SIMCA) (see, for example, Wold, 1976); partial least squares analysis (PLS) (see, for example, Wold, 1966; Joreskog, 1982; Frank, 1984; Bro, R., 1997); linear descriminant analysis (LDA) (see, for example, Nillson, 1965); K-nearest neighbour analysis (KNN) (sec, for example, Brown et al., 1996); artificial neural networks (ANN) (see, for example, Wasserman, 1989; Anker et al., 1992; Hare, 1994); probabilistic neural networks (PNNs) (see, for example, Parzen, 1962; Bishop, 1995; Speckt, 1990; Broomhead et al., 1988; Patterson, 1996); rule induction (RI) (see, for example, Quinlan, 1986); and, Bayesian methods (see, for example, Bretthorst, 1990a, 1990b, 1988). In one embodiment, the classifier for identifying tumor subtypes based on gene expression data is the centroid based method described in Mullins et al. (2007) Clin Chem. 53(7):1273-9, each of which is herein incorporated by reference in its entirety.


In other embodiments, an unsupervised training approach is employed, and therefore, no training set is used.


Referring to sample training sets for supervised learning approaches again, in some embodiments, a sample training set(s) can include expression data of a plurality or all of the classifier biomarkers (e.g., all the classifier biomarkers of Table 1 or Table 3) from a HNSCC sample. The plurality of classifier biomarkers can comprise at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1. The plurality of classifier biomarkers can comprise at least two classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3. In some embodiments, the sample training set(s) are normalized to remove sample-to-sample variation.


In some embodiments, comparing can include applying a statistical algorithm, such as, for example, any suitable multivariate statistical analysis model, which can be parametric or non-parametric. In some embodiments, applying the statistical algorithm can include determining a correlation between the expression data obtained from the human head and neck tissue sample and the expression data from the HNSCC training set(s). In some embodiments, cross-validation is performed, such as (for example), leave-one-out cross-validation (LOOCV). In some embodiments, integrative correlation is performed. In some embodiments, a Spearman correlation is performed. In some embodiments, a centroid based method is employed for the statistical algorithm as described in Mullins et al. (2007) Clin Chem. 53(7):1273-9, and based on gene expression data, which is herein incorporated by reference in its entirety.


Results of the gene expression performed on a sample from a subject (test sample) may be compared to a biological sample(s) or data derived from a biological sample(s) that is known or suspected to be normal (“reference sample” or “normal sample”, e.g., non-HNSCC sample). In some embodiments, a reference sample or reference gene expression data is obtained or derived from an individual known to have a particular molecular subtype of SCC, i.e., BA, MS, AT or CL.


The reference sample may be assayed at the same time, or at a different time from the test sample. Alternatively, the biomarker level information from a reference sample may be stored in a database or other means for access at a later date.


The biomarker level results of an assay on the test sample may be compared to the results of the same assay on a reference sample. In some cases, the results of the assay on the reference sample are from a database, or a reference value(s). In some cases, the results of the assay on the reference sample are a known or generally accepted value or range of values by those skilled in the art. In some cases, the comparison is qualitative. In other cases, the comparison is quantitative. In some cases, qualitative or quantitative comparisons may involve but are not limited to one or more of the following: comparing fluorescence values, spot intensities, absorbance values, chemiluminescent signals, histograms, critical threshold values, statistical significance values, expression levels of the genes described herein, mRNA copy numbers.


In one embodiment, an odds ratio (OR) is calculated for each biomarker level panel measurement. Here, the OR is a measure of association between the measured biomarker values for the patient and an outcome, e.g., HNSCC subtype. For example, see, J Can. Acad. Child Adolesc. Psychiatry 2010; 19(3): 227-229, which is incorporated by reference in its entirety for all purposes.


In one embodiment, a specified statistical confidence level may be determined in order to provide a confidence level regarding the Head and Neck cancer subtype. For example, it may be determined that a confidence level of greater than 90% may be a useful predictor of the Head and Neck cancer subtype. In other embodiments, more or less stringent confidence levels may be chosen. For example, a confidence level of about or at least about 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 97.5%, 99%, 99.5%, or 99.9% may be chosen. The confidence level provided may in some cases be related to the quality of the sample, the quality of the data, the quality of the analysis, the specific methods used, and/or the number of gene expression values (i.e., the number of genes) analyzed. The specified confidence level for providing the likelihood of response may be chosen on the basis of the expected number of false positives or false negatives. Methods for choosing parameters for achieving a specified confidence level or for identifying markers with diagnostic power include but are not limited to Receiver Operating Characteristic (ROC) curve analysis, binormal ROC, principal component analysis, odds ratio analysis, partial least squares analysis, singular value decomposition, least absolute shrinkage and selection operator analysis, least angle regression, and the threshold gradient directed regularization method.


Determining the HNSCC subtype in some cases can be improved through the application of algorithms designed to normalize and or improve the reliability of the gene expression data. In some embodiments of the present invention, the data analysis utilizes a computer or other device, machine or apparatus for application of the various algorithms described herein due to the large number of individual data points that are processed. A “machine learning algorithm” refers to a computational-based prediction methodology, also known to persons skilled in the art as a “classifier,” employed for characterizing a gene expression profile or profiles, e.g., to determine the HNSCC subtype. The biomarker levels, determined by, e.g., microarray-based hybridization assays, sequencing assays, NanoString assays, etc., are in one embodiment subjected to the algorithm in order to classify the profile. Supervised learning generally involves “training” a classifier to recognize the distinctions among subtypes such as BA positive, MS positive, AT positive or CL positive, and then “testing” the accuracy of the classifier on an independent test set. Therefore, for new, unknown samples the classifier can be used to predict, for example, the class (e.g., BA vs. MS vs. AT vs. CL) in which the samples belong.


In some embodiments, a robust multi-array average (RMA) method may be used to normalize raw data. The RMA method begins by computing background-corrected intensities for each matched cell on a number of microarrays. In one embodiment, the background corrected values are restricted to positive values as described by Irizarry et al. (2003). Biostatistics April 4 (2): 249-64, incorporated by reference in its entirety for all purposes. After background correction, the base-2 logarithm of each background corrected matched-cell intensity is then obtained. The background corrected, log-transformed, matched intensity on each microarray is then normalized using the quantile normalization method in which for each input array and each probe value, the array percentile probe value is replaced with the average of all array percentile points, this method is more completely described by Bolstad et al. Bioinformatics 2003, incorporated by reference in its entirety. Following quantile normalization, the normalized data may then be fit to a linear model to obtain an intensity measure for each probe on each microarray. Tukey's median polish algorithm (Tukey, J. W., Exploratory Data Analysis. 1977, incorporated by reference in its entirety for all purposes) may then be used to determine the log-scale intensity level for the normalized probe set data.


Various other software programs may be implemented. In certain methods, feature selection and model estimation may be performed by logistic regression with lasso penalty using glmnet (Friedman et al. (2010). Journal of statistical software 33(1): 1-22, incorporated by reference in its entirety). Raw reads may be aligned using TopHat (Trapnell et al. (2009). Bioinformatics 25(9): 1105-11, incorporated by reference in its entirety). In methods, top features (N ranging from 10 to 200) are used to train a linear support vector machine (SVM) (Suykens J A K, Vandewalle J. Least Squares Support Vector Machine Classifiers. Neural Processing Letters 1999; 9(3): 293-300, incorporated by reference in its entirety) using the e1071 library (Meyer D. Support vector machines: the interface to libsvm in package e1071. 2014, incorporated by reference in its entirety). Confidence intervals, in one embodiment, are computed using the pROC package (Robin X, Turck N, Hainard A, et al. pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC bioinformatics 2011; 12: 77, incorporated by reference in its entirety).


In addition, data may be filtered to remove data that may be considered suspect. In one embodiment, data derived from microarray probes that have fewer than about 4, 5, 6, 7 or 8 guanosine+cytosine nucleotides may be considered to be unreliable due to their aberrant hybridization propensity or secondary structure issues. Similarly, data deriving from microarray probes that have more than about 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, or 22 guanosine+cytosine nucleotides may in one embodiment be considered unreliable due to their aberrant hybridization propensity or secondary structure issues.


In some embodiments of the present invention, data from probe-sets may be excluded from analysis if they are not identified at a detectable level (above background).


In some embodiments of the present disclosure, probe-sets that exhibit no, or low variance may be excluded from further analysis. Low-variance probe-sets are excluded from the analysis via a Chi-Square test. In one embodiment, a probe-set is considered to be low-variance if its transformed variance is to the left of the 99 percent confidence interval of the Chi-Squared distribution with (N−1) degrees of freedom. (N−1)*Probe-set Variance/(Gene Probe-set Variance). Chi-Sq(N−1) where N is the number of input CEL files, (N−1) is the degrees of freedom for the Chi-Squared distribution, and the “probe-set variance for the gene” is the average of probe-set variances across the gene. In some embodiments of the present invention, probe-sets for a given mRNA or group of mRNAs may be excluded from further analysis if they contain less than a minimum number of probes that pass through the previously described filter steps for GC content, reliability, variance and the like. For example in some embodiments, probe-sets for a given gene or transcript cluster may be excluded from further analysis if they contain less than about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or less than about 20 probes.


Methods of biomarker level data analysis in one embodiment, further include the use of a feature selection algorithm as provided herein. In some embodiments of the present invention, feature selection is provided by use of the LIMMA software package (Smyth, G. K. (2005). Limma: linear models for microarray data. In: Bioinformatics and Computational Biology Solutions using R and Bioconductor, R. Gentleman, V. Carey, S. Dudoit, R. Irizarry, W. Huber (eds.), Springer, New York, pages 397-420, incorporated by reference in its entirety for all purposes).


Methods of biomarker level data analysis, in one embodiment, include the use of a pre-classifier algorithm. For example, an algorithm may use a specific molecular fingerprint to pre-classify the samples according to their composition and then apply a correction/normalization factor. This data/information may then be fed in to a final classification algorithm which would incorporate that information to aid in the final diagnosis.


Methods of biomarker level data analysis, in one embodiment, further include the use of a classifier algorithm as provided herein. In one embodiment of the present invention, a diagonal linear discriminant analysis, k-nearest neighbor algorithm, support vector machine (SVM) algorithm, linear support vector machine, random forest algorithm, or a probabilistic model-based method or a combination thereof is provided for classification of microarray data. In some embodiments, identified markers that distinguish samples (e.g., of varying biomarker level profiles, and/or varying molecular subtypes of HNSCC (e.g., basal, mesemchymal, atypical, classical)) are selected based on statistical significance of the difference in biomarker levels between classes of interest. In some cases, the statistical significance is adjusted by applying a Benjamin Hochberg or another correction for false discovery rate (FDR).


In some cases, the classifier algorithm may be supplemented with a meta-analysis approach such as that described by Fishel and Kaufman et al. 2007 Bioinformatics 23(13): 1599-606, incorporated by reference in its entirety for all purposes. In some cases, the classifier algorithm may be supplemented with a meta-analysis approach such as a repeatability analysis.


Methods for deriving and applying posterior probabilities to the analysis of biomarker level data are known in the art and have been described for example in Smyth, G. K. 2004 Stat. Appi. Genet. Mol. Biol. 3: Article 3, incorporated by reference in its entirety for all purposes. In some cases, the posterior probabilities may be used in the methods of the present invention to rank the markers provided by the classifier algorithm.


A statistical evaluation of the results of the biomarker level profiling may provide a quantitative value or values indicative of one or more of the following: molecular subtype of HNSCC (e.g., basal, mesenchymal, atypical, classical); the likelihood of the success of a particular therapeutic intervention, e.g., angiogenesis inhibitor therapy, chemotherapy, or immunotherapy. In one embodiment, the data is presented directly to the physician in its most useful form to guide patient care, or is used to define patient populations in clinical trials or a patient population for a given medication. The results of the molecular profiling can be statistically evaluated using a number of methods known to the art including, but not limited to: the students T test, the two sided T test, Pearson rank sum analysis, hidden Markov model analysis, analysis of q-q plots, principal component analysis, one way ANOVA, two way ANOVA, LIMMA and the like.


In some cases, accuracy may be determined by tracking the subject over time to determine the accuracy of the original diagnosis. In other cases, accuracy may be established in a deterministic manner or using statistical methods. For example, receiver operator characteristic (ROC) analysis may be used to determine the optimal assay parameters to achieve a specific level of accuracy, specificity, positive predictive value, negative predictive value, and/or false discovery rate.


In some cases, the results of the biomarker level profiling assays, are entered into a database for access by representatives or agents of a molecular profiling business, the individual, a medical provider, or insurance provider. In some cases, assay results include sample classification, identification, or diagnosis by a representative, agent or consultant of the business, such as a medical professional. In other cases, a computer or algorithmic analysis of the data is provided automatically. In some cases the molecular profiling business may bill the individual, insurance provider, medical provider, researcher, or government entity for one or more of the following: molecular profiling assays performed, consulting services, data analysis, reporting of results, or database access.


In some embodiments of the present invention, the results of the biomarker level profiling assays are presented as a report on a computer screen or as a paper record. In some embodiments, the report may include, but is not limited to, such information as one or more of the following: the levels of biomarkers (e.g., as reported by copy number or fluorescence intensity, etc.) as compared to the reference sample or reference value(s); the likelihood the subject will respond to a particular therapy, based on the biomarker level values and the HNSCC subtype and proposed therapies.


In one embodiment, the results of the gene expression profiling may be classified into one or more of the following: basal positive, mesenchymal positive, atypical positive or classical positive, basal negative, mesenchymal negative, atypical negative or classical negative; likely to respond to surgery (e.g., neck dissection), radiotherapy, angiogenesis inhibitor, immunotherapy or chemotherapy; unlikely to respond to surgery (e.g., neck dissection), radiotherapy, angiogenesis inhibitor, immunotherapy or chemotherapy; or a combination thereof. In a further embodiment, the results of the gene expression profiling may be further classified into being HPV positive or HPV negative.


In some embodiments of the present invention, results are classified using a trained algorithm. Trained algorithms of the present invention include algorithms that have been developed using a reference set of known gene expression values and/or normal samples, for example, samples from individuals diagnosed with a particular molecular subtype of HNSCC. In some cases, a reference set of known gene expression values are obtained from individuals who have been diagnosed with a particular molecular subtype of HNSCC, and are also known to respond (or not respond) to angiogenesis inhibitor therapy. In some cases, a reference set of known gene expression values are obtained from individuals who have been diagnosed with a particular molecular subtype of HNSCC, and are also known to respond (or not respond) to immunotherapy. In some cases, a reference set of known gene expression values are obtained from individuals who have been diagnosed with a particular molecular subtype of HNSCC, and are also known to respond (or not respond) to chemotherapy. In some cases, the reference sets described above are HPV positive. In some cases, the reference sets described above are HPV negative.


Algorithms suitable for categorization of samples include but are not limited to k-nearest neighbor algorithms, support vector machines, linear discriminant analysis, diagonal linear discriminant analysis, updown, naive Bayesian algorithms, neural network algorithms, hidden Markov model algorithms, genetic algorithms, or any combination thereof.


When a binary classifier is compared with actual true values (e.g., values from a biological sample), there are typically four possible outcomes. If the outcome from a prediction is p (where “p” is a positive classifier output, such as the presence of a deletion or duplication syndrome) and the actual value is also p, then it is called a true positive (TP); however if the actual value is n then it is said to be a false positive (FP). Conversely, a true negative has occurred when both the prediction outcome and the actual value are n (where “n” is a negative classifier output, such as no deletion or duplication syndrome), and false negative is when the prediction outcome is n while the actual value is p. In one embodiment, consider a test that seeks to determine whether a person is likely or unlikely to respond to angiogenesis inhibitor therapy. A false positive in this case occurs when the person tests positive, but actually does respond. A false negative, on the other hand, occurs when the person tests negative, suggesting they are unlikely to respond, when they actually are likely to respond. The same holds true for classifying a Head and Neck cancer subtype.


The positive predictive value (PPV), or precision rate, or post-test probability of disease, is the proportion of subjects with positive test results who are correctly diagnosed as likely or unlikely to respond, or diagnosed with the correct Head and Neck cancer subtype, or a combination thereof. It reflects the probability that a positive test reflects the underlying condition being tested for. Its value does however depend on the prevalence of the disease, which may vary. In one example the following characteristics are provided: FP (false positive); TN (true negative); TP (true positive); FN (false negative). False positive rate (□)=FP/(FP+TN)−specificity; False negative rate (□)=FN/(TP+FN)−sensitivity; Power=sensitivity=1−□□; Likelihood-ratio positive=sensitivity/(1−specificity); Likelihood-ratio negative=(1−sensitivity)/specificity. The negative predictive value (NPV) is the proportion of subjects with negative test results who are correctly diagnosed.


In some embodiments, the results of the biomarker level analysis of the subject methods provide a statistical confidence level that a given diagnosis is correct. In some embodiments, such statistical confidence level is at least about, or more than about 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% 99.5%, or more.


In some embodiments, the method further includes classifying the Head and Neck tissue sample as a particular Head and Neck cancer subtype based on the comparison of biomarker levels in the sample and reference biomarker levels, for example present in at least one training set. In some embodiments, the Head and Neck tissue sample is classified as a particular subtype if the results of the comparison meet one or more criterion such as, for example, a minimum percent agreement, a value of a statistic calculated based on the percentage agreement such as (for example) a kappa statistic, a minimum correlation (e.g., Pearson's correlation) and/or the like.


It is intended that the methods described herein can be performed by software (stored in memory and/or executed on hardware), hardware, or a combination thereof. Hardware modules may include, for example, a general-purpose processor, a field programmable gate array (FPGA), and/or an application specific integrated circuit (ASIC). Software modules (executed on hardware) can be expressed in a variety of software languages (e.g., computer code), including Unix utilities, C, C++, Java™, Ruby, SQL, SAS®, the R programming language/software environment, Visual Basic™, and other object-oriented, procedural, or other programming language and development tools. Examples of computer code include, but are not limited to, micro-code or micro-instructions, machine instructions, such as produced by a compiler, code used to produce a web service, and files containing higher-level instructions that are executed by a computer using an interpreter. Additional examples of computer code include, but are not limited to, control signals, encrypted code, and compressed code.


Some embodiments described herein relate to devices with a non-transitory computer-readable medium (also can be referred to as a non-transitory processor-readable medium or memory) having instructions or computer code thereon for performing various computer-implemented operations and/or methods disclosed herein. The computer-readable medium (or processor-readable medium) is non-transitory in the sense that it does not include transitory propagating signals per se (e.g., a propagating electromagnetic wave carrying information on a transmission medium such as space or a cable). The media and computer code (also can be referred to as code) may be those designed and constructed for the specific purpose or purposes. Examples of non-transitory computer-readable media include, but are not limited to: magnetic storage media such as hard disks, floppy disks, and magnetic tape; optical storage media such as Compact Disc/Digital Video Discs (CD/DVDs), Compact Disc-Read Only Memories (CD-ROMs), and holographic devices; magneto-optical storage media such as optical disks; carrier wave signal processing modules; and hardware devices that are specially configured to store and execute program code, such as Application-Specific Integrated Circuits (ASICs), Programmable Logic Devices (PLDs), Read-Only Memory (ROM) and Random-Access Memory (RAM) devices. Other embodiments described herein relate to a computer program product, which can include, for example, the instructions and/or computer code discussed herein.


In some embodiments, a single biomarker, or from about 18 to about 36, from about 36 to about 54, from about 54 to about 72, from about 72 to about 90, from about 90 to about 108, from about 108 to about 126, from about 126 to about 144, from about 36 to about 72, from about 36 to about 108, from about 36 to about 144 biomarkers (e.g., as disclosed in Table 1) is capable of classifying subtypes of HNSCC with a predictive success of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between. In some embodiments, any combination of biomarkers disclosed herein (e.g., in Table 1) can be used to obtain a predictive success of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between.


In some embodiments, a single biomarker, or from about 10 to about 20, from about 20 to about 30, from about 20 to about 40, from about 40 to about 50, from about 40 to about 60, from about 60 to about 70, from about 60 to about 80, from about 20 to about 60, from about 20 to about 80, from about 40 to about 80 biomarkers (e.g., as disclosed in Table 3) is capable of classifying subtypes of HNSCC with a predictive success of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between. In some embodiments, any combination of biomarkers disclosed herein (e.g., in Table 3) can be used to obtain a predictive success of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between.


In some embodiments, a single biomarker, or from about 18 to about 36, from about 36 to about 54, from about 54 to about 72, from about 72 to about 90, from about 90 to about 108, from about 108 to about 126, from about 126 to about 144, from about 36 to about 72, from about 36 to about 108, from about 36 to about 144 biomarkers (e.g., as disclosed in Table 1) is capable of classifying subtypes of HNSCC with a sensitivity or specificity of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between. In some embodiments, any combination of biomarkers disclosed herein can be used to obtain a sensitivity or specificity of at least about 70%, at least about 71%, at least about 72%, about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between.


In some embodiments, a single biomarker, or from about 10 to about 20, from about 20 to about 30, from about 20 to about 40, from about 40 to about 50, from about 40 to about 60, from about 60 to about 70, from about 60 to about 80, from about 20 to about 60, from about 20 to about 80, from about 40 to about 80 biomarkers (e.g., as disclosed in Table 3) is capable of classifying subtypes of HNSCC with a sensitivity or specificity of at least about 70%, at least about 71%, at least about 72%, at least about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between. In some embodiments, any combination of biomarkers disclosed herein can be used to obtain a sensitivity or specificity of at least about 70%, at least about 71%, at least about 72%, about 73%, at least about 74%, at least about 75%, at least about 76%, at least about 77%, at least about 78%, at least about 79%, at least about 80%, at least about 81%, at least about 82%, at least about 83%, at least about 84%, at least about 85%, at least about 86%, at least about 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, up to 100%, and all values in between.


Classifier Gene Selection

In one embodiment, the methods and compositions provided herein are useful for determining the HNSCC subtype of a sample (e.g., Head and Neck tissue sample) from a patient by analyzing the expression of a set of biomarkers, whereby the set of biomarkers comprise a fewer number of biomarkers that methods known in the art for molecularly classifying HNSCC subtype. In some cases, the set of biomarkers is less than 250, 240, 230, 220, 210, 200, 150, 100, 95 or 90 biomarkers. In some cases, the set of biomarkers is less than 150 biomarkers. In some cases, the set of biomarkers is the set of 144 biomarkers listed in Table 1. In some cases, the set of biomarkers is a sub-set of biomarkers listed Table 1 such as, for example, the 80 biomarkers of Table 3. The biomarkers or classifier genes useful in the methods and compositions provided herein can be selected from one or more HNSCC datasets from one or more databases. The databases can be public databases. In one embodiment, classifier genes (e.g., one or more genes listed in Table 1 and Table 3) useful in the methods and compositions provided herein for detecting or diagnosing HNSCC subtypes were selected from a HNSCC RNAseq dataset from The Cancer Genome Atlas (TCGA). In one embodiment, classifier genes useful for the methods and compositions provided herein such as those in Table 1 are selected by subjecting a large set of classifier genes to an in silico based process in order to determine the minimum number of genes whose expression profile can be used to determine an HNSCC subtype of sample obtained from a subject. In some cases, the large set of classifier genes can be a HNSCC RNAseq dataset such as, for example, from TCGA. In some cases, the large set of classifier genes can be 840-gene classifier described herein, whereby the 840-gene classifier can serve to define gold standard subtype. The in silico process for selecting a gene cassette as provided herein for determining HNSCC subtype of a sample from a patient can comprise, applying or using a Classifying arrays to Nearest Centroid (CLaNC) algorithm with modification on the standard 840 classifier genes to choose an equal number of negatively and positively correlated genes for each subtype. For determination of the optimal number of genes (e.g, 36 per subtype as shown in Table 1 or 20 per subtype as shown in Table 3) to include in the signature, the process can further comprise performing a 5-fold cross validation using TCGA HNSCC dataset as provided herein to produce cross-validation curves as shown in FIG. 1. To get the final list of gene classifiers, the method can further comprise applying the Classifying arrays to Nearest Centroid (CLaNC) to the entire TCGA data set minus 20% of samples with the lowest gold standard subtype prediction strength, and removing an equal number from each subtype.


In one embodiment, the method further comprises validating the gene classifiers. Validation can comprise testing the expression of the classifiers in several fresh frozen publicly available array and RNAseq datasets and calling the subtype based on said expression levels and subsequently comparing the expression with the gold standard subtype calls as defined by the previously published 840-gene signature. Final validation of the gene signature (e.g., Table 1 or Table 3) can then be performed in a newly collected RNAseq dataset of archived formalin-fixed paraffin-embedded (FFPE) HNSCC samples to assure comparable performance in the FFPE samples. In one embodiment, the classifier biomarkers of Table 1 or Table 3 were selected based on the in silico CLaNC process described herein. The gene symbols and official gene names are listed in column 2 and column 3, respectively of Tables 1 and 3.


In one embodiment, the methods of the invention require the detection of at least 1, 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 18, at least 20, at least 22, at least 24, at least 26, at least 28, at least 30, at least 32, at least 34 or up to 36 classifier biomarkers in a Head and Neck cancer cell sample obtained from a patient which expression is altered in order to identify a BA, MS, AT or CL HNSCC subtype. The same applies for other classifier gene expression datasets as provided herein.


In another embodiment, the methods of the invention require the detection of a total of at least 1, at least 2, at least 5, at least 8, at least 10, at least 18, at least 36, at least 54, at least 72, at least 108, at least 126, or up to 144 classifier biomarkers out of the 144 gene biomarkers of Table 1 in a Head and Neck cancer cell sample (e.g., HNSCC sample) obtained from a patient in order to identify a BA, MS, AT or CL HNSCC subtype. In another embodiment, the methods of the invention require the detection of a total of at least 1, at least 2, at least 5, at least 10, at least 20, at least 40, at least 60 or up to 80 classifier biomarkers out of the 80 gene biomarkers of Table 3 in a Head and Neck cancer cell sample (e.g., HNSCC sample) obtained from a patient in order to identify a basal, classical, atypical or mesenchymal Head and Neck squamous cell carcinoma subtype. The same applies for other classifier gene expression datasets as provided herein.


In one embodiment, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 10, at least 18, at least 36, at least 54, at least 72, at least 108, at least 126, or up to 144 classifier biomarkers out of the 144 gene biomarkers of Table 1 are “up-regulated” in a specific subtype of HNSCC. In another embodiment, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 10, at least 18, at least 36, at least 54, at least 72, at least 108, at least 126, or up to 144 classifier biomarkers out of the 144 gene biomarkers of Table 1 are “down-regulated” in a specific subtype of HNSCC. In one embodiment, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 or up to 10 biomarkers of Table 3 are “up-regulated” in a specific subtype of head and neck squamous cell carcinoma. In another embodiment, at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9 or up to 10 biomarkers of Table 3 are “down-regulated” in a specific subtype of head and neck squamous cell carcinoma. The same applies for other classifier gene expression datasets as provided herein.


In one embodiment, the expression level of an “up-regulated” biomarker as provided herein is increased by about 0.5-fold, about 1-fold, about 1.5-fold, about 2-fold, about 2.5-fold, about 3-fold, about 3.5-fold, about 4-fold, about 4.5-fold, about 5-fold, and any values in between. In another embodiment, the expression level of a “down-regulated” biomarker as provided herein is decreased by about 0.8-fold, about 1.4-fold, about 2-fold, about 2.6-fold, about 3.2-fold, about 3.6-fold, about 4-fold, and any values in between.


It is recognized that additional genes or proteins can be used in the practice of the invention. In general, genes useful in classifying the subtypes of HNSCC, include those that are independently capable of distinguishing between normal versus tumor, or between different classes or grades of HNSCC. A gene is considered to be capable of reliably distinguishing between subtypes if the area under the receiver operator characteristic (ROC) curve is approximately 1.


HPV Status

In one embodiment, the Human Papillomavirus (HPV) status of a subject is determined. The HPV status of the subject can be determined using any of the HPV-specific tests known in the art, alone or in combination, as described in Lewis Jr et al., (2018) Human Papillomavirus Testing in Head and Neck Carcinomas: Guideline From the College of American Pathologists. Archives of Pathology & Laboratory Medicine: May 2018, Vol. 142, No. 5, pp. 559-597, or Venuti A, Paolini F. HPV Detection Methods in Head and Neck Cancer. Head and Neck Pathology. 2012; 6(Suppl 1):63-74, each of which is herein incorporated by reference. Use of any of the known HPV-specific tests can also be used in combination with examining the surrogate marker p16 by immunohistochemistry (IHC) and/or examining hematoxylin-eosin morphology of tissue samples from the subject. The surrogate marker p16 can be markedly overexpressed in tumor cells with transcriptionally active HPV because the viral E7 oncoprotein destabilizes pRb, functionally removing suppression of p16 expression and allowing tumor cells with high p16 levels to bypass pRb-dependent cell cycle arrest as described in Moody C A. Laimins L A. Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer. 2010; 10(8):550-560 and Munger K. Baldwin A. Edwards K M. et al. Mechanisms of human papillomavirus-induced oncogenesis. J Virol. 2004; 78(21):11451-11460. The result can be marked overexpression of p16, which can make it an excellent surrogate marker of viral infection in the correct context. Results obtained from use of the HPV-specific tests and/or examination of p16 IHC and/or hematoxylin-eosin morphology can indicate that the subject is experiencing ongoing HPV replication. As a result, said subject can be said to be HPV positive.


In one embodiment, the HPV status of a subject is assessed by determining the presence, absence or level of expression of one or more genes or gene products derived therefrom (e.g., messenger RNA (mRNA)) of HPV in a sample obtained from the subject. Determining the presence, absence or level of one or more genes or gene products derived therefrom (e.g., messenger RNA (mRNA)) of an HPV can indicate that said subject is experiencing ongoing HPV replication. Measuring or detecting the presence, absence or expression levels of one or more HPV genes can be done using sequencing (e.g., RNASeq), amplification (e.g., qRT-PCR) or hybridization assays (e.g., microarray analysis or in situ hybridization (ISH) assays). The sequencing assay can be any sequencing assay known in the art such as, for example, Cervista HPV 16/18 assay. The amplification assay can be any amplification assay known in the art such as, for example, the Hologic Aptima HPV assay or Roche Cobas HPV test. The hybridization assay can be any hybridization assay known in the art such as, for example, the Qiagen/Digene HC2 high-risk HPV test or Hologic Cervista HPV HR assay. In one embodiment, HPV status is determined using sequencing such as next-generation sequencing (NGS). For example, HPV status can be determined using NGS RNA sequencing (RNASeq) in order to detect read counts of one or more HPV genes (e.g., HPV E6 and/or E7). In one embodiment, read counts of greater than or equal to 200, 300, 400, 500, 600, 700, 800, 900, 1000, 1250, 1500, 1750 or 2000 are indicative of ongoing HPV replication. In one embodiment, the one or more genes or gene products derived therefrom (e.g., messenger RNA (mRNA)) can be the HPV E6 and/or E7 gene. The one or more genes can be the entire HPV genome or subsets thereof. In one embodiment, HPV status is determined by measuring or detecting expression of the HPV E6 gene and/or E7 gene in combination with one or more additional HPV genes. The HPV can be any type of HPV. In one embodiment, the HPV is selected from HPV types 16, 18, 31, 33, 35, 39, 45, 51, 52, 56, 58, 59, and 68 or any combination thereof. The detection of HPV E6 and E7 mRNA (or any other gene from an HPV genome) can be performed by ISH as described in Ukpo O C et al, High-risk human papillomavirus E6/E7 mRNA detection by a novel in situ hybridization assay strongly correlates with p16 expression and patient outcomes in oropharyngeal squamous cell carcinoma. Am J Surg Pathol. 2011; 35(9):1343-1350, Bishop J A. Ma X-J. Wang H. et al. Detection of transcriptionally active high-risk HPV in patients with head and neck squamous cell carcinoma as visualized by a novel E6/E7 mRNA in situ hybridization method. Am J Surg Pathol. 2012; 36(12):1874-1882, and Kerr D A. Arora K S. Mahadevan K K. et al. Performance of a branch chain RNA in situ hybridization assay for the detection of high-risk human papillomavirus in head and neck squamous cell carcinoma. Am J Surg Pathol. 2016; 39(12):1643-1652, each of which is herein incorporated by reference. In some cases, genes from HPV genomes can be detected using an ISH method as described in Ang K K. Harris J. Wheeler R. et al. Human papillomavirus and survival of patients with oropharyngeal cancer. N Engl J Med. 2010; 363(1):24-35, which is herein incorporated by reference. In some cases, genes from HPV genomes can be detected using the multiplex PCR and ISH methods as described in Fakhry C. Westra W H. Li S. et al. Improved survival of patients with human papillomavirus-positive head and neck squamous cell carcinoma in a prospective clinical trial. J Natl Cancer Inst. 2008; 100(4):261-269, which is herein incorporated by reference. In some cases, genes from HPV genomes can be detected using the Qiagen HC2 capture assay as described in Elke A. Jarboe, Mark Willis, Brandon Bentz, Luke Buchmann, Jason Hunt, Gary Ellis, Lester Layfield; Detection of Human Papillomavirus Using Hybrid Capture 2 in Oral Brushings From Patients With Oropharyngeal Squamous Cell Carcinoma, American Journal of Clinical Pathology, Volume 135, Issue 5, 1 May 2011, Pages 766-769, which is incorporated by herein by reference. In some cases, the presence of HPV genomes can be detected using RNA-seq and/or whole-exome sequencing methods as described in Parfenov M, Pedamallu C S, Gehlenborg N, et al. Characterization of HPV and host genome interactions in primary head and neck cancers. Proceedings of the National Academy of Sciences of the United States of America. 2014; 111(43):15544-15549, which is herein incorporated by reference. In some cases, the detection of HPV genes (e.g., HPV E6 and E7 mRNA) can be performed using qRT-PCR and/or RNA-seq as described in Kalu N N, Mazumdar T, Peng S, et al. Genomic characterization of human papillomavirus-positive and -negative human squamous cell cancer cell lines. Oncotarget. 2017; 8(49):86369-86383, which is herein incorporated by reference. In some cases, the genes from the HPV genome can be detected in a sample (e.g., head and neck sample) from a subject using the Roche Cobas HPV test as described in Angelique W. Levi, Jane I. Bernstein, Pei Hui, Kara Duch, Kevin Schofield, and David C. Chhieng (2016) A Comparison of the Roche Cobas HPV Test With the Hybrid Capture 2 Test for the Detection of High-Risk Human Papillomavirus Genotypes. Archives of Pathology & Laboratory Medicine: February 2016, Vol. 140, No. 2, pp. 153-157, which is herein incorporated by reference. In some cases, the genes from the HPV genome can be detected in a sample (e.g., head and neck sample) from a subject using the Hologic Aptima HPV assay as described in Max C, Michael G, Dan J, Lilian D M, Bernard J, et al. (2017) Performance of Aptima E6/E7 mRNA HPV assays on fine needle aspirates from cervical lymph nodes of patients with metastatic oropharyngeal squamous cell carcinoma. Otorhinolaryngol Head Neck Surg 2: DOI: 10.15761/OHNS.1000153, or the Hologic Cervista HPV assay as described in Guo, M., Khanna, A., Dhillon, J., Patel, S. J., Feng, J., Williams, M. D., Bell, D. M., Gong, Y., Katz, R. L., Sturgis, E. M. and Staerkel, G. A. (2014), Cervista HPV assays for fine-needle aspiration specimens are a valid option for human papillomavirus testing in patients with oropharyngeal carcinoma. Cancer Cytopathology, 122: 96-103, which is herein incorporated by reference.


In one embodiment, determining a subject's HPV status (e.g., by determining or detecting the presence, absence or level of one or more genes of HPV) is performed in addition to determining an HNSCC subtype of the subject. Further to this embodiment, the subtype is determined by detecting the expression levels of one or more classifier biomarkers using sequencing (e.g., RNASeq), amplification (e.g., qRT-PCR) or hybridization assays (e.g., microarray analysis) as described herein. The one or more biomarkers can be selected from a publically available database (e.g., TCGA HNSCC RNASeq gene expression datasets or any other publically available HNSCC gene expression datasets provided herein). In some embodiments, the biomarkers of Table 1 or Table 3 can be used to specifically determine the subtype of a HNSCC sample obtained from a patient as described herein. Further to these embodiments, determination of the HNSCC subtype can be performed prior to, concurrently with, or following determination of the HPV status of the subject. The HPV status can be determined using the methods provided herein. The HPV can be any type of HPV known in the art (e.g., the 202 types of HPV recognized by the International Human Papillomavirus Reference Center). In one embodiment, the HPV type is 16 18, 33, 35 or any combination thereof. As provided herein, the HPV status can be determined by measuring the gene expression (e.g., gene expression signatures) of HPV markers. In some cases, HPV status can be determined by determining the levels of surrogate HPV markers such as p16 and/or by examining the hematoxylin eosin morphology of tissue samples. In some cases, determining the levels of p16 and/or hematoxylin-eosin morphology can be performed in combination with measuring the gene expression of HPV markers as provided herein. The levels of p16 can be determined using any method known in the art and/or provided herein. The HPV markers can consist of, consist essentially of or comprise the whole HPV genome, or subsets thereof. In one embodiment, the HPV markers consist of, consist essentially of or comprise the HPV E6 and/or E7 gene. In another embodiment, the HPV markers consist of, consist essentially of or comprise the HPV E6 and/or E7 gene in combination with one or more HPV genes. The HPV markers can be measured in the same and/or different sample used to subtype the HNSCC sample as described herein. In one embodiment, the HPV status is determined by detecting read counts of one or more HPV genes (e.g., HPV E6 and/or E7 gene) from an RNAseq analysis of RNA isolated from a sample obtained from a subject such that reads counts above a predetermined threshold are indicative of ongoing, active HPV replication. The predetermined threshold can be 1000 read counts.


In another embodiment, determining a subject's HPV status (e.g., by determining or detecting the presence, absence or level of one or more genes of HPV) is used to determine an HNSCC subtype of the subject. Further to this embodiment, the subtype is determined by detecting the expression levels of one or more classifier biomarkers using sequencing (e.g., RNASeq), amplification (e.g., qRT-PCR) or hybridization assays (e.g., microarray analysis) as described herein in combination with determining the HPV status. The one or more biomarkers can be selected from a publically available database (e.g., TCGA HNSCC RNASeq gene expression datasets or any other publically available HNSCC gene expression datasets provided herein). In some embodiments, the biomarkers of Table 1 or Table 3 can be used in combination with determining the subject's HPV status in order to determine the subtype of a HNSCC sample obtained from the subject as described herein. The HPV status can be determined using the methods provided herein. The HPV can be any type of HPV known in the art (e.g., the 202 types of HPV recognized by the International Human Papillomavirus Reference Center). In one embodiment, the HPV type is 16, 18, 33, 35 or any combination thereof. As provided herein, the HPV status can be determined by measuring the gene expression (e.g., gene expression signatures) of HPV markers. In some cases, HPV status can be determined by determining the levels of surrogate HPV markers such as p16 and/or by examining the hematoxylin eosin morphology of tissue samples. In some cases, determining the levels of p16 and/or hematoxylin-eosin morphology can be performed in combination with measuring the gene expression of HPV markers as provided herein. The levels of p16 can be determined using any method known in the art and/or provided herein. The HPV markers can consist of, consist essentially of or comprise the whole HPV genome, or subsets thereof. In one embodiment, the HPV markers consist of, consist essentially of or comprise the HPV E6 and/or E7 gene. In another embodiment, the HPV markers consist of, consist essentially of or comprise the HPV E6 and/or E7 gene in combination with one or more HPV genes. The HPV markers can be measured in the same and/or different sample used to measure other classifier biomarkers (e.g., biomarkers from Tables 1 or 3) as described herein. In one embodiment, the HPV status is determined by detecting read counts of one or more HPV genes (e.g., HPV E6 and/or E7 gene) from an RNAseq analysis of RNA isolated from a sample obtained from a subject such that read counts above a predetermined threshold are indicative of ongoing, active HPV replication. The predetermined threshold can be 1000 read counts.


Clinical/Therapeutic Uses

In one embodiment, a method is provided herein for determining a disease outcome or prognosis for a patient suffering from cancer. In some cases, the cancer is head and neck squamous cell carcinoma. The disease outcome or prognosis can be measured by examining the overall survival for a period of time or intervals (e.g., 0 to 36 months or 0 to 60 months). In one embodiment, survival is analyzed as a function of subtype (e.g., for HNSCC (BA, MS, AT and CL)). The HNSCC subtype can be determined using the methods provided herein such as, for example, determining the expression of all or subsets of the genes in Tables 1 or 3 alone or in combination with determining the HPV status. Relapse-free and overall survival can be assessed using standard Kaplan-Meier plots as well as Cox proportional hazards modeling. For example, as shown in FIG. 17, a subject whose gene expression based HNSCC subtyping is indicative of a basal, atypical, or HPV positive (“Atypical-like”) HNSCC subtype can have an overall survival that is better than a subject whose gene expression based HNSCC subtyping is indicative of an HNSCC that is mesenchymal or classical. In one embodiment, assessing the HPV status of a subject as a means of assisting in subtyping HNSCC or in conjunction with subtyping HNSCC is more predictive of said subject's prognosis than assessing HPV status alone or determining HNSCC subtype via gene expression based HNSCC subtyping without assessing HPV status. For example, as shown in FIG. 2A, a subject whose HPV status is positive and whose gene expression based HNSCC subtyping is indicative of an atypical HNSCC subtype can have an overall survival that is better than a subject whose HPV status is positive and whose gene expression based HNSCC subtyping is indicative of an HNSCC that is not atypical. The HPV status can be performed using any of the methods provided herein such as, for example, detecting the expression of one or more HPV genes (e.g., HPV E6 and/or E7 genes). The gene expression based HNSCC subtyping can be performed using any of the methods provided herein such as, for example, detecting the expression of one or more of the biomarkers listed in Tables 1 or 3.


In another embodiment, assessing the HPV status of a subject as a means of assisting in gene expression based HNSCC subtyping or in conjunction with gene expression based HNSCC subtyping is more predictive of said subject's response to a particular type of therapy (e.g., immunotherapy, radiotherapy, surgical intervention) than assessing HPV status alone or determining HNSCC subtype without assessing HPV status. The HPV status can be performed using any of the methods provided herein such as, for example, detecting the expression of one or more HPV genes (e.g., HPV E6 and/or E7 genes). The gene expression based HNSCC subtyping can be performed using any of the methods provided herein such as, for example, detecting the expression of one or more of the biomarkers listed in Tables 1 or 3.


In one embodiment, upon determining a patient's HNSCC subtype (e.g., by measuring the expression of all or subsets of the genes in Tables 1 or 3 alone or in combination with determining the HPV status), the patient is selected for suitable therapy, for example, radiotherapy (radiation therapy), surgical intervention, target therapy, chemotherapy or drug therapy with an angiogenesis inhibitor or immunotherapy or combinations thereof. In some embodiments, the suitable treatment can be any treatment or therapeutic method that can be used for a HNSCC patient. In one embodiment, upon determining a patient's HNSCC subtype, the patient is administered a suitable therapeutic agent, for example chemotherapeutic agent(s) or an angiogenesis inhibitor or immunotherapeutic agent(s). In one embodiment, the therapy is immunotherapy, and the immunotherapeutic agent is a checkpoint inhibitor, monoclonal antibody, biological response modifier, therapeutic vaccine or cellular immunotherapy. In some embodiments, the determination of a suitable treatment can identify treatment responders. In some embodiments, the determination of a suitable treatment can identify treatment non-responders. In some embodiments, upon determining a patient's HNSCC subtype, the HNSCC patients can be selected for any combination of suitable therapies. For example, chemotherapy or drug therapy with a radiotherapy, a neck dissection with an immunotherapy or a chemotherapeutic agent with a radiotherapy. In some embodiments, immunotherapy, or immunotherapeutic agent can be a checkpoint inhibitor, monoclonal antibody, biological response modifier, therapeutic vaccine or cellular immunotherapy.


The methods of present invention are also useful for evaluating clinical response to therapy, as well as for endpoints in clinical trials for efficacy of new therapies. The extent to which sequential diagnostic expression profiles move towards normal can be used as one measure of the efficacy of the candidate therapy.


In one embodiment, the methods of the invention also find use in predicting response to different lines of therapies based on the subtype of HNSCC. For example, chemotherapeutic response can be improved by more accurately assigning tumor subtypes. Likewise, treatment regimens can be formulated based on the tumor subtype.


Angiogenesis Inhibitors


In one embodiment, upon determining a patient's HNSCC subtype, the patient is selected for drug therapy with an angiogenesis inhibitor.


In one embodiment, the angiogenesis inhibitor is a vascular endothelial growth factor (VEGF) inhibitor, a VEGF receptor inhibitor, a platelet derived growth factor (PDGF) inhibitor or a PDGF receptor inhibitor.


Each biomarker panel can include one, two, three, four, five, six, seven, eight or more biomarkers usable by a classifier (also referred to as a “classifier biomarker”) to assess whether a HNSCC patient is likely to respond to angiogenesis inhibitor therapy; to select a HNSCC patient for angiogenesis inhibitor therapy; to determine a “hypoxia score” and/or to subtype a HNSCC sample as basal, mesenchymal, atypical, or classical molecular subtype. As used herein, the term “classifier” can refer to any algorithm for statistical classification, and can be implemented in hardware, in software, or a combination thereof. The classifier can be capable of 2-level, 3-level, 4-level, or higher, classification, and can depend on the nature of the entity being classified. One or more classifiers can be employed to achieve the aspects disclosed herein.


In general, methods of determining whether a HNSCC patient is likely to respond to angiogenesis inhibitor therapy, or methods of selecting a HNSCC patient for angiogenesis inhibitor therapy are provided herein. In one embodiment, the method comprises assessing whether the patient's HNSCC subtype is basal, mesenchymal, atypical, or classical using the methods described herein (e.g., assessing the expression of one or more classifier biomarkers of Table 1 or Table 3 alone or in combination with assessing the expression of one or more HPV genes) and probing a HNSCC sample from the patient for the levels of at least five biomarkers selected from the group consisting of RRAGD, FABP5, UCHL1, GAL, PLOD, DDIT4, VEGF, ADM, ANGPTL4, NDRG1, NP, SLC16A3, and C140RF58 (see Table 5) at the nucleic acid level. In a further embodiment, the probing step comprises mixing the sample with five or more oligonucleotides that are substantially complementary to portions of nucleic acid molecules of the at least five biomarkers under conditions suitable for hybridization of the five or more oligonucleotides to their complements or substantial complements, detecting whether hybridization occurs between the five or more oligonucleotides to their complements or substantial complements; and obtaining hybridization values of the sample based on the detecting steps. The hybridization values of the sample are then compared to reference hybridization value(s) from at least one sample training set, wherein the at least one sample training set comprises (i) hybridization value(s) of the at least five biomarkers from a sample that overexpresses the at least five biomarkers, or overexpresses a subset of the at least five biomarkers, (ii) hybridization values of the at least five biomarkers from a reference basal, mesenchymal, atypical, or classical sample, or (iii) hybridization values of the at least five biomarkers from a HNSCC free head and neck sample. A determination of whether the patient is likely to respond to angiogenesis inhibitor therapy, or a selection of the patient for angiogenesis inhibitor is then made based upon (i) the patient's HNSCC subtype and (ii) the results of comparison.









TABLE 5







Biomarkers for hypoxia profile











GenBank


Name
Abbreviation
Accession No.





RRAGD
Ras-related GTP binding D
BC003088


FABP5
fatty acid binding protein 5
M94856


UCHL1
ubiquitin carboxyl-terminal esterase L1
NM_004181


GAL
Galanin
BC030241


PLOD
procollagen-lysine, 2-oxoglutarate 5-
M98252



dioxygenase lysine hydroxylase


DDIT4
DNA-damage-inducible transcript 4
NM_019058


VEGF
vascular endothelial growth factor
M32977


ADM
Adrenomedullin
NM_001124


ANGPTL4
angiopoietin-like 4
AF202636


NDRG1
N-myc downstream regulated gene 1
NM_006096


NP
nucleoside phosphorylase
NM 000270


SLC16A3
solute carrier family 16 monocarboxylic
NM_004207



acid transporters, member 3


C14ORF58
chromosome 14 open reading frame 58
AK000378









The aforementioned set of thirteen biomarkers, or a subset thereof, is also referred to herein as a “hypoxia profile”.


In one embodiment, the method provided herein includes determining the levels of at least five biomarkers, at least six biomarkers, at least seven biomarkers, at least eight biomarkers, at least nine biomarkers, or at least ten biomarkers, or five to thirteen, six to thirteen, seven to thirteen, eight to thirteen, nine to thirteen or ten to thirteen biomarkers selected from RRAGD, FABP5, UCHL1, GAL, PLOD, DDIT4, VEGF, ADM, ANGPTL4, NDRG1, NP, SLC16A3, and C140RF58 in a HNSCC sample obtained from a subject. Biomarker expression in some instances may be normalized against the expression levels of all RNA transcripts or their expression products in the sample, or against a reference set of RNA transcripts or their expression products. The reference set as explained throughout, may be an actual sample that is tested in parallel with the HNSCC sample, or may be a reference set of values from a database or stored dataset. Levels of expression, in one embodiment, are reported in number of copies, relative fluorescence value or detected fluorescence value. The level of expression of the biomarkers of the hypoxia profile together with HNSCC subtype as determined using the methods provided herein can be used in the methods described herein to determine whether a patient is likely to respond to angiogenesis inhibitor therapy.


In one embodiment, the levels of expression of the thirteen biomarkers (or subsets thereof, as described above, e.g., five or more, from about five to about 13), are normalized against the expression levels of all RNA transcripts or their non-natural cDNA expression products, or protein products in the sample, or of a reference set of RNA transcripts or a reference set of their non-natural cDNA expression products, or a reference set of their protein products in the sample.


In one embodiment, angiogenesis inhibitor treatments include, but are not limited to an integrin antagonist, a selectin antagonist, an adhesion molecule antagonist, an antagonist of intercellular adhesion molecule (ICAM)-1, ICAM-2, ICAM-3, platelet endothelial adhesion molecule (PCAM), vascular cell adhesion molecule (VCAM)), lymphocyte function-associated antigen 1 (LFA-1), a basic fibroblast growth factor antagonist, a vascular endothelial growth factor (VEGF) modulator, a platelet derived growth factor (PDGF) modulator (e.g., a PDGF antagonist).


In one embodiment of determining whether a subject is likely to respond to an integrin antagonist, the integrin antagonist is a small molecule integrin antagonist, for example, an antagonist described by Paolillo et al. (Mini Rev Med Chem, 2009, volume 12, pp. 1439-1446, incorporated by reference in its entirety), or a leukocyte adhesion-inducing cytokine or growth factor antagonist (e.g., tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), monocyte chemotactic protein-1 (MCP-1) and a vascular endothelial growth factor (VEGF)), as described in U.S. Pat. No. 6,524,581, incorporated by reference in its entirety herein.


The methods provided herein are also useful for determining whether a subject is likely to respond to one or more of the following angiogenesis inhibitors: interferon gamma 1β, interferon gamma 1β (Actimmune®) with pirfenidone, ACUHTR028, aV135, aminobenzoate potassium, amyloid P, ANG1122, ANG1170, ANG3062, ANG3281, ANG3298, ANG4011, anti-CTGF RNAi, Aplidin, Astragalus membranaceus extract with salvia and Schisandra chinensis, atherosclerotic plaque blocker, Azol, AZX100, BB3, connective tissue growth factor antibody, CT140, danazol, Esbriet, EXC001, EXC002, EXC003, EXC004, EXC005, F647, FG3019, Fibrocorin, Follistatin, FT011, a galectin-3 inhibitor, GKT137831, GMCT01, GMCT02, GRMD01, GRMD02, GRN510, Heberon Alfa R, interferon α-2β, ITMN520, JKB119, JKB121, JKB122, KRX168, LPA1 receptor antagonist, MGN4220, MIA2, microRNA 29a oligonucleotide, MMI0100, noscapine, PBI4050, PBI4419, PDGFR inhibitor, PF-06473871, PGN0052, Pirespa, Pirfenex, pirfenidone, plitidepsin, PRM151, Px102, PYN17, PYN22 with PYN17, Relivergen, rhPTX2 fusion protein, RXI109, secretin, STX100, TGF-β Inhibitor, transforming growth factor, β-receptor 2 oligonucleotide, VA999260, XV615 or a combination thereof.


In another embodiment, a method is provided for determining whether a subject is likely to respond to one or more endogenous angiogenesis inhibitors. In a further embodiment, the endogenous angiogenesis inhibitor is endostatin, a 20 kDa C-terminal fragment derived from type XVIII collagen, angiostatin (a 38 kDa fragment of plasmin), a member of the thrombospondin (TSP) family of proteins. In a further embodiment, the angiogenesis inhibitor is a TSP-1, TSP-2, TSP-3, TSP-4 and TSP-5. Methods for determining the likelihood of response to one or more of the following angiogenesis inhibitors are also provided a soluble VEGF receptor, e.g., soluble VEGFR-1 and neuropilin 1 (NPR1), angiopoietin-1, angiopoietin-2, vasostatin, calreticulin, platelet factor-4, a tissue inhibitor of metalloproteinase (TIMP) (e.g., TIMP1, TIMP2, TIMP3, TIMP4), cartilage-derived angiogenesis inhibitor (e.g., peptide troponin I and chrondomodulin I), a disintegrin and metalloproteinase with thrombospondin motif 1, an interferon (IFN), (e.g., IFN-α, IFN-β, IFN-γ), a chemokine, e.g., a chemokine having the C-X-C motif (e.g., CXCL10, also known as interferon gamma-induced protein 10 or small inducible cytokine B10), an interleukin cytokine (e.g., IL-4, IL-12, IL-18), prothrombin, antithrombin III fragment, prolactin, the protein encoded by the TNFSF15 gene, osteopontin, maspin, canstatin, proliferin-related protein.


In one embodiment, a method for determining the likelihood of response to one or more of the following angiogenesis inhibitors is provided is angiopoietin-1, angiopoietin-2, angiostatin, endostatin, vasostatin, thrombospondin, calreticulin, platelet factor-4, TIMP, CDAI, interferon α, interferon β, vascular endothelial growth factor inhibitor (VEGI) meth-1, meth-2, prolactin, VEGI, SPARC, osteopontin, maspin, canstatin, proliferin-related protein (PRP), restin, TSP-1, TSP-2, interferon gamma 1β, ACUHTR028, αVβ5, aminobenzoate potassium, amyloid P, ANG1122, ANG1170, ANG3062, ANG3281, ANG3298, ANG4011, anti-CTGF RNAi, Aplidin, Astragalus membranaceus extract with salvia and Schisandra chinensis, atherosclerotic plaque blocker, Azol, AZX100, BB3, connective tissue growth factor antibody, CT140, danazol, Esbriet, EXC001, EXC002, EXC003, EXC004, EXC005, F647, FG3019, Fibrocorin, Follistatin, FT011, a galectin-3 inhibitor, GKT137831, GMCT01, GMCT02, GRMD01, GRMD02, GRN510, Heberon Alfa R, interferon α-2β, ITMN520, JKB119, JKB121, JKB122, KRX168, LPA1 receptor antagonist, MGN4220, MIA2, microRNA 29a oligonucleotide, MMI0100, noscapine, PBI4050, PBI4419, PDGFR inhibitor, PF-06473871, PGN0052, Pirespa, Pirfenex, pirfenidone, plitidepsin, PRM151, Px102, PYN17, PYN22 with PYN17, Relivergen, rhPTX2 fusion protein, RXI109, secretin, STX100, TGF-β Inhibitor, transforming growth factor, β-receptor 2 oligonucleotide, VA999260, XV615 or a combination thereof.


In yet another embodiment, the angiogenesis inhibitor can include pazopanib (Votrient), sunitinib (Sutent), sorafenib (Nexavar), axitinib (Inlyta), ponatinib (Iclusig), vandetanib (Caprelsa), cabozantinib (Cometrig), ramucirumab (Cyramza), regorafenib (Stivarga), ziv-aflibercept (Zaltrap), motesanib, or a combination thereof. In another embodiment, the angiogenesis inhibitor is a VEGF inhibitor. In a further embodiment, the VEGF inhibitor is axitinib, cabozantinib, aflibercept, brivanib, tivozanib, ramucirumab or motesanib. In yet a further embodiment, the angiogenesis inhibitor is motesanib.


In one embodiment, the methods provided herein relate to determining a subject's likelihood of response to an antagonist of a member of the platelet derived growth factor (PDGF) family, for example, a drug that inhibits, reduces or modulates the signaling and/or activity of PDGF-receptors (PDGFR). For example, the PDGF antagonist, in one embodiment, is an anti-PDGF aptamer, an anti-PDGF antibody or fragment thereof, an anti-PDGFR antibody or fragment thereof, or a small molecule antagonist. In one embodiment, the PDGF antagonist is an antagonist of the PDGFR-α or PDGFR-β. In one embodiment, the PDGF antagonist is the anti-PDGF-β aptamer E10030, sunitinib, axitinib, sorefenib, imatinib, imatinib mesylate, nintedanib, pazopanib HCl, ponatinib, MK-2461, dovitinib, pazopanib, crenolanib, PP-121, telatinib, imatinib, KRN 633, CP 673451, TSU-68, Ki8751, amuvatinib, tivozanib, masitinib, motesanib diphosphate, dovitinib dilactic acid, linifanib (ABT-869).


Upon making a determination of whether a patient is likely to respond to angiogenesis inhibitor therapy, or selecting a patient for angiogenesis inhibitor therapy, in one embodiment, the patient is administered the angiogenesis inhibitor. The angiogenesis in inhibitor can be any of the angiogenesis inhibitors described herein.


Immunotherapy


In one embodiment, provided herein is a method for determining whether a HNSCC cancer patient is likely to respond to immunotherapy by determining the subtype of HNSCC of a sample obtained from the patient and, based on the HNSCC subtype, assessing whether the patient is likely to respond to immunotherapy. In another embodiment, provided herein is a method of selecting a patient suffering from HNSCC for immunotherapy by determining a HNSCC subtype of a sample from the patient and, based on the HNSCC subtype, selecting the patient for immunotherapy. The determination of the HNSCC subtype of the sample obtained from the patient can be performed using any method for subtyping HNSCC known in the art. The determination of the HNSCC subtype of the sample obtained from the patient can be performed using any method for subtyping HNSCC provided herein. In one embodiment, the sample obtained from the patient has been previously diagnosed as being HNSCC, and the methods provided herein are used to determine the HNSCC subtype of the sample. The previous diagnosis can be based on a histological analysis. The histological analysis can be performed by one or more pathologists. In one embodiment, the HNSCC subtyping is performed via gene expression analysis of a set or panel of biomarkers or subsets thereof in order to generate an expression profile. The gene expression analysis can be performed on a head and neck cancer sample (e.g., HNSCC sample) obtained from a patient in order to determine the presence, absence or level of expression of one or more biomarkers selected from a publically available head and neck cancer database described herein and/or Table 1 or 3 provided herein. The gene expression analysis can further comprise determining the HPV status of the sample obtained from the subject. The HPV status can be assessed as provided herein (e.g., detecting the expression of one or more HPV genes). The HNSCC subtype can be selected from the group consisting of basal, atypical, mesenchymal or classical. The immunotherapy can be any immunotherapy provided herein. In one embodiment, the immunotherapy comprises administering one or more checkpoint inhibitors. The checkpoint inhibitors can be any checkpoint inhibitor provided herein such as, for example, a checkpoint inhibitor that targets PD-1, PD-LI or CTLA4.


As disclosed herein, the biomarkers panels, or subsets thereof, can be those disclosed in any publically available HNSCC gene expression dataset or datasets alone or in combination with one or more biomarkers of HPV. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the cancer genome atlas (TCGA) HNSCC RNAseq gene expression dataset (n=520). In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset (n=134) disclosed in Keck et al. (Clin Cancer Res. 2014; 21: 870-881.), the contents of which are herein incorporated by reference in its entirety. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset (n=138) disclosed in Von Walter et al. (PLoS One, 8(2):e56823), the contents of which are herein incorporated by reference in its entirety. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset (n=270) disclosed in Wichman et al. (Intl Jrnl Cancer 2015; 137: 2846-2857), the contents of which are herein incorporated by reference in its entirety. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset disclosed in Table 1 or Table 3. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset disclosed in Table 1 or Table 3 in combination with one or more biomarkers from a publically available HNSCC expression dataset. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset disclosed in Table 1 or Table 3 in combination with one or more biomarkers of HPV. In one embodiment, the head and neck cancer is SCC and the biomarker panel or subset thereof is, for example, the HNSCC gene expression dataset disclosed in Table 1 or Table 3 in combination with one or more biomarkers from a publically available HNSCC expression dataset and one or more biomarkers of HPV. In Table 2 or Table 4, the first column of the table represents the biomarker list for distinguishing atypical. The second column of the table represents the biomarker list for mesenchymal. The third column of the table represents the biomarker list for distinguishing classical. The last column of the table represents the biomarker list for distinguishing basal. In some cases, as shown in Table 2, a total of 144 biomarkers can be used for HNSCC subtype determination. For each HNSCC subtype in Table 2, 18 of the 36 biomarkers can be negatively correlated genes, while 18 can be positively correlated genes which can be selected as the gene signature of a specific HNSCC subtype. In some cases, as shown in Table 4, a total of 80 biomarkers can be used for HNSCC subtype determination. For each HNSCC subtype in Table 4, 10 of the 20 biomarkers can be negatively correlated genes, while 10 can be positively correlated genes which can be selected as the gene signature of a specific HNSCC subtype.


In some embodiments, the method for HNSCC subtyping includes detecting expression levels of a classifier biomarker set alone or in combination with one or more biomarkers of HPV. The classifier biomarker set can be a set of biomarkers from a publically available database such as, for example, TCGA HNSCC RNASeq gene expression dataset(s) or any other dataset provided herein. In some embodiments, the detecting includes all of the classifier biomarkers of Table 1 or Table 3 or any other dataset provided herein at the nucleic acid level or protein level. In another embodiment, a single classifier biomarker of Table 1 or Table 3 or a subset of the classifier biomarkers of Table 1 or Table 3 or any other dataset provided herein are detected, for example, from about five to about twenty. In another embodiment, a single classifier biomarker of Table 1 or a subset of the classifier biomarkers of Table 1 and/or any other dataset provided herein are detected, for example, from about 18 to about 144. In another embodiment, a single classifier biomarker of Table 3 or a subset of the classifier biomarkers of Table 3 and/or any other dataset provided herein are detected, for example, from about 10 to about 80. In another embodiment, all of the classifier biomarkers of Table 1 or Table 3 or any other dataset provided herein are detected. In another embodiment, at least one or all of the classifier biomarkers of Table 1 or Table 3 in combination with one or more classifier biomarkers of any other HNSCC dataset provided herein are detected. The detecting can be performed by any suitable technique including, but not limited to, RNA-seq, a reverse transcriptase polymerase chain reaction (RT-PCR), a microarray hybridization assay, or another hybridization assay, e.g., a NanoString assay for example, with primers and/or probes specific to the classifier biomarkers, and/or the like. In some cases, the primers useful for the amplification methods (e.g., RT-PCR or qRT-PCR) are any forward and reverse primers suitable for binding to a classifier gene from a dataset provided herein alone or in combination.


In one embodiment, from about 1 to about 5, about 5 to about 10, from about 5 to about 15, from about 5 to about 20, from about 5 to about 25, from about 5 to about 30, from about 5 to about 35, from about 5 to about 40, from about 5 to about 45, from about 5 to about 50, from about 5 to about 55, from about 5 to about 60, from about 5 to about 65, from about 5 to about 70, from about 5 to about 75, or from about 5 to about 80 of the biomarkers in any of the HNSCC gene expression datasets provided herein, including, for example, Table 1 or Table 3 for an HNSCC sample are detected in a method to determine the HNSCC subtype as provided herein. In another embodiment, each of the biomarkers from any one of the HNSCC gene expression datasets provided herein, including, for example, Table 1 or Table 3 for an HNSCC sample are detected in a method to determine the HNSCC subtype as provided herein. Further to the above embodiments, the HPV status can be determined by measuring one or more biomarkers of HPV as described herein.


In one embodiment, the methods provided herein further comprise determining the presence, absence or level of immune activation in a HNSCC subtype. The presence or level of immune cell activation can be determined by creating an expression profile or detecting the expression of one or more biomarkers associated with innate immune cells and/or adaptive immune cells associated with each HNSCC subtype in a sample obtained from a patient. In one embodiment, immune cell activation associated with a HNSCC subtype is determined by monitoring the immune cell signatures of Bindea et al (Immunity 2013; 39(4); 782-795), the contents of which are herein incorporated by reference in its entirety. In one embodiment, the method further comprises measuring single gene immune biomarkers, such as, for example, CTLA4, PDCD1 and CD274 (PD-LI), PDCDLG2(PD-L2) and/or IFN gene signatures. The presence or a detectable level of immune activation (Innate and/or Adaptive) associated with a HNSCC subtype can indicate or predict that a patient with said HNSCC subtype may be amendable to immunotherapy. The immunotherapy can be treatment with a checkpoint inhibitor as provided herein. In one embodiment, a method is provided herein for detecting the expression of at least one classifier biomarker provided herein in a sample (e.g., HNSCC sample) obtained from a patient further comprises administering an immunotherapeutic agent following detection of immune activation as provided herein in said sample.


In one embodiment, the method comprises determining a subtype of a HNSCC sample and subsequently determining a level of immune cell activation of said sub-type. In one embodiment, the subtype is determined by determining the expression levels of one or more classifier biomarkers using sequencing (e.g., RNASeq), amplification (e.g., qRT-PCR) or hybridization assays (e.g., microarray analysis) as described herein. The one or more biomarkers can be selected from a publically available database (e.g., TCGA HNSCC RNASeq gene expression datasets or any other publically available HNSCC gene expression datasets provided herein). In some embodiments, the biomarkers of Table 1 or Table 3 can be used to specifically determine the subtype of a HNSCC sample obtained from a patient. In some embodiments, the subtyping can further comprises determining the HPV status by measuring one or more biomarkers of HPV as described herein. In some embodiments, the subtyping can be in combination with also determining the HPV status by measuring one or more biomarkers of HPV as described herein. In one embodiment, the level of immune cell activation is determined by measuring gene expression signatures of immunomarkers. The immunomarkers can be measured in the same and/or different sample used to subtype the HNSCC sample as described herein. The immunomarkers that can be measured can comprise, consist of, or consistently essentially of innate immune cell (IIC) and/or adaptive immune cell (AIC) gene signatures, interferon (IFN) gene signatures, individual immunomarkers, major histocompatability complex class II (MHC class II) genes or a combination thereof. The gene expression signatures for both IICs and AICs can be any known gene signatures for said cell types known in the art. For example, the immune gene signatures can be those from Bindea et al. (Immunity 2013; 39(4); 782-795). In one embodiment, the immunomarkers for use in the methods provided herein are selected from Table 6A and/or Table 6B. The individual immunomarkers can be CTLA4, PDCD1 and CD274 (PD-L1). In one embodiment, the individual immunomarkers for use in the methods provided herein are selected from Table 7. The immunomarkers can be one or more interferon (INF) genes. In one embodiment, the immunomarkers for use in the methods provided herein are selected from Table 8. The immunomarkers can be one or more MHCII genes. In one embodiment, the immunomarkers for use in the methods provided herein are selected from Table 9. In yet another embodiment, the immunomarkers for use in the methods provided herein are selected from Tables 6A, 6B, 7, 8, 9, or a combination thereof.









TABLE 6A





Adaptive immune cell (AIC) gene signature immunomarkers for use in the methods provided herein.


Cell Type






















B cells
T cells
T helper cells
Tcm
Tem
Th1 cells





Human
ABCB4 (ATP
BCL11B
ANP32B
AQP3 (aquaporine 3;
AKT3 (AKT
APBB2 (amyloid


Gene
binding
(B-cell
(acidic nuclear
NM_004925.4)
serine/threonine
beta precursor


(Gene
cassette
lymphoma/
phosphoprotein

kinase 3;
protein binding


Name;
subfamily B
leukaemia 11B;
32 family member B;

NM_005465.4)
family B member 2;


GenBank
member 4;
AJ404614.1)
NM_006401.2)


NM_001166054.1)


Accession
NM_000443)


No.*)
BACH2 (BTB
CD2
ASF1A
ATF7IP (activating
C7orf54
APOD



domain and
(CD2 molecule;
(anti-silencing
transcription factor 7
(staphylococcal
(apolipoprotein D;



CNC homolog 2;
NM_001328609.1)
function
interacting protein;
nuclease and tudor
NM_001647.3)



NM_021813.3)

1A histone
NM_181352.1)
domain containing 1





chaperone;

(SND1);





NM_014034.2)

NG_051199.1)



BCL11A
CD28 (CD28
ATF2
ATM (ATM
CCR2 (C-C motif
ATP9A (ATPase



(B-cell CLL/
molecule;
(activating
serine/threonine
chemokine receptor
phospholipid



lymphoma 11A;
NM_001243078.1)
transcription
kinase;
2;
transporting 9A;



NM_022893.3)

factor 2;
NM_000051.3)
NM_001123396.1)
NM_006045.2)





NM_001256093.1)



BLK (BLK proto-
CD3D (CD3d
BATF
CASP8 (caspase 8;
DDX17 (DEAD-box
BST2 (bone marrow



oncogene,
molecule;
(basic leucine
NM_001228.4)
helicase 17;
stromal cell antigen



Src family
NM_000732.4)
zipper ATF-like

NM_006386.4)
2; NM_004335.3)



tyrosine kinase;

transcription



NM_001715.2)

factor;





NM_006399.3)



BLNK
CD3E (CD3e
C13orf34
CDC14A (cell division
EWSR1 (EWS RNA
BTG3 (BTG anti-



(B-cell linker;
molecule;
(aurora
cycle 14A;
binding protein 1;
proliferation factor



NM_013314.3)
NM_000733.3)
borealis;
NM_003672.3)
NM_013986.3)
3;





EU834129.1)


NM_001130914.1)



CCR9
CD3G (CD3g
CD28 (CD28
CEP68 (centrosomal
FLI1 (Fli-1 proto-
CCL4 (C-C motif



(C-C motif
molecule;
molecule;
protein 68;
oncogene, ETS
chemokine ligand 4;



chemokine
NM_000073.2)
NM_006139.3)
NM_015147.2)
transcription factor;
NM_002984.3)



receptor 9;



NM_002017.4)



NM_031200.2)



CD19 (CD19
CD6 (CD6
DDX50 (DEAD-box
CG030 (BRCA2
GDPD5
CD38 (CD38



molecule;
molecule;
helicase 50;
region, mRNA
(glycerophosphodiester
molecule;



NM_001178098.1)
NM_006725.4)
NM_024045.1)
sequence CG030;
phosphodiesterase
NM_001775.3)






U50531.1)
domain containing







5; NM_030792.6)



CD72 (CD72
CD96 (CD96
FAM111A (family
CLUAP1 (clusterin
LTK (leukocyte
CD70 (CD70



molecule;
molecule;
with sequence
associated
receptor tyrosine
molecule;



NM_001782.2)
NM_198196.2)
similarity 111
protein 1;
kinase;
NM_001252.4)





member A;
NM_015041.2)
NM_002344.5)





NM_022074.3)



COCH (cochlin;
GIMAP5
FRYL
CREBZF
MEFV
CMAH (cytidine



NM_001135058.1)
(GTPase,
(FRY like
(CREB/ATF
(Mediterranean
monophospho-N-




IMAP family
transcription
bZIP transcription
fever;
acetylneuraminic




member 5;
coactivator;
factor;
NM_000243.2)
acid hydroxylase,




NM_018384.4)
NM_015030.1)
NM_001039618.2)

pseudogene;








NR_002174.2)



CR2 (complement
ITM2A (integral
FUSIP1
CYLD
NFATC4 (nuclear
CSF2 (colony



C3d receptor 2;
membrane protein
(serine and
(CYLD lysine 63
factor of
stimulating



NM_001006658.2)
2A; NM_004867.4)
arginine
deubiquitinase;
activated
factor 2;





rich splicing
NM_015247.2)
T-cells 4;
NM_000758.3)





factor 10;

NM_001136022.2)





NM_006625.5)



DTNB
LCK
GOLGA8A
CYorf15B (taxilin
PRKY (protein
CTLA4



(dystrobrevin
(LCK proto-
(golgin A8
gamma pseudogene,
kinase, Y-linked,
(cytotoxic T-



beta;
oncogene,
family
Y-linked;
pseudogene;
lymphocyte



NM_021907.4)
Src family
member A;
NR_045128.1)
NR_028062.1)
associated protein




tyrosine kinase;
NM_181077.3)


4; NM_005214.4)




NM_001042771.2)



FAM30A (family
NCALD
ICOS
DOCK9 (dedicator of
TBC1D5 (TBC1
DGKI



with sequence
(neurocalcin
(inducible
cytokinesis 9;
domain family
(diacylglycerol



similarity 30,
delta;
T-cell
NM_015296.2)
member 5;
kinase iota;



member A;
NM_001040624.1)
costimulator;

NM_001134381.1)
NM_004717.3)



NR_026800.2)

NM_012092.3)



FCRL2
PRKCQ (protein
ITM2A (integral
FOXP1 (forkhead box
TBCD (tubulin
DOK5 (docking



(Fc receptor
kinase C theta;
membrane protein
P1; NM_032682.5)
folding cofactor D;
protein 5;



like 2;
NM_006257.4)
2A; NM_004867.4)

NM_005993.4)
NM_018431.4)



NM_030764.3)



GLDC (glycine
SH2D1A
LRBA (LPS
FYB (FYN binding
TRA (T cell
DPP4 (dipeptidyl



decarboxylase;
(SH2 domain
responsive
protein;
receptor
peptidase 4;



NM_000170.2)
containing 1A;
beige-like
NM_001465.4)
alpha delta locus;
NM_001935.3)




NM_002351.4)
anchor protein;

NG_001332.3)





NM_001199282.2)



GNG7
SKAP1 (src
NAP1L4
HNRPH1
VIL2 (ezrin;
DUSP5 (dual



(G protein
kinase
(nucleosome
(heterogeneous
NM_003379.4)
specificity



subunit
associated
assembly
nuclear

phosphatase 5;



gamma 7;
phosphoprotein 1;
protein 1
ribonucleoprotein H1

NM_004419.3)



NM_052847.2)
NM_001075099.1)
like 4;
(H);





NM_005969.3)
NM_001257293.1)



HLA-DOB (major
TRA (T cell
NUP107
INPP4B (inositol

EGFL6 (EGF like



histocompatibility
receptor
(nucleoporin
polyphosphate-4-

domain multiple 6;



complex,
alpha delta
107;
phosphatase type II

NM_015507.3)



class II, DO
locus;
NM_020401.3)
B; NM_003866.3)



beta;
NG_001332.3)



NM_002120.3)



HLA-DQA1 (major
TRAC (nuclear
PHF10
KLF12

GGT1 (gamma-



histocompatibility
receptor
(PHD finger
(Kruppel like

glutamyltransferase



complex,
corepressor
protein 10;
factor 12;

1; NM_013421.2)



class II, DQ
2; NM_006312.5)
NM_018288.3)
NM_007249.4)



alpha 1;



NM_002122.3)



IGHA1
TRAT1 (T cell
PPP2R5C
LOC202134 (family

HBEGF (heparin



(immunoglobulin
receptor
(protein
with sequence

binding EGF like



heavy locus;
associated
phosphatase 2
similarity 153

growth factor;



NG_001019.6)
transmembrane
regulatory
member B;

NM_001945.2)




adaptor 1;
subunit B′,
NM_001265615.1)




NM_016388.3)
gamma;





NM_001161725.1)



IGHG1
TRBC1 (T cell
RPA1
MAP3K1 (mitogen-

IFNG (interferon



(immunoglobulin
receptor
(replication
activated protein

gamma;



heavy locus;
beta locus;
protein A1;
kinase kinase kinase

NM_000619.2)



NG_001019.6)
NG_001333.2)
NM_002945.3)
1, E3 ubiquitin






protein ligase;






NM_005921.1)



IGHM

SEC24C
MLL (lysine (K)-

IL12RB2



(immunoglobulin

(SEC24
specific

(interleukin



heavy locus;

homolog C,
methyltransferase

12 receptor



NG_001019.6)

COPII
2A; NM_005933.3)

subunit





coat complex


beta 2;





component;


NM_001319233.1)





NM_004922.3)



IGKC

SLC25A12
NEFL (neurofilament,

IL22



(immunoglobulin

(solute
light polypeptide;

(interleukin 22;



kappa locus,

carrier
NM_006158.4)

NM_020525.4)



proximal

family 25



V-cluster

member 12;



and J-C cluster;

NM_003705.4)



NG_000834.1)



IGL

TRA
NFATC3 (nuclear

LRP8



(immunoglobulin

(T cell
factor of activated T-

(LDL receptor



lambda locus;

receptor
cells 3;

related



NG_000002.1)

alpha delta
NM_173165.2)

protein 8;





locus;


NM_017522.4)





NG_001332.3)



KIAA0125 (family

UBE2L3
PCM1 (pericentriolar

LRRN3



with sequence

(ubiquitin
material 1;

(leucine rich



similarity 30,

conjugating
NM_001315507.1)

repeat



member A;

enzyme E2 L3;


neuronal 3;



NR_026800.2)

NM_003347.3)


NM_018334.4)



MEF2C (myocyte

YME1L1
PCNX (pecanex

LTA



enhancer

(YME1 like 1
homolog 1;

(lymphotoxin



factor 2C;

ATPase;
NM_014982.2)

alpha;



NM_001308002.1)

NM_001253866.1)


NM_000595.3)



MICAL3


PDXDC2 (pyridoxal

SGCB



(microtubule


dependent

(sarcoglycan,



associated


decarboxylase

beta (43 kDa



monooxygenase,


domain containing 2,

dystrophin-



calponin


pseudogene;

associated



and LIM domain


NR_003610.1)

glycoprotein);



containing 3;




NM_000232.4)



NM_001136004.3)



MS4A1


PHC3 (polyhomeotic

SYNGR3



(membrane


homolog 3;

(synaptogyrin 3;



spanning 4-domains


NM_001308116.1)

NM_004209.5)



A1; NM_021950.3)



OSBPL10


POLR2J2 (RNA

ZBTB32



(oxysterol


polymerase

(zinc finger



binding


II subunit

and BTB domain



protein like 10;


J2; NM_032959.5)

containing 32;



NM_017784.4)




NM_014383.2)



PNOC


PSPC1 (paraspeckle



(prepronociceptin;


component 1;



NM_001284244.1)


NM_001042414.2)



QRSL1


REPS1 (RALBP1



(glutaminyl-


associated Eps



tRNA synthase


domain containing 1;



(glutamine-


NM_001128617.2)



hydrolyzing)-



like 1;



NM_018292.4)



SCN3A (sodium


RP11-74E24.2 (zinc



voltage-gated


finger CCCH-type



channel alpha


domain-containing-



subunit 3;


like;



NM_001081677.1)


NM_001271675.1)



SLC15A2


RPP38 (ribonuclease



(solute


P/MRP subunit p38;



carrier


NM_001265601.1)



family 15



member 2;



XM_017007074.1)



SPIB (Spi-B


SLC7A6 (solute



transcription


carrier family 7



factor;


member 6;



NM_001244000.1)


NM_003983.5)



TCL1A (T-cell


SNRPN (small nuclear



leukemia/


ribonucleoprotein



lymphoma 1A;


polypeptide N;



NM_001098725.1)


NM_022807.3)



TNFRSF17


ST3GAL1 (ST3 beta-



(TNF receptor


galactoside alpha-



superfamily


2,3-sialyltransferase



member 17;


1; NM_173344.2)



NM_001192.2)






STX16 (syntaxin 16;






NM_001204868.1)






TIMM8A (translocase






of inner






mitochondrial






membrane 8






homolog A;






NM_001145951.1)






TRAF3IP3 (TRAF3






interacting






protein 3;






NM_001320144.1)






TXK (TXK tyrosine






kinase;






NM_003328.2)






USP9Y (ubiquitin






specific






peptidase 9,






Y-linked;






NG_008311.1)















Th2 cells
TFH
Th17 cells
TReg





Human
ADCY1
B3GAT1
IL17A
FOXP3


Gene
(adenylate
(beta-1,3-
(interleukin
(forkhead


(Gene
cyclase 1;
glucuronyl-
17A;
box P3;


Name;
NM_001281768.1)
transferase 1;
NM_002190.2)
NM_014009.3)


GenBank

NM_018644.3)


Accession
AHI1
BLR1 (c-x-c
IL17RA


No.*)
(Abelson
chemokine
(interleukin 17



helper
receptor
receptor A;



integration
type 5;
NM_014339.6)



site 1;
EF444957.1)



NM_001134831.1)



AI582773
C18orf1
RORC (RAR



(tn17d08.x1
(low density
related orphan



NCI_CGAP_Brn25
lipoprotein
receptor C;




Homo sapiens

receptor
NM_001001523.1)



cDNA clone;
class A domain



AI582773.1)
containing 4;




NM_181481.4)



ANK1
CDK5R1 (cyclin



(ankyrin 1;
dependent kinase 5



NM_020476.2)
regulatory subunit




1; NM_003885.2)



BIRC5
CHGB



(baculoviral
(chromogranin B;



IAP repeat
NM_001819.2)



containing 5;



NM_001012271.1)



CDC25C (cell
CHI3L2



division cycle
(chitinase 3



25C;
like 2;



NM_001318098.1)
NM_001025199.1)



CDC7 (cell
CXCL13



division
(C-X-C motif



cycle 7;
chemokine



NM_001134420.1)
ligand 13;




NM_006419.2)



CENPF
HEY1



(centromere
(hes related



protein F;
family bHLH



NM_016343.3)
transcription




factor with




YRPW motif 1;




NM_001282851.1)



CXCR6
HIST1H4K



(killer cell
(histone



lectin like
cluster 1



receptor B1;
H4 family



NM_002258.2)
member k;




NM_003541.2)



DHFR
ICA1 (islet cell



(dihydrofolate
autoantigen 1;



reductase;
NM_001136020.2)



NM_001290354.1)



EVI5 (ecotropic
KCNK5 (potassium



viral
two pore domain



integration
channel



site 5;
subfamily K



NM_001308248.1)
member 5;




NM_003740.3)



GATA3 (GATA
KIAA1324



binding
(KIAA1324;



protein 3;
NM_001284353.1)



NM_001002295.1)



GSTA4
MAF (MAF bZIP



(glutathione S-
transcription



transferase alpha
factor;



4; NM_001512.3)
NM_001031804.2)



HELLS
MAGEH1 (MAGE



(helicase,
family member H1;



lymphoid-
NM_014061.4)



specific;



NM_001289074.1)



IL26
MKL2



(interleukin
(MKL1/myocardin



26;
like 2;



NM_018402.1)
NM_014048.4)



LAIR2
MYO6 (myosin VI;



(leukocyte
NM_001300899.1)



associated



immunoglobulin



like receptor 2;



NM_021270.4)



LIMA1 (LIM
MYO7A (myosin



domain and
VIIA;



actin
NM_001127179.2)



binding 1;



NM_001243775.1)



MB (myoglobin;
PASK (PAS domain



NM_203377.1)
containing




serine/threonine




kinase;




NM_001252119.1)



MICAL2
PDCD1



(microtubule
(programmed cell



associated
death 1;



monooxygenase,
NM_005018.2)



calponin



and LIM



domain



containing 2;



NM_001282663.1)



NEIL3
POMT1 (protein O-



(nei like DNA
mannosyl-



glycosylase
transferase 1;



3; NM_018248.2)
NM_001136114.1)



PHEX (phosphate
PTPN13 (protein



regulating
tyrosine



endopeptidase
phosphatase, non-



homolog, X-
receptor type 13;



linked;
NM_080685.2)



NM_000444.5)



PMCH (pro-
PVALB



melanin
(parvalbumin;



concentrating
NM_001315532.1)



hormone;



NM_002674.3)



PTGIS (I2
SH3TC1 (SH3



synthase;
domain and



NM_000961.3)
tetratricopeptide




repeats 1;




NM_018986.4)



SLC39A14
SIRPG (signal



(solute carrier
regulatory



family 39
protein gamma;



member 14;
NM_018556.3)



NM_001135153.1)



SMAD2 (SMAD
SLC7A10 (solute



family member 2;
carrier family 7



NM_001135937.2)
member 10;




NM_019849.2)



SNRPD1 (small
SMAD1 (SMAD



nuclear
family member 1;



ribonucleoprotein
NM_001003688.1)



D1 polypeptide;



NM_001291916.1)



WDHD1 (WD
ST8SIA1



repeat and HMG-
(ST8 alpha-



box DNA binding
N-acetyl-



protein 1;
neuraminide alpha-



NM_001008396.2)
2,8-




sialyltransferase




1;




NM_001304450.1)




STK39




(serine/threonine




kinase 39;




NM_013233.2)




THADA (THADA,




armadillo repeat




containing;




NM_001271644.1)




TOX (thymocyte




selection associated




high mobility group




box; NM_014729.2)




TSHR (thyroid




stimulating




hormone receptor;




NM_000369.2)




ZNF764 (zinc finger




protein 764;




NM_001172679.1)
















CD8 T cells
Tgd
Cytotoxic cells







Human
ABT1
C1orf61
APBA2 (amyloid



Gene
(activator
(chromosome 1
beta precursor



(Gene
of basal
open reading
protein binding



Name;
transcription 1;
frame 61;
family A member



GenBank
NM_013375.3)
NM_006365.2)
2; NM_005503.3)



Accession
AES (amino-
CD160 (CD160
APOL3



No.*)
terminal
molecule;
(apolipoprotein




enhancer
NM_007053.3)
L3;




of split;

NM_014349.2)




NM_198969.1)




APBA2 (amyloid
FEZ1
CTSW




beta precursor
(Fasciculation
(cathepsin W;




protein binding
And Elongation
NM_001335.3)




family A member 2;
Protein Zeta 1;




NM_001130414.1)
AF123659.1)




ARHGAP8 (Rho
TARP (TCR
DUSP2 (dual




GTPase activating
gamma alternate
specificity




protein 8;
reading frame
phosphatase 2;




NM_001198726.1)
protein;
NM_004418.3)





NM_001003806.1)




C12orf47
TRD (T cell
GNLY




(MAPKAPK5
receptor alpha
(granulysin;




antisense RNA 1;
delta locus;
NM_012483.3)




NR_015404.1)
NG_001332.3)




C19orf6
TRGV9 (Tcell
GZMA (granzyme




(transmembrane
receptor gamma
A; NM_006144.3)




protein 259;
V region 9;




NM_001033026.1)
X69385.1)




C4orf15

GZMH




(HAUS augmin

(granzyme H;




like complex

NM_001270781.1)




subunit 3;




NM_001303143.1)




CAMLG

KLRB1




(calcium

(killer cell




modulating

lectin like




ligand;

receptor B1;




NM_001745.3)

NM_002258.2)




CD8A (CD8a

KLRD1




molecule;

(killer cell




NM_001768.6)

lectin like






receptor D1;






NM_001114396.1)




CD8B (CD8b

KLRF1




molecule;

(killer cell




NM_001178100.1)

lectin like






receptor F1;






NM_001291822.1)




CDKN2AIP

KLRK1




(CDKN2A

(killer cell




interacting

lectin like




protein;

receptor K1;




NM_001317343.1)

NM_007360.3)




DNAJB1

NKG7




(DnaJ heat

(natural




shock protein

killer




family (Hsp40)

cell granule




member B1;

protein 7;




NM_001313964.1)

NM_005601.3)




FLT3LG (fms

RORA (RAR




related tyrosine

related orphan




kinase 3 ligand;

receptor A;




NM_001278638.1)

NM_134262.2)




GADD45A (growth

RUNX3 (runt




arrest and DNA

related




damage inducible

transcription




alpha;

factor 3;




NM_001199742.1)

NM_004350.2)




GZMM (granzyme

SIGIRR




M;

(single Ig




NM_001258351.1)

and TIR domain






containing;






NM_001135054.1)




KLF9

WHDC1L1




(Kruppel like

(WAS protein




factor 9;

homolog




NM_001206.2)

associated with






actin, golgi






membranes and






microtubules






pseudogene 3;






NR_003521.1)




LEPROTL1

ZBTB16 (zinc




(leptin

finger and BTB




receptor

domain




overlapping

containing 16;




transcript-

NM_001018011.1)




like 1;




NM_001128208.1)




LIME1 (Lck




interacting




transmembrane




adaptor 1;




NM_017806.3)




MYST3 (MYST




histone




acetyl-




transferase




(monocytic




leukemia) 3;




NM_006766.4)




PF4




(platelet




factor 4;




NM_002619.3)




PPP1R2 (protein




phosphatase 1




regulatory




inhibitor




subunit 2;




NM_001291504.1)




PRF1




(perforin 1;




NM_005041.4)




PRR5




(proline




rich 5;




NM_181333.3)




RBM3




(RNA binding




motif




(RNP1, RRM)




protein 3;




NM_006743.4)




SF1




(splicing




factor 1;




NM_004630.3)




SFRS7




(serine and




arginine rich




splicing




factor 7;




NM_001031684.2)




SLC16A7




(solute carrier




family 16




member 7;




NM_001270622.1)




TBCC (tubulin




folding




cofactor C;




NM_003192.2)




THUMPD1




(THUMP




domain




containing




1; NM_017736.4)




TMC6




(transmembrane




channel like 6;




NM_001321185.1)




TSC22D3




(TSC22




domain family




member 3;




NM_001318470.1)




VAMP2 (vesicle




associated




membrane protein




2; NM_014232.2)




ZEB1 (zinc




finger E-




box binding




homeobox 1;




NM_001128128.2)




ZFP36L2




(ZFP36




ring finger




protein




like 2;




NM_006887.4)




ZNF22




(zinc finger




protein 22;




NM_006963.4)




ZNF609




(zinc finger




protein 609;




NM_015042.1)




ZNF91




(zinc finger




protein 91;




NM_001300951.1)







*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.













TABLE 6B





Innate immune cell (IIC) gene signature immunomarkers for use in the methods provided herein.


Cell Type



















NK cells
NK CD56dim cells
NK CD56bright cells





Human
ADARB1 (adenosine
EDG8 (sphingosine-1-
BG255923


Gene
deaminase, RNA specific
phosphate receptor 5;
(lysophosphatidylcholine


(Gene
B1; NM_001112)
NM_001166215.1)
acyltransferase 4;


Name;


NM_153613.2)


GenBank
AF107846
FLJ20699 (cDNA
DUSP4 (dual specificity


Accession
(neuroendocrine-specific
FLJ20699 fis, clone
phosphatase 4;


No.*)
Golgi protein p55;
KAIA2372; AK000706.1)
NM_057158.3)



AF107846.1)



AL080130 (cDNA
GTF3C1 (general
FOXJ1 (forkhead box J1;



DKFZp434E033 (from clone
transcription factor IIIC
NM_001454.3)



DKFZp434E033);
subunit 1;



AL080130.1)
NM_001286242.1)



ALDH1B1 (aldehyde
GZMB (granzyme B;
MADD (MAP kinase



dehydrogenase 1 family
NM_004131.4)
activating death domain;



member B1; NM_000692.4)

NM_001135944.1)



ARL6IP2 (atlastin GTPase 2;
IL21R (interleukin 21
MPPED1



NM_001330461.1)
receptor;
(metallophosphoesterase




NM_181079.4)
domain containing 1,





mRNA;





NM_001044370.1)



BCL2 (apoptosis regulator
KIR2DL3 (killer cell
MUC3B (mucin 3B cell



(BCL2); NM_000633.2)
immunoglobulin like
surface associated;




receptor, two Ig
JQ511939.1)




domains and long




cytoplasmic tail 3;




NM_015868.2)



CDC5L (cell division cycle 5
KIR2DS1 (killer cell
NIBP (NIK and IKKbetta-



like; NM_001253.3)
immunoglobulin like
binding protein;




receptor, two Ig
AY630619.1)




domains and short




cytoplasmic tail 1;




NM_014512.1)



FGF18 (fibroblast growth
KIR2DS2 (killer cell
PLA2G6 (phospholipase



factor 18; NM_003862.2)
immunoglobulin like
A2 group VI;




receptor, two Ig
NM_001004426.1)




domains and short




cytoplasmic tail 2;




NM_001291700.1)



FUT5 (fucosyltransferase 5;
KIR2DS5 (killer cell
RRAD (Ras related



NM_002034.2)
immunoglobulin like
glycolysis inhibitor and




receptor, two Ig
calcium channel




domains and short
regulator;




cytoplasmic tail 5;
NM_001128850.1)




NM_014513.2)



FZR1 (fizzy/cell division
KIR3DL1 (killer cell
SEPT6 (septin 6;



cycle 20 related 1;
immunoglobulin like
NM_145802.3)



XM_005259573.4)
receptor, three Ig




domains and long




cytoplasmic tail 1;




NM_013289.2)



GAGE2 (G antigen 2;
KIR3DL2 (killer cell
XCL1 (X-C motif



NM_001127212.1)
immunoglobulin like
chemokine ligand 1;




receptor, three Ig
NM_002995.2)




domains and long




cytoplasmic tail 2;




NM_006737.3)



IGFBP5 (insulin like growth
KIR3DL3 (killer cell



factor binding protein 5;
immunoglobulin like



NM_000599.3)
receptor, three Ig




domains and long




cytoplasmic tail 3;




NM_153443.4)



LDB3 (LIM domain binding
KIR3DS1 (killer cell



3; NM_001171611.1)
immunoglobulin like




receptor, three Ig




domains and short




cytoplasmic tail 1;




NM_001083539.2)



LOC643313 (similar to
SPON2 (spondin 2;



hypothetical protein
NM_001199021.1)



LOC284701; XM_933043.1)



LOC730096 (hypothetical
TMEPAI (prostate



protein LOC730096;
transmembrane



NC_000022.9)
protein, androgen




induced 1;




NM_199169.2)



MAPRE3 (microtubule



associated protein RP/EB



family member 3;



NM_001303050.1)



MCM3AP



(minichromosome



maintenance complex



component 3 associated



protein; NM_003906.4)



MRC2 (mannose receptor C



type 2; NM_006039.4)



NCR1 (natural cytotoxicity



triggering receptor 1;



NM_001242357.2)



NM_014114 (PR00097



protein; NM_014114.1)



NM_014274 (transient



receptor potential cation



channel, subfamily V,



member 6; NM_014274.3)



NM_017616 (KN motif and



ankyrin repeat domains 2;



NM_015493.6)



PDLIM4 (PDZ and LIM



domain 4; NM_003687.3)



PRX (periaxin;



NM_020956.2)



PSMD4 (proteasome 26S



subunit, non-ATPase 4;



NM_001330692.1)



RP5-886K2.1 (neuronal



thread protein AD7c-NTP;



AF010144.1)



SLC30A5 (solute carrier



family 30 member 5;



NM_001251969.1)



SMEK1 (protein



phosphatase 4 regulatory



subunit 3A;



NM_001284280.1)



SPN (sialophorin;



NM_003123.4)



TBXA2R (thromboxane A2



receptor; NM_001060.5)



TCTN2 (tectonic family



member 2;



NM_001143850.2)



TINAGL1 (tubulointerstitial



nephritis antigen like 1;



NM_001204415.1)



XCL1 (X-C motif chemokine



ligand 1; NM_002995.2)



XCL2 (X-C motif chemokine



ligand 2; NM_003175.3)



ZNF205 (zinc finger protein



205; NM_001278158.1)



ZNF528 (zinc finger protein



528; NM_032423.2)



ZNF747 (zinc finger protein



747; NM_023931.3)















DC
iDC







Human
CCL13 (C-C motif
ABCG2 (ATP-binding



Gene
chemokine ligand 13;
cassette, sub-family G



(Gene
NM_005408.2)
(WHITE), member 2



Name;

(Junior blood group);



GenBank

NM_001257386.1)



Accession
CCL17 (C-C motif
BLVRB (biliverdin



No.*)
chemokine ligand 17;
reductase B;




NM_002987.2)
NM_000713.2)




CCL22 (C-C motif
CARD9 (caspase




chemokine ligand 22;
recruitment domain




NM_002990.4)
family member 9;





NM_052814.3)




CD209 (CD209 molecule;
CD1A (CD1a molecule;




NM_001144899.1)
NM_001763.2)




HSD11B1 (hydroxysteroid
CD1B (CD1b molecule;




11-beta dehydrogenase
NM_001764.2)




1; NM_001206741.1)




NPR1 (natriuretic peptide
CD1C (CD1c molecule;




receptor 1;
NM_001765.2)




NM_000906.3)




PPFIBP2 (PPFIA binding
CD1E (CD1e molecule;




protein 2; XR_930917.2)
NM_001185115.1)





CH25H (cholesterol 25-





hydroxylase;





NM_003956.3)





CLEC10A (C-type lectin





domain family 10 member





A; NM_001330070.1)





CSF1R (colony stimulating





factor 1 receptor;





NM_001288705.1)





CTNS (cystinosin,





lysosomal cystine





transporter;





NM_001031681.2)





F13A1 (factor XIII a





subunit; AH002691.2)





FABP4 (fatty acid binding





protein 4; NM_001442.2)





FZD2 (frizzled class





receptor 2; NM_001466.3)





GSTT1 (glutathione S-





transferase theta 1;





NM_001293814.1)





GUCA1A (guanylate





cyclase activator 1A;





NM_001319062.1)





HS3ST2 (heparan sulfate





(glucosamine) 3-O-





sulfotransferase 2;





NM_006043.1)





LMAN2L (lectin, mannose





binding 2 like;





NM_001322355.1)





MMP12 (matrix





metallopeptidase 12;





NM_002426.5)





MS4A6A (membrane





spanning 4-domains A6A;





NM_001330275.1)





NM_021941





(chromosome 21 open





reading frame 97;





NM_021941.1)





NUDT9 (nudix hydrolase 9;





NM_001248011.1)





PPARG (peroxisome





proliferator activated





receptor gamma;





NM_005037.5)





PREP (prolyl





endopeptidase;





NM_002726.4)





RAP1GAP (RAP1 GTPase





activating protein;





NM_001330383.1)





SLC26A6 (solute carrier





family 26 member 6;





NM_001281733.1)





SLC7A8 (solute carrier





family 7 member 8;





NR_049767.1)





SYT17 (synaptotagmin 17;





NM_001330509.1)





TACSTD2 (tumor-





associated calcium signal





transducer 2;





NM_002353.2)





TM7SF4 (dendrocyte





expressed seven





transmembrane protein;





NM_001257317.1)





VASH1 (vasohibin 1;





NM_014909.4)















aDC
pDC
Eosinophils





Human
CCL1
IL3RA (interleukin 3 receptor
ABHD2 (abhydrolase


Gene
(Chemokine (C-C
subunit alpha; NM_001267713.1)
domain containing 2;


(Gene
motif) ligand 1;

NM_007011.7)


Name;
NM_002981)


GenBank
EBI3 (Epstein-Barr

ACACB (acetyl-CoA


Accession
virus induced 3;

carboxylase beta;


No.*)
NM_005755.2)

NM_001093.3)



INDO

C9orf156 (tRNA



(indoleamine-

methyltransferase O;



pyrrole 2,3

NM_001330725.1)



dioxygenase;



AY221100.1)



LAMP3 (lysosomal

CAT (catalase;



associated

NM_001752.3)



membrane protein



3; NM_014398.3)



OAS3 (2′-5′-

CCR3 (C-C motif



oligoadenylate

chemokine receptor 3;



synthetase 3;

NM_178329.2)



NM_006187.3)





CLC (Charcot-Leyden





crystal galectin;





NM_001828.5)





CYSLTR2 (cysteinyl





leukotriene receptor





2; NM_001308471.1)





EMR1 (EGF-like





module containing





mucin-like hormone





receptor-like 1;





DQ217942.1)





EPN2 (epsin 2;





NM_001102664.1)





GALC





(galactosylceramidase;





NM_000153.3)





GPR44 (orphan G





protein-coupled





receptor; AF118265.1)





HES1 (hes family bHLH





transcription factor 1;





NM_005524.3)





HIST1H1C (histone





cluster 1 H1 family





member c;





NM_005319.3)





HRH4 (histamine





receptor H4;





NM_001143828.1)





IGSF2





(immunoglobulin





superfamily, member





2; BC130327.1)





IL5RA (interleukin 5





receptor subunit





alpha;





NM_001243099.1)





KBTBD11 (kelch





repeat and BTB





domain containing 11;





NM_014867.2)





KCNH2 (potassium





voltage-gated





channel, subfamily H





(eag-related), member





2; NM_000238.3)





LRP5L (LDL receptor





related protein 5 like;





NM_001135772.1)





MYO15B (myosin XVB;





NM_001309242.1)





RCOR3 (REST





corepressor 3;





NM_001136224.2)





RNASE2 (ribonuclease





A family member 2;





NM_002934.2)





RNU2 (U2 snRNA;





U57614.1)





RRP12 (ribosomal RNA





processing 12





homolog;





NM_001284337.1)





SIAH1 (siah E3





ubiquitin protein





ligase 1;





NM_003031.3)





SMPD3





(sphingomyelin





phosphodiesterase 3;





NM_018667.3)





SYNJ1 (synaptojanin 1;





NM_001160302.1)





TGIF1 (TGFB induced





factor homeobox 1;





NM_174886.2)





THBS1





(thrombospondin 1;





NM_003246.3)





THBS4





(thrombospondin 4;





NM_001306213.1)





TIPARP (TCDD





inducible poly(ADP-





ribose) polymerase;





NM_001184718.1)





TKTL1 (transketolase





like 1;





NM_001145934.1)
















Macrophages
Mast cells
Neutrophils







Human
APOE (apolipoprotein
ABCC4
ALPL (alkaline



Gene
E; NM_001302691.1)
(ATP binding
phosphatase,



(Gene

cassette
liver/bone/kidney;



Name;

subfamily C
NM_001127501.3)



GenBank

member 4;



Accession

NM_001301829.1)



No.*)
ATG7 (autophagy
ADCYAP1 (adenylate
BST1




related 7;
cyclase activating
(bone marrow




NM_001144912.1)
polypeptide 1;
stromal cell





NM_001117.4)
antigen 1;






NM_004334.2)




BCAT1 (branched
CALB2
CD93




chain amino acid
(calbindin 2;
(CD93 molecule;




transaminase 1;
NM_001740.4)
NM_012072.3)




NM_001178094.1)




CCL7 (C-C motif
CEACAM8
CEACAM3




chemokine ligand 7;
(carcinoembryonic
(carcinoembryonic




NM_006273.3)
antigen related cell
antigen related cell





adhesion molecule 8;
adhesion molecule 3;





NM_001816.3)
NM_001277163.2)




CD163 (CD163
CMA1 (chymase 1,
CREB5 (cAMP




molecule;
mast cell;
responsive element




NM_203416.3)
NM_001308083.1)
binding protein 5;






NM_001011666.2)




CD68 (CD68
CPA3
CRISPLD2




molecule;
(carboxypeptidase
(cysteine




NM_001040059.1)
A3; NM_001870.3)
rich secretory






protein






LCCL domain






containing 2;






NM_031476.3)




CD84 (CD84
CTSG (cathepsin G;
CSF3R (colony




molecule;
NM_001911.2)
stimulating




NM_001184881.1)

factor 3






receptor;






NM_172313.2)




CHI3L1
ELA2 (neutrophil
CYP4F3




(chitinase 3
elastase;
(cytochrome




like 1;
EU617980.1)
P450 family 4




NM_001276.2)

subfamily






F member 3;






NM_001199209.1)




CHIT1 (chitinase 1;
GATA2
DYSF (dysferlin;




NM_001270509.1)
(GATA binding
NM_001130455.1)





protein 2;





NM_001145661.1)




CLEC5A
HDC (histidine
FCAR (Fc




(C-type lectin
decarboxylase;
fragment of




domain family 5
NM_002112.3)
IgA receptor;




member A;

NM_133278.3)




NM_001301167.1)




COL8A2 (collagen
HPGD
FCGR3B




type VIII alpha 2
(hydroxyprostaglandin
(Fc fragment




chain;
dehydrogenase 15-
of IgG




NM_001294347.1)
(NAD);
receptor IIIb;





NM_001256307.1)
NM_001271035.1)




COLEC12 (collectin
KIT (KIT proto-
FU11151




subfamily member
oncogene receptor
(hypothetical




12; NM_130386.2)
tyrosine kinase;
protein





NM_000222.2)
FLJ11151;






BC006289.2)




CTSK
LOC339524 (long
FPR1




(cathepsin K;
intergenic
(formyl peptide




NM_000396.3)
non-protein
receptor 1;





coding RNA 1140;
NM_001193306.1)





NR_026985.1)




CXCL5
LOH11CR2A (BCSC-1
FPRL1




(C-X-C motif
isoform;
(formyl peptide




chemokine ligand 5;
AY366508.1)
receptor-like




NM_002994.4)

receptor;






M84562.1)




CYBB
MAOB (monoamine
G0S2 (G0/G1




(cytochrome b-
oxidase B;
switch 2;




245 beta chain;
NM_000898.4)
NM_015714.3)




NM_000397.3)




DNASE2B
MLPH (melanophilin;
HIST1H2BC




(deoxyribonuclease 2
NM_001042467.2)
(histone




beta; NM_058248.1)

cluster 1






H2B family






member c;






NM_003526.2)




EMP1 (epithelial
MPO
HPSE (heparanase;




membrane protein 1;
(myeloperoxidase;
NM_001098540.2)




NM_001423.2)
NM_000250.1)




FDX1 (ferredoxin 1;
MS4A2 (membrane
IL8RA




NM_004109.4)
spanning 4-domains
(interleukin 8





A2; NM_001256916.1)
receptor alpha;






L19591.1)




FN1
NM_003293 (tryptase
IL8RB




(fibronectin 1;
alpha/beta 1;
(interleukin-8




NM_001306131.1)
NM_003294.3)
receptor type B;






U11878.1)




GM2A (GM2
NR0B1 (nuclear
KCNJ15 (potassium




ganglioside
receptor
voltage-gated channel




activator;
subfamily 0
subfamily J member




NM_000405.4)
group B member 1;
15; NM_001276438.1)





NM_000475.4)




GPC4 (glypican 4;
PGDS
KIAA0329 (tectonin




NM_001448.2)
(hematopoietic
beta-propeller





prostaglandin D
repeat containing 2;





synthase;
NM_014844.4)





NM_014485.2)




KAL1 (anosmin 1;
PPM1H (protein
LILRB2 (leukocyte




NM_000216.3)
phosphatase,
immunoglobulin





Mg2+/Mn2+
like receptor B2;





dependent 1H;
NR_103521.2)





NM_020700.1)




MARCO (macrophage
PRG2
MGAM (maltase-




receptor with
(proteoglycan 2,
glucoamylase;




collagenous
pro eosinophil
NM_004668.2)




structure;
major basic protein;




NM_006770.3)
NM_001302927.1)




ME1 (malic enzyme
PTGS1
MME (membrane




1; NM_002395.5)
(prostaglandin-
metalloendopeptidase;





endoperoxide
NM_007289.2)





synthase 1;





NM_000962.3)




MS4A4A (membrane
SCG2
PDE4B




spanning 4-domains
(secretogranin
(phosphodiesterase




A4A;
II; NM_003469.4)
4B; NM_001297440.1)




NM_001243266.1)




MSR1 (macrophage
SIGLEC6
S100A12




scavenger receptor 1;
(sialic acid
(S100 calcium




NM_138716.2)
binding Ig
binding





like lectin 6;
protein A12;





NM_198845.5)
NM_005621.1)




PCOLCE2
SLC18A2 (solute
SIGLEC5




(procollagen C-
carrier family 18
(sialic acid




endopeptidase
member A2;
binding Ig




enhancer 2;
NM_003054.4)
like lectin 5;




NM_013363.3)

NM_003830.3)




PTGDS
SLC24A3 (solute
SLC22A4




(prostaglandin
carrier family 24
(solute carrier




D2 synthase;
member 3;
family 22 member 4;




NM_000954.5)
NM_020689.3)
NM_003059.2)




RAI14
TAL1
SLC25A37 (solute




(retinoic acid
(T-cell acute
carrier family 25




induced 14;
lymphocytic leukemia
member 37;




NM_001145525.1)
1; X51990.1)
NM_001317812.1)




SCARB2 (scavenger
TPSAB1 (tryptase
TNFRSF10C (TNF




receptor class B
alpha/beta 1;
receptor




member 2;
NM_003294.3)
superfamily




NM_001204255.1)

member 10c;






NM_003841.3)




SCG5 (secretogranin
TPSB2
VNN3 (vanin 3;




V; NM_001144757.2)
(tryptase beta 2;
NM_001291703.1)





NM_024164.5)




SGMS1




(sphingomyelin




synthase 1;




NM_147156.3)




SULT1C2




(sulfotransferase




family 1C member 2;




NM_176825.2)







*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.













TABLE 7







Individual Immunomarkers for use in the methods provided herein.











GenBank


Gene Name
Abbreviation
Accession No.*





Programmed Death Ligand 1
PDL1
NM_014143


programmed death ligand 2
PDL2
AY254343


programmed cell death 1
PDCD1
NM_005018


cytotoxic T-lymphocyte associated
CTLA4
NM_005214


protein 4





*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.













TABLE 8







Interferon (IFN) Genes for use in the methods provided herein.











GenBank


Gene Name
Abbreviation
Accession No.*





Chemokine (C-X-C Motif) Ligand 10
CXCL10
NM_001565


C-X-C motif chemokine ligand 9
CXCL9
NM_002416


Interferon alpha inducible protein 27
IFI27
NM_001130080


Interferon induced protein with
IFIT1
NM_001548


tetratricopeptide repeats 1


interferon induced protein with
IFIT2
NM_001547


tetratricopeptide repeats 2


interferon induced protein with
IFIT3
NM_001549


tetratricopeptide repeats 3


MX dynamin like GTPase 1
MX1
NM_001144925


MX dynamin like GTPase 2
MX2
XM_005260983


2′-5′-oligoadenylate synthetase 1
OAS1
NM_016816


2′-5′-oligoadenylate synthetase 2
OAS2
NM_016817


signal transducer and activator of
STAT1
NM_007315


transcription 1


signal transducer and activator of
STAT2
NM_005419


transcription 2





*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.













TABLE 9







MHC class II genes for use in the methods provided herein.











GenBank


Name
Abbreviation
Accession No.*





CD74

Homo sapiens CD74 molecule (CD74)

NM_001025159


CIITA
class II major histocompatibility
NM_001286402



complex transactivator


CTSH
cathepsin H
NM_004390


HLA-DMA

Homo sapiens major histocompatibility

NM_006120



complex, class II, DM alpha


HLA-DPA1

Homo sapiens major histocompatibility

NM_033554



complex, class II, DP alpha 1


HLA-DPB1
Human MHC class II lymphocyte
M83664



antigen (HLA-DP) beta chain


HLA-DQA1

Homo sapiens major histocompatibility

NM_002122



complex, class II, DQ alpha 1


HLA-DRB1

Homo sapiens major histocompatibility

NM_002124



complex, class II, DR beta 1


HLA-DRB5

Homo sapiens major histocompatibility

NM_002125



complex, class II, DR beta 5


HLA-DRB6

Homo sapiens major histocompatibility

NR_001298



complex, class II, DR beta 6


NCOA1

Homo sapiens nuclear receptor

NM_003743



coactivator 1





*Each GenBank Accession Number is a representative or exemplary GenBank Accession Number for the listed gene and is herein incorporated by reference in its entirety for all purposes. Further, each listed representative or exemplary accession number should not be construed to limit the claims to the specific accession number.






In one embodiment, upon determining a patient's HNSCC cancer subtype using any of the methods and classifier biomarkers panels or subsets thereof as provided herein alone or in combination with determining expression of one or more immune cell markers as provided herein and/or expression of HPV genes, the patient is selected for treatment with or administered an immunotherapeutic agent. The immunotherapeutic agent can be a checkpoint inhibitor, monoclonal antibody, biological response modifiers, therapeutic vaccine or cellular immunotherapy.


In another embodiment, the immunotherapeutic agent is a checkpoint inhibitor. In some cases, a method for determining the likelihood of response to one or more checkpoint inhibitors is provided. In one embodiment, the checkpoint inhibitor is a PD-1/PD-LI checkpoint inhibitor. The PD-1/PD-LI checkpoint inhibitor can be nivolumab, pembrolizumab, atezolizumab, durvalumab, lambrolizumab, or avelumab. In one embodiment, the checkpoint inhibitor is a CTLA-4 checkpoint inhibitor. The CTLA-4 checkpoint inhibitor can be ipilimumab or tremelimumab. In one embodiment, the checkpoint inhibitor is a combination of checkpoint inhibitors such as, for example, a combination of one or more PD-1/PD-LI checkpoint inhibitors used in combination with one or more CTLA-4 checkpoint inhibitors.


In one embodiment, the immunotherapeutic agent is a monoclonal antibody. In some cases, a method for determining the likelihood of response to one or more monoclonal antibodies is provided. The monoclonal antibody can be directed against tumor cells or directed against tumor products. The monoclonal antibody can be panitumumab, matuzumab, necitumunab, trastuzumab, amatuximab, bevacizumab, ramucirumab, bavituximab, patritumab, rilotumumab, cetuximab, immu-132, or demcizumab.


In yet another embodiment, the immunotherapeutic agent is a therapeutic vaccine. In some cases, a method for determining the likelihood of response to one or more therapeutic vaccines is provided. The therapeutic vaccine can be a peptide or tumor cell vaccine. The vaccine can target MAGE-3 antigens, NY-ESO-1 antigens, p53 antigens, survivin antigens, or MUC1 antigens. The therapeutic cancer vaccine can be GVAX (GM-CSF gene-transfected tumor cell vaccine), belagenpumatucel-L (allogeneic tumor cell vaccine made with four irradiated NSCLC cell lines modified with TGF-beta2 antisense plasmid), MAGE-A3 vaccine (composed of MAGE-A3 protein and adjuvant AS15), (1)-BLP-25 anti-MUC-1 (targets MUC-1 expressed on tumor cells), CimaVax EGF (vaccine composed of human recombinant Epidermal Growth Factor (EGF) conjugated to a carrier protein), WT1 peptide vaccine (composed of four Wilms' tumor suppressor gene analogue peptides), CRS-207 (live-attenuated Listeria monocytogenes vector encoding human mesothelin), Bec2/BCG (induces anti-GD3 antibodies), GV1001 (targets the human telomerase reverse transcriptase), TG4010 (targets the MUC1 antigen), racotumomab (anti-idiotypic antibody which mimicks the NGcGM3 ganglioside that is expressed on multiple human cancers), tecemotide (liposomal BLP25; liposome-based vaccine made from tandem repeat region of MUC1) or DRibbles (a vaccine made from nine cancer antigens plus TLR adjuvants).


In one embodiment, the immunotherapeutic agent is a biological response modifier. In some cases, a method for determining the likelihood of response to one or more biological response modifiers is provided. The biological response modifier can trigger inflammation such as, for example, PF-3512676 (CpG 7909) (a toll-like receptor 9 agonist), CpG-ODN 2006 (downregulates Tregs), Bacillus Calmette-Guerin (BCG), mycobacterium vaccae (SRL172) (nonspecific immune stimulants now often tested as adjuvants). The biological response modifier can be cytokine therapy such as, for example, IL-2+ tumor necrosis factor alpha (TNF-alpha) or interferon alpha (induces T-cell proliferation), interferon gamma (induces tumor cell apoptosis), or Mda-7 (IL-24) (Mda-7/IL-24 induces tumor cell apoptosis and inhibits tumor angiogenesis). The biological response modifier can be a colony-stimulating factor such as, for example granulocyte colony-stimulating factor. The biological response modifier can be a multi-modal effector such as, for example, multi-target VEGFR: thalidomide and analogues such as lenalidomide and pomalidomide, cyclophosphamide, cyclosporine, denileukin diftitox, talactoferrin, trabecetedin or all-trans-retinmoic acid.


In one embodiment, the immunotherapy is cellular immunotherapy. In some cases, a method for determining the likelihood of response to one or more cellular therapeutic agents. The cellular immunotherapeutic agent can be dendritic cells (DCs) (ex vivo generated DC-vaccines loaded with tumor antigens), T-cells (ex vivo generated lymphokine-activated killer cells; cytokine-induce killer cells; activated T-cells; gamma delta T-cells), or natural killer cells.


In some cases, specific subtypes of HNSCC have different levels of immune activation (e.g., innate immunity and/or adaptive immunity) such that subtypes with elevated or detectable immune activation (e.g., innate immunity and/or adaptive immunity) are selected for treatment with one or more immunotherapeutic agents described herein. In some cases, specific subtypes of HNSCC have high or elevated levels of immune activation. In some cases, the MS subtype of AD has elevated levels of immune activation (e.g., innate immunity and/or adaptive immunity) as compared to other HNSCC subtypes. In some cases, the HPV positive, AT-like subtype of HNSCC has elevated levels of immune activation (e.g., innate immunity and/or adaptive immunity) as compared to other HNSCC subtypes. In one embodiment, HNSCC subtypes with low levels of or no immune activation (e.g., innate immunity and/or adaptive immunity) are not selected for treatment with one or more immunotherapeutic agents described herein.


Radiotherapy

In one embodiment, provided herein is a method for determining whether a HNSCC cancer patient is likely to respond to radiotherapy by determining the subtype of HNSCC of a sample obtained from the patient and, based on the HNSCC subtype, assessing whether the patient is likely to respond to radiotherapy. In another embodiment, provided herein is a method of selecting a patient suffering from HNSCC for radiotherapy by determining a HNSCC subtype of a sample from the patient and, based on the HNSCC subtype, selecting the patient for radiotherapy. The determination of the HNSCC subtype of the sample obtained from the patient can be performed using any method for subtyping HNSCC known in the art. The determination of the HNSCC subtype of the sample obtained from the patient can be performed using any method for subtyping HNSCC provided herein. In some embodiments, the method for HNSCC subtyping includes detecting expression levels of a classifier biomarker set alone or in combination with one or more biomarkers of HPV. The classifier biomarker set can be a set of biomarkers from a publically available database such as, for example, TCGA HNSCC RNASeq gene expression dataset(s) or any other dataset provided herein. In some embodiments, the detecting includes all of the classifier biomarkers of Table 1 or Table 3 or any other dataset provided herein at the nucleic acid level or protein level. In one embodiment, from about 1 to about 5, about 5 to about 10, from about 5 to about 15, from about 5 to about 20, from about 5 to about 25, from about 5 to about 30, from about 5 to about 35, from about 5 to about 40, from about 5 to about 45, from about 5 to about 50, from about 5 to about 55, from about 5 to about 60, from about 5 to about 65, from about 5 to about 70, from about 5 to about 75, or from about 5 to about 80 of the biomarkers in any of the HNSCC gene expression datasets provided herein, including, for example, Table 1 or Table 3 for an HNSCC sample are detected in a method to determine the HNSCC subtype as provided herein. In another embodiment, each of the biomarkers from any one of the HNSCC gene expression datasets provided herein, including, for example, Table 1 or Table 3 for an HNSCC sample are detected in a method to determine the HNSCC subtype as provided herein. Further to the above embodiments, the HPV status can be determined by measuring one or more biomarkers of HPV as described herein.


In some embodiments, the radiotherapy can include but are not limited to proton therapy and external-beam radiation therapy. In some embodiments, the radiotherapy can include any types or forms of treatment that is suitable for HNSCC patients. In some embodiments, the surgery can include laser technology, excision, lymph node dissection or neck dissection, and reconstructive surgery.


In some embodiments, an HNSCC can have or display resistance to radiotherapy. Radiotherapy resistance in any HNSCC subtype can be determined by measuring or detecting the expression levels of one or more genes known in the art and/or provided herein associated with or related to the presence of radiotherapy resistance. Genes associated with radiotherapy resistance can include NFE2L2, KEAP1 and CUL3. In some embodiments, radiotherapy resistance can be associated with the alterations of KEAP1 (Kelch-like ECH-associated protein 1)/NRF2 (nuclear factor E2-related factor 2) pathway. Association of a particular gene to radiotherapy resistance can be determined by examining expression of said gene in one or more patients known to be radiotherapy non-responders and comparing expression of said gene in one or more patients known to be radiotherapy responders. In one embodiment, the HNSCC subtype that has radiotherapy resistance can be a CL subtype. In some embodiments, the HNSCC subtype that has radiotherapy resistance can be a BA subtype. In some embodiments, the HNSCC subtype that has radiotherapy resistance can be a MS subtype. In some embodiments, the HNSCC subtype that has radiotherapy resistance can be an AT subtype. In some embodiments, the HNSCC subtype that has radiotherapy resistance can be any HNSCC subtypes. In one embodiment, the HNSCC subtype is a CL subtype. The HNSCC patient can be HPV-negative or positive. In some embodiments, the methods for clinical applications as described herein can determine radiotherapy resistance for surgically resectable HPV-negative or HPV-positive HNSCC cases.


Surgical Intervention

In one embodiment, provided herein is a method for determining whether a HNSCC cancer patient is likely to respond to surgical intervention by determining the subtype of HNSCC of a sample obtained from the patient and, based on the HNSCC subtype, assessing whether the patient is likely to respond to surgery. In another embodiment, provided herein is a method of selecting a patient suffering from HNSCC for surgery by determining a HNSCC subtype of a sample from the patient and, based on the HNSCC subtype, selecting the patient for surgery. The determination of the HNSCC subtype of the sample obtained from the patient can be performed using any method for subtyping HNSCC known in the art. The determination of the HNSCC subtype of the sample obtained from the patient can be performed using any method for subtyping HNSCC provided herein. In some embodiments, the method for HNSCC subtyping includes detecting expression levels of a classifier biomarker set alone or in combination with one or more biomarkers of HPV. The classifier biomarker set can be a set of biomarkers from a publically available database such as, for example, TCGA HNSCC RNASeq gene expression dataset(s) or any other dataset provided herein. In some embodiments, the detecting includes all of the classifier biomarkers of Table 1 or Table 3 or any other dataset provided herein at the nucleic acid level or protein level. In one embodiment, from about 1 to about 5, about 5 to about 10, from about 5 to about 15, from about 5 to about 20, from about 5 to about 25, from about 5 to about 30, from about 5 to about 35, from about 5 to about 40, from about 5 to about 45, from about 5 to about 50, from about 5 to about 55, from about 5 to about 60, from about 5 to about 65, from about 5 to about 70, from about 5 to about 75, or from about 5 to about 80 of the biomarkers in any of the HNSCC gene expression datasets provided herein, including, for example, Table 1 or Table 3 for an HNSCC sample are detected in a method to determine the HNSCC subtype as provided herein. In another embodiment, each of the biomarkers from any one of the HNSCC gene expression datasets provided herein, including, for example, Table 1 or Table 3 for an HNSCC sample are detected in a method to determine the HNSCC subtype as provided herein. Further to the above embodiments, the HPV status can be determined by measuring one or more biomarkers of HPV as described herein.


In some embodiments, surgery approaches for use herein can include but are not limited to minimally invasive or endoscopic head and neck surgery (eHNS), Transoral Robotic Surgery (TORS), Transoral Laser Microsurgery (TLM), Endoscopic Thyroid and Neck Surgery, Robotic Thyroidectomy, Minimally Invasive Video-Assisted Thyroidectomy (MIVAT), and Endoscopic Skull Base Tumor Surgery. In some embodiments, the surgery can include any types of surgical treatment that is suitable for HNSCC patients. In one embodiment, the suitable treatment is surgery.


Prediction of Overall Survival Rate and Metastasis for HNSCC Patients

The present disclosure provides methods for predicting overall survival rate for a HNSCC patient. In some embodiments, the prediction of overall survival rate can involve obtaining a head and neck tissue sample for a HNSCC patient. In some embodiments, the HNSCC patients can have various stages of cancers. In some embodiments, the overall survival rate can be determined by detecting the expression level of at least one subtype classifier of a publically available head and neck cancer database or dataset. In some embodiments, an overall survival rate can be determined by detecting the expression level (e.g., protein and/or nucleic acid) of any subtype classifiers that are relevant to HNSCC. In one embodiment, the subtype classifiers can be all or a subset of classifiers from Table 1 or Table 3. The method can further entail determining the HPV status of the HNSCC patient. HPV status can be determined as provided herein. The HNSCC patient or subject can be HPV-negative or HPV-positive.


In some embodiments, the present disclosure further provide methods of predicting overall survival in HNSCC from specific areas of the head and neck such as, for example, the oral cavity (i.e., oral cavity squamous cell carcinoma (OCSCC)) or larynx (i.e., larynx squamous cell carcinoma (LSCC)). In some embodiments, the prediction includes detecting an expression level of at least one gene from an HNSCC dataset (e.g., Table 1 or Table 3) in a head and neck tissue sample (e.g., sample from oral cavity or larynx) obtained from a patient. In some embodiments, the detection of the expression level of a subtype classifier from an HNSCC dataset (e.g., Table 1 or Table 3) using the methods provided herein specifically identifies a BA, MS, AT or CL OCSCC or LSCC subtype. In some embodiments, the identification of the OCSCC subtype is indicative of the overall survival in the patient. A mesenchymal subtype of OCSCC as ascertained by measuring one or more subtype classifiers in a sample obtained from an OCSCC patient as provided herein can indicate a poor overall survival of an OCSCC patient as compared to patients with other subtypes of OCSCC. In some embodiments, the identification of the LSCC subtype is indicative of the overall survival in the patient. A classical subtype of LSCC as ascertained by measuring one or more subtype classifiers in a sample obtained from a LSCC patient as provided herein can indicate a poor overall survival of a LSCC patient as compared to patients with other subtypes of LSCC.


The present disclosure provides methods for predicting nodal metastasis for a HNSCC patient. In some embodiments, the prediction of nodal metastasis can involve obtaining a head and neck tissue sample for a HNSCC patient. In some embodiments, the HNSCC patients can have various stages of cancers. In some embodiments, the nodal metastasis can be determined by detecting the expression level of at least one subtype classifier from a head and neck gene set. The head and neck gene set can be a publically available head and neck database or a head and neck gene set provided herein (e.g. Table 1 or Table 3) or a combination thereof. The publically available head and neck gene set can be the TCGA HNSCC gene set. In one embodiment, nodal metastasis of HNSCC can be determined by detecting the expression level of all the subtype classifiers or subsets thereof of the classifiers found in Table 1 or Table 3. The HNSCC subject can be HPV-negative or HPV-positive.


In some embodiments, the MS subtype of HNSCC can be more likely to be associated with nodal metastasis compared with other subtypes such as CL, BA or AT. In some embodiments, the OCSCC MS subtype can be most likely associated with positive lymph node metastasis compared with other OCSCC subtypes such as CL, BA or AT. In some embodiments, the OCSCC MS subtype can be at least about 0.1 times, at least about 0.2 times, at least about 0.3 times, at least about 0.4 times, at least about 0.5 times, at least about 0.6 times, at least about 0.7 times, at least about 0.8 times, at least about 0.9 times, at least about 1 time, at least about 1.2 times, at least about 1.5 times, at least about 1.7 times, at least about 2.0 times, at least about 2.2 times, at least about 2.5 times, at least about 2.7 times, at least about 3.0 times, at least about 3.2 times, at least about 3.5 times, at least about 3.7 times, at least about 4.0 times, at least about 4.2 times, at least about 4.5 times, at least about 4.7 times, at least about 5.0 times, inclusive of all ranges and subranges therebetween, more likely to have occult nodal metastasis compared to other OCSCC subtypes such as CL, BA or AT. In one embodiment, the OCSCC MS subtype can be at least about 3 times more likely to have occult nodal metastasis compared to the BA subtype.


Detection Methods

In one embodiment, the methods and compositions provided herein allow for the detection of at least one nucleic acid in a HNSCC sample obtained from a subject. The at least one nucleic acid can be a classifier biomarker and/or HPV gene(s) provided herein. In one embodiment, the at least one nucleic acid detected using the methods and compositions provided herein are selected from Table 1 or Table 3 alone or in combination with one or more HPV genes. In one embodiment, the methods of detecting the nucleic acid(s) (e.g., classifier biomarkers) in the HNSCC sample obtained from the subject comprises, consists essentially of, or consists of measuring the expression level of at least one or a plurality of biomarkers using any of the methods provided herein. The biomarkers can be selected from Table 1 or Table 3. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or all 144 biomarkers nucleic acids of Table 1. In some cases, the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or all 80 biomarkers nucleic acids of Table 1. The detection can be at the nucleic acid level. The detection can be by using any amplification, hybridization and/or sequencing assay disclosed herein.


In another embodiment, the methods and compositions provided herein allow for the detection of at least one nucleic acid or a plurality of nucleic acids in a head and neck cancer sample (e.g. HNSCC sample) obtained from a subject such that the at least one nucleic acid is or the plurality of nucleic acids are selected from the biomarkers listed in Table 1 or Table 3 alone or in combination with one or more HPV genes and the detection of at least one biomarker from a set of biomarkers whose presence, absence and/or level of expression is indicative of immune activation. The set of biomarkers for indicating immune activation can be gene expression signatures of and/or Adaptive Immune Cells (AIC) (e.g., Table 6A) and/or Innate Immune Cells (IIC) (e.g., Table 6B), individual immune biomarkers (e.g., Table 7), interferon genes (e.g., Table 8), major histocompatibility complex, class II (MHC II) genes (e.g., Table 9) or a combination thereof. The gene expression signatures of both IIC and AIC can be any gene signatures known in the art such as, for example, the gene signature listed in Bindea et al. (Immunity 2013; 39(4); 782-795). The detection can be at the nucleic acid level. The detection can be by using any amplification, hybridization and/or sequencing assay disclosed herein.


Kits

Kits for practicing the methods of the invention can be further provided. By “kit” can encompass any manufacture (e.g., a package or a container) comprising at least one reagent, e.g., an antibody, a nucleic acid probe or primer, etc., for specifically detecting the expression of a biomarker of the invention. The kit may be promoted, distributed, or sold as a unit for performing the methods of the present invention. Additionally, the kits may contain a package insert describing the kit and methods for its use.


In one embodiment, kits for practicing the methods of the invention are provided. Such kits are compatible with both manual and automated immunocytochemistry techniques (e.g., cell staining). These kits comprise at least one antibody directed to a biomarker of interest, chemicals for the detection of antibody binding to the biomarker, a counterstain, and, optionally, a bluing agent to facilitate identification of positive staining cells. Any chemicals that detect antigen-antibody binding may be used in the practice of the invention. The kits may comprise 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, or more antibodies for use in the methods of the invention.


EXAMPLES

The present invention is further illustrated by reference to the following Examples. However, it should be noted that these Examples, like the embodiments described above, are illustrative and are not to be construed as restricting the scope of the invention in any way.


Example 1—Development and Validation of the Head and Neck Squamous Cell Carcinoma (HNSCC) Subtyping Signature
Background

Head and Neck Squamous Cell Carcinoma (HNSCC) is comprised of cancers arising from the oral cavity, oropharynx, nasopharynx, hypopharynx, and larynx are responsible for approximately 3% of all malignancies (NCI HNSCC https://www.cancer.gov/types/head-and-neck/hp accessed 6-7-17). The most significant predisposing factors include heavy smoking and/or alcohol use, and more recently an increasing proportion of HNSCC tumors caused by Human Papilloma Virus (HPV) Infection. In the United States, it is projected that there are approximately 60,000 new cases and 12,000 deaths in 2015 [1]. HNSCC is traditionally managed with surgery, radiation therapy, and/or chemotherapy; early stage tumors are often managed with a single treatment modality while advanced stage tumors require multimodality therapy. Risk stratification and treatment decisions vary by anatomic site, stage at presentation, histologic characteristics of the tumor, and patient factors.


Recent advances in cancer genomics have led to an increased understanding of mutational and gene expression profiles in HNSCC. HNSCC subtypes, as defined by underlying genomic features, have shown varied cell of origin, tumor drivers, proliferation, immune responses, and prognosis [2,3,4]. While traditionally associated with tobacco and alcohol use, an increased number of incident oropharyngeal cancers are caused by human papillomavirus (HPV), thus there has been a growing interest in studies of HPV associated HNSCC tumors. With the exception of the use of P16 immunohistochemistry as a marker of HPV infection in oropharyngeal tumors, the molecular characteristics of HNSCC have largely not been incorporated into risk stratification, drug response stratification, nor clinical management decisions (chemotherapy, etc).


Objective

This example was initiated to address the need for an efficient method for improved tumor classification that could inform prognosis, drug response and patient management based on underlying genomic and biologic tumor characteristics. Using multiple available public datasets, including the TCGA [2] and Von Walter et al. (GSE 39366) [3] Keck et al. (GSE 40774) [4] and Wichman et al. (GSE 65858) [5] and the accompanying HPV gene expression results, where available, an HNSCC subtyping method and algorithm was developed. The diagnostic method developed in this example includes evaluation of gene expression subtypes followed by HPV gene expression and application of an algorithm for categorization of HNSCC tumors into one of 5 subtypes (Atypical (AT), Mesenchymal (MS), Classical (CL), Basal (BA), and HPV “Atypical-like”).


Methods

The TCGA HNSCC gene expression data [2] generated from whole-transcriptome RNAseq was examined using the 840-gene classifier previously developed by Von Walter et al. [3] to differentiate the BA, CL, MS and AT HNSCC subtypes. To develop a reduced and clinically applicable gene signature for evaluation of HNSCC tumors, a 5-fold cross-validation (CV) on the entire TCGA HNSCC dataset (n=520) was conducted to find the number of genes that would be required to provide differentiation of the subtypes with sufficient agreement with the previously developed gold standard (i.e., aforementioned 840-gene classifier) as shown in FIG. 1. Prototype samples were then chosen based on the gold standard silhouette (n=416 samples selected). A minimal gene set that optimally classified the Basal (BA), Classical (CL), Mesenchymal (MS), and Atypical (AT) subtypes was identified using a modification of the software packages ClaNC [6]. The Clanc t-statistics were calculated for all 840 gold standard subtyping genes [2.3] using the prototype samples and 144 genes were selected based on the ranks of the strongest t-statistics using a 50% high and 50% low modification (i.e., select an equal number of negatively and positively correlated genes for each HNSCC subtype). A nearest centroid classifier was fit using the 144 genes and the prototypes only followed by an evaluation of the full TCGA dataset (n=520). Validation of the reduced gene signature was compared to the gold standard 840 gene signature in TCGA datasets and in several other publicly available datasets including Keck, Von Walter, and Wichman, references [3,4,5]. FIG. 11 and FIG. 16 depict the patient characteristics for the datasets used to develop (i.e., TCGA HNSCC dataset) and validate (i.e., TCGA, Keck, von Walter and Wichman) the HNSCC 144 subtyping gene signature. Agreement with the gold standard was measured on the full TCGA dataset as well as the remaining datasets. The Keck data set was markedly enriched for HPV+ samples and HNSCC subtyping took this into account by adjusting gene centering values to reflect the HPV distribution in the training set using methods similar to Tibshirani et al. [7]. In addition, signatures that used subsets of the 144 genes were evaluated and compared to the gold standard and 144 subtyping gene signature.


Samples were also evaluated for ongoing HPV replication by evaluation of HPV gene expression. Ongoing HPV replication was assessed by RNAseq evaluation of HPV aligned sequences in HPV types 16, 18, 33, and 35 at levels >1000 counts. HPV reference sequence data was based on the PaVe website: https://pave.niaid.nih.gov/. Read Counts of >1000 for HPV RNAseq (TCGA) or HPV E6 gene expression [4] were used as the criterion for ongoing HPV replication and an HPV positive tumor designation.


Most HPV positive HNSCC tumors subtype as “Atypical”, however not all HPV positive tumors are “atypical”. For those tumors with >1000 HPV read counts and a gene expression subtype other than “atypical”, the correlation to the nearest centroid, to the Atypical centroid, and the silhouette score were evaluated to assist in categorizing that tumor as HPV “atypical-like” group or as one of the other non-HPV gene expression subtypes (Basal, Classical, or Mesenchymal). In addition to atypical subtype, HPV+ samples, a high correlation to the atypical subtype with a low silhouette score was used to assess the need for inclusion of additional HPV positive samples into the HPV positive “atypical-like” subtype. The survival differences between HPV “atypical-like” as compared to other non-atypical HPV positive subtypes was evaluated.


Results
Development of the 144 Gene Signature

The 144 gene signature gene list developed in this study is shown in Table 1, while the 80 gene signature gene list can be found in Table 2. Agreement of subtype calls using the 144 gene signature with the published 840 gold standard gene signature subtype call in several different test datasets is shown in FIG. 12. The newly developed 144 gene signature demonstrated agreement of 0.87 in the TCGA dataset and a range of 0.83-0.86 in the other 3 test datasets. FIG. 11 and FIG. 16 provide a summary of the test datasets. The smaller gene signature (80 genes) showed marginally lower concordance to the gold standard (84% vs. 87%) than the 144 gene signature (see FIGS. 14 and 15).


Evaluation of HPV Status and Assignment to the HPV “Atypical-Like” Subtype

The majority but not all of the HPV positive samples (as evaluated by gene expression) belonged to the atypical gene expression subtype (see FIG. 13). As seen in FIG. 13, this observation was true using either the gold standard or 144 gene signature as the subtyping tool. Some tumors displayed biologic characteristics that were more like other subtypes despite the presence of HPV gene expression. These tumors appeared to be more similar to smoking related tumors and were reflective of a different biology and possibly different prognosis. For this reason, a subtyping algorithm was developed that incorporated both the HPV status and the gene expression subtype in identifying 5 relevant subtypes in HNSCC (i.e., Atypical (AT), Mesenchymal (MS), Classical (CL), Basal (BA), and HPV “Atypical-like”).


As shown in the survival curves in comparing HPV atypical vs. HPV positive non-Atypical tumors in TCGA using the 144 gene gold standard (FIG. 2A), the HPV positive samples that did not belong to the atypical gene expression subtype or “atypical-like” subtype demonstrated a worse survival and may be more similar to smoking induced non-HPV HNSCC tumors. This observation was corroborated by the survival curves comparing HPV atypical vs. HPV positive non-Atypical tumors in Keck [4] with and without adjustment by correlation and silhouette score using the 144 gene gold standard (FIG. 3) and survival curves comparing HPV positive atypical to HPV positive non-Atypical tumors in the TCGA dataset with and without adjustment by correlation and silhouette score using the 840 gene gold standard (FIGS. 4A and 4B). It was noted that in FIGS. 4A and 4B, a few non-atypical samples having both high correlation with the atypical centroid and with low silhouettes may reflect underlying biology more similar to the HPV atypical-like subtype. When these samples were included in the HPV+ atypical-like subtype, the survival differences were enhanced (see FIG. 4C). Correlation with the atypical centroid and silhouette scores for the non-atypical HPV positive tumors was included to assist in determining inclusion of an HPV positive tumor in the “HPV atypical-like” subtype (see FIGS. 2B, 3 and 4B).


Conclusion

Development and validation of a 144 and 80 gene signature for HNSCC subtyping was described. The resulting 144 and 80 gene signatures maintain low misclassification rates when applied to several independent test sets. Further, the 144 gene signature in combination with evaluation of HPV status, as defined by HPV gene expression, was developed to classify tumors of HNSCC into 5 subtypes, Basal, Classical, Mesenchymal, Atypical, and HPV Atypical-like. The 5 subtypes showed differences in HPV status, underlying biology, prognosis, immune response, and likely response to a variety of therapeutics. Important differences in prognosis and survival of HPV positive tumors that differ in their gene expression subtype (those that are atypical-like vs HPV tumors that are not atypical-like but rather more closely resemble other HNSCC tumor subtypes) were demonstrated. The different HPV tumors demonstrate differences in prognosis that may be meaningful to therapeutic management.


Incorporation by Reference

The following references are incorporated by reference in their entireties for all purposes.

  • 1.) Siegel R L, Miller K D, Jemal A. Cancer Statistics, 2015. CA Cancer J Clin. 2015; 65: 5-29. doi:10.3322/caac.21254
  • 2.) Lawrence M S, Sougnez C, Lichtenstein L, Cibulskis K, Lander E, Gabriel S B, et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015; 517: 576-582. doi:10.1038/nature14129
  • 3.) Von Walter, Yin X, Wilkerson M D, Cabanski C R, Zhao N, Du Y, Ang M K, Hayward M C, Salazar A H, Hoadley K A, Fritchie K, Sailey C J, Weissler M C, Shockley W W, Zanation A M, Hackman T, Thorne L B, Funkhouser W D, Muldrew K L, Olshan A F, Randell S H, Wright F A, Shores C G, Hayes D N. (2013). Molecular Subtypes in Head and Neck Cancer Exhibit Distinct Patterns of Chromosomal Gain and Loss of Canonical Cancer Genes. PLoS One, 8(2):e56823. PMCID: 3579892.
  • 4.) Keck M K, Zuo Z, Khattri a., Stricker T P, Brown C D, Imanguli M, et al. Integrative Analysis of Head and Neck Cancer Identifies Two Biologically Distinct HPV and Three Non-HPV Subtypes. Clin Cancer Res. 2014; 21: 870-881. doi:10.1158/1078-0432.CCR-14-2481
  • 5.) Wichman G, Rosolowski M, Krohn K, et al. The role of HPV RNA transcription, immune response-related gene expression and disruptive TP53 mutations in diagnostic and prognostic profiling of head and neck cancer. Intl Jrnl Cancer 2015; 137: 2846-2857.
  • 6.) Dabney A R. ClaNC: Point-and-click software for classifying microarrays to nearest centroids. Bioinformatics. 2006; 22: 122-123. doi:10.1093/bioinformatics/bti756
  • 7.) Tibshirani R, Hastie T, Narasimhan B, Chu G. Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci USA. 2002; 99: 6567-72. doi:10.1073/pnas.082099299
  • 8.) Bindea G, Mlecnik B, Tosolini M, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 2013; 39(4):782-95.


Example 2—Immune Cell Activation Differences Among HNSCC Intrinsic Subtypes as Determined Using HNSCC Subtyping Gene Signature from Example 1
Methods

Using previously published Bindea et al. (8) immune cell gene signatures (24 in total) and the TCGA HNSCC gene expression dataset (HNSCC n=520), immune cell expression and immunomarkers were examined in relation to the 5 HNSCC subtypes as defined using the 144 gene signature from Example 1. Gene expression signatures of both Innate Immune Cells (IIC) and Adaptive Immune Cells (AIC) as well as single gene immune biomarkers (CTLA4, PDCD1, and CD274 (PD-L1), PDCDLG2 (PD-L2)) and IFN were examined in all 5 HNSCC subtypes. Immune cell signature associations with tumor subtype and with CD274 expression were evaluated using linear regression. Survival immune cell signature associations were evaluated with stratified cox proportional hazard models allowing for different baseline hazards in each dataset.


Results

Heatmap analysis (FIG. 5) and hierarchical clustering (FIG. 7) of immune cell gene signatures provided separation of intrinsic subtypes of HNSCC. Further, immune cell signature gene expression patterns were consistent across multiple HNSCC (see FIG. 9) datasets. Strength of association of CD274 (PD-L1) expression and individual immune markers versus that for subtype (using the 144 gene signature) was conducted. As shown in FIG. 8, for HNSCC subtypes, association strengths (adjusted R squared from linear model) were mixed showing CD274 association greater for some cells (Treg, Tgd and Th1 cells), while HNSCC subtype association greater for others (B cells, T cells, T helper cells, cytotoxic cells, CD8 T cells, TFH, Th2, Tem, Th17, and Tcm).


Using cox proportional hazard models, subtype specific hazard ratios for one unit of increased expression were calculated. Subtype specific HR's were adjusted for pathologic stage and confidence intervals were calculated. The HR and CI for cell signatures or genes showing significant survival associations for one or more of the subtypes are shown in FIG. 10.


In summary, immune cell expression was significantly different across the subtypes and was often higher in HPV positive tumors and in the Mesenchymal subtype tumors (see FIGS. 5, 6 and 7). Classical and Basal subtypes demonstrated lower immune expression, but were differentiated by the presence of elevated CD274 (PD-L1) and PDCD1LG2 (PD-L2) expression in the basal subtype (FIG. 6). Subtype and HPV status was a better predictor than by CD274 (PD-L1) expression for AIC expression (FIG. 8). Improved survival was associated with increased expression of T memory, T follicular helper, and NK CD56bright cells in the mesenchymal subtype (p<0.05), whereas Th2 cells, CD274 (PD-L1) and gene PDCD1LG2 (PD-L2) were associated with lower survival (p<0.05) (FIG. 6). A lower mutation burden was observed in the HPV “Atypical-like” tumors and improved survival was associated with increased expression of Tgamma delta cells and PDCD1 (PD-1) expression (p=0.01) (see FIG. 6).


Conclusion

Intrinsic biologic subtypes of HNSCC as defined by gene expression and by HPV gene expression reveal key differences in immune cell expression, which were not always correlated with CD274(PD-L1) expression. Accordingly, using the 144 gene signature (or the reduced 80 gene signature) in combination with HPV gene expression, immune cell/marker differences in HNSCC tumors that may inform immunotherapy treatments including checkpoint inhibitors as well as other therapeutic targets was demonstrated.


Incorporation by Reference

The following references are incorporated by reference in their entireties for all purposes.

  • 1.) Siegel R L, Miller K D, Jemal A. Cancer Statistics, 2015. C A Cancer J Clin. 2015; 65: 5-29. doi:10.3322/caac.21254
  • 2.) Lawrence M S, Sougnez C, Lichtenstein L, Cibulskis K, Lander E, Gabriel S B, et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015; 517: 576-582. doi:10.1038/nature14129
  • 3.) Von Walter, Yin X, Wilkerson M D, Cabanski C R, Zhao N, Du Y, Ang M K, Hayward M C, Salazar A H, Hoadley K A, Fritchie K, Sailey C J, Weissler M C, Shockley W W, Zanation A M, Hackman T, Thorne L B, Funkhouser W D, Muldrew K L, Olshan A F, Randell S H, Wright F A, Shores C G, Hayes D N. (2013). Molecular Subtypes in Head and Neck Cancer Exhibit Distinct Patterns of Chromosomal Gain and Loss of Canonical Cancer Genes. PLoS One, 8(2):e56823. PMCID: 3579892.
  • 4.) Keck M K, Zuo Z, Khattri a., Stricker T P, Brown C D, Imanguli M, et al. Integrative Analysis of Head and Neck Cancer Identifies Two Biologically Distinct HPV and Three Non-HPV Subtypes. Clin Cancer Res. 2014; 21: 870-881. doi:10.1158/1078-0432.CCR-14-2481
  • 5.) Wichman G, Rosolowski M, Krohn K, et al. The role of HPV RNA transcription, immune response-related gene expression and disruptive TP53 mutations in diagnostic and prognostic profiling of head and neck cancer. Intl Jrnl Cancer 2015; 137: 2846-2857.
  • 6.) Dabney A R. ClaNC: Point-and-click software for classifying microarrays to nearest centroids. Bioinformatics. 2006; 22: 122-123. doi:10.1093/bioinformatics/bti756
  • 7.) Tibshirani R, Hastie T, Narasimhan B, Chu G. Diagnosis of multiple cancer types by shrunken centroids of gene expression. Proc Natl Acad Sci USA. 2002; 99: 6567-72. doi:10.1073/pnas.082099299
  • 8.) Bindea G, Mlecnik B, Tosolini M, et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 2013; 39(4):782-95.


Numbered Embodiments of the Disclosure

Other subject matter contemplated by the present disclosure is set out in the following numbered embodiments:


1. A method for determining a head and neck squamous cell carcinoma (HNSCC) subtype of a head and neck tissue sample obtained from a patient, the method comprising detecting an expression level of at least one classifier biomarker of Table 1 or Table 3, wherein the detection of the expression level of the classifier biomarker specifically identifies a basal (BA), mesenchymal (MS), atypical (AT) or classical (CL) HNSCC subtype.


2. The method of embodiment 1, wherein the method further comprises comparing the detected levels of expression of the at least one classifier biomarkers of Table 1 or Table 3 to the expression of the at least one classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the sample as BA, MS, AT or CL subtype based on the results of the comparing step.


3. The method of embodiment 2, wherein the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm.


4. The method of any of the above embodiments, wherein the expression level of the classifier biomarker is detected at the nucleic acid level.


5. The method of embodiment 4, wherein the nucleic acid level is RNA or cDNA.


6. The method embodiment 4 or 5, wherein the detecting an expression level comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.


7. The method of embodiment 6, wherein the expression level is detected by performing qRT-PCR.


8. The method of embodiment 7, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers specific for at least one classifier biomarker of Table 1 or Table 3.


9. The method of any of the above embodiments, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.


10. The method of embodiment 9, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


11. The method of any one of the above embodiments, wherein the at least one classifier biomarker comprises a plurality of classifier biomarkers.


12. The method of embodiment 11, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1.


13. The method of any of embodiments 1-10, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 1.


14. The method of embodiment 11, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 5 classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3.


15. The method of any of embodiments 1-10, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 3.


16. The method of any of the above embodiments, wherein the method further comprises determining the HPV status of the patient.


17. The method of embodiment 16, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


18. The method of embodiment 17, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


19. A method for determining a HNSCC subtype of a head and neck tissue sample obtained from a patient comprising detecting an expression level of at least one nucleic acid molecule that encodes a classifier biomarker having a specific expression pattern in head and neck cancer cells, wherein the classifier biomarker is selected from the group consisting of the classifier genes set forth in Table 1 or Table 3, the method comprising: (a) isolating nucleic acid material from a head and neck tissue sample from a patient; (b) mixing the nucleic acid material with oligonucleotides that are substantially complementary to portions of nucleic acid molecule of the classifier biomarker; and (c) detecting expression of the classifier biomarker.


20. The method of embodiment 19, wherein the method further comprises comparing the detected levels of expression of the at least one classifier biomarkers of Table 1 or Table 3 to the expression of the at least one classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the sample as BA, MS, AT or CL subtype based on the results of the comparing step.


21. The method of embodiment 20, wherein the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm.


22. The method of any of embodiments 19-21, wherein the detecting the expression level comprises performing qRT-PCR or any hybridization-based gene assays.


23. The method of embodiment 22, wherein the expression level is detected by performing qRT-PCR.


24. The method of embodiment 23, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers specific for at least one classifier biomarker of Table 1 or Table 3.


25. The method of any of embodiments 19-24, further comprising predicting the response to a therapy for treating a subtype of HNSCC based on the detected expression level of the classifier biomarker.


26. The method of embodiment 25, wherein the therapy is radiotherapy, surgical intervention, chemotherapy, angiogenesis inhibitors and/or immunotherapy.


27. The method of any one of embodiments 19-26, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets or a bodily fluid obtained from the patient.


28. The method of embodiment 27, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


29. The method of any of embodiments 19-28, wherein the at least one nucleic acid molecule that encodes a classifier biomarker comprises a plurality of nucleic acid molecules that encode a plurality of classifier biomarkers.


30. The method of embodiment 29, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1.


31. The method of any of embodiments 19-28, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 1.


32. The method of embodiment 29, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 5 classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3.


33. The method of any of embodiments 19-28, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 3.


34. The method of embodiments 19-33, wherein the method further comprises determining the HPV status of the patient.


35. The method of embodiment 34, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


36. The method of embodiment 35, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


37. A method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method comprising measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay.


38. The method of embodiment 37, wherein the head neck tissue sample was previously diagnosed as being squamous cell carcinoma.


39. The method of embodiment 38, wherein the previous diagnosis was by histological examination.


40. The method of any of embodiments 37-39, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.


41. The method of embodiment 40, wherein the expression level is detected by performing qRT-PCR.


42. The method of embodiment 41, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3.


43. The method of any of embodiments 37-42, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.


44. The method of embodiment 43, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


45. The method of any of embodiments 37-44, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.


46. The method of any of embodiments 37-44, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1.


47. The method of any of embodiments 37-44, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.


48. The method of any of embodiments 37-44, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3.


49. The method of embodiments 37-48, wherein the method further comprises determining the HPV status of the patient.


50. The method of embodiment 49, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


51. The method of embodiment 50, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


52. A method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method consisting essentially of measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay.


53. The method of embodiment 52, wherein the head and neck tissue sample was previously diagnosed as being squamous cell carcinoma.


54. The method of embodiment 53, wherein the previous diagnosis was by histological examination.


55. The method of any of embodiments 52-54, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.


56. The method of embodiment 55, wherein the expression level is detected by performing qRT-PCR.


57. The method of embodiment 56, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3.


58. The method of any of embodiments 52-57, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.


59. The method of embodiment 58, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


60. The method of any of embodiments 52-59, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.


61. The method of any of embodiments 52-59, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1.


62. The method of any of embodiments 52-59, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.


63. The method of any of embodiments 52-59, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3.


64. The method of embodiments 52-63, wherein the method further comprises determining the HPV status of the patient.


65. The method of embodiment 64, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


66. The method of embodiment 65, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


67. A method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method consisting of measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay.


68. The method of embodiment 67, wherein the head and neck tissue sample was previously diagnosed as being squamous cell carcinoma.


69. The method of embodiment 68, wherein the previous diagnosis was by histological examination.


70. The method of any of embodiments 67-69, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.


71. The method of embodiment 70, wherein the expression level is detected by performing qRT-PCR.


72. The method of embodiment 71, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3.


73. The method of any of embodiments 67-72, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.


74. The method of embodiment 73, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


75. The method of any of embodiments 67-74, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.


76. The method of any of embodiments 67-74, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1.


77. The method of any of embodiments 67-74, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.


78. The method of any of embodiments 67-74, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3.


79. The method of embodiments 67-78, wherein the method further comprises determining the HPV status of the patient.


80. The method of embodiment 79, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


81. The method of embodiment 80, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


82. A method of determining whether a HNSCC patient is likely to respond to immunotherapy, the method comprising,

    • determining the HNSCC subtype of a head and neck tissue sample from the patient, wherein the HNSCC subtype is selected from the group consisting of basal, mesenchymal, atypical and classical; and
    • based on the subtype, assessing whether the patient is likely to respond to immunotherapy.


83. A method for selecting a HNSCC patient for immunotherapy, the method comprising, determining a HNSCC subtype of a head and neck tissue sample from the patient, based on the subtype; and selecting the patient for immunotherapy.


84. The method of embodiment 82 or 83, wherein the immunotherapy comprises checkpoint inhibitor therapy.


85. The method of embodiment 84, wherein the checkpoint inhibitor targets PD-1 or PD-L1.


86. The method of embodiment 84, wherein the checkpoint inhibitor targets CTLA-4.


87. The method of embodiment 85, wherein the checkpoint inhibitor is Pembrolizumab, Nivolumab or an antigen fragment binding fragment thereof.


88. The method of embodiment 86, wherein the checkpoint inhibitor is Ipilimumab or an antigen binding fragment thereof.


89. The method of any one of embodiments 82-88, wherein the patient is initially determined to have HNSCC via a histological analysis of a sample.


90. The method of any one of embodiments 82-89, wherein the patient's HNSCC molecular subtype is selected from basal, mesenchymal, atypical or classical and is determined via a histological analysis of a sample obtained from the patient.


91. The method of any one of embodiments 89-90, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, or a bodily fluid obtained from the patient.


92. The method of embodiment 91, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


93. The method of any one of embodiments 82-92, wherein the determining the HNSCC subtype comprises determining expression levels of a plurality of classifier biomarkers.


94. The method of embodiment 93, wherein the determining the expression levels of the plurality of classifier biomarkers is at a nucleic acid level by performing RNA sequencing, reverse transcriptase polymerase chain reaction (RT-PCR) or hybridization based analyses.


95. The method of embodiment 93 or 94, wherein the plurality of classifier biomarkers for determining the HNSCC subtype is selected from a publically available HNSCC dataset.


96. The method of embodiment 95, wherein the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset.


97. The method of embodiment 94, wherein the plurality of classifier biomarkers for determining the HNSCC subtype is selected from Table 1 or Table 3.


98. The method of embodiment 97, wherein the RT-PCR is quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR).


99. The method of embodiment 98, wherein the RT-PCR is performed with primers specific to the plurality of classifier biomarkers of Table 1 or Table 3.


100. The method of any one of embodiments 93-99, further comprising comparing the detected levels of expression of the plurality of classifier biomarkers of Table 1 or Table 3 to the expression of the plurality of classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the first sample as BA, MS, AT or CL based on the results of the comparing step.


101. The method of embodiment 100, wherein the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm.


102. The method of any of embodiments 93-101, wherein the plurality of the classifier biomarkers comprise each of the classifier biomarkers set forth in Table 1 or Table 3.


103. The method of embodiments 82-102, wherein the method further comprises determining the HPV status of the patient.


104. The method of embodiment 103, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


105. The method of embodiment 104, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


106. A method of treating HNSCC in a subject, the method comprising:

    • measuring the expression level of at least one biomarker nucleic acid in a HNSCC sample obtained from the subject, wherein the at least one biomarker nucleic acid is selected from a set of biomarkers listed in Table 1 or Table 3, wherein the presence, absence and/or level of the at least one biomarker indicates a subtype of the HNSCC; and
    • administering an immunotherapeutic agent based on the subtype of the HNSCC.


107. The method of embodiment 106, wherein the head and neck sample is a HNSCC sample.


108. The method of embodiment 107, wherein the at least one biomarker nucleic acid selected from the set of biomarkers comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.


109. The method of embodiment 107, wherein the at least one biomarker nucleic acid selected from the set of biomarkers comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.


110. The method of any of embodiments 106-109, wherein the head and neck tissue sample was previously diagnosed as HNSCC.


111. The method of embodiment 109, wherein the previous diagnosis was by histological examination.


112. The method of any one of embodiments 106-110, further comprising measuring the expression of at least one biomarker from an additional set of biomarkers.


113. The method of embodiment 112, wherein the additional set of biomarkers comprise gene expression signatures of Innate Immune Cells (IIC), Adaptive Immune Cells (AIC), one or more individual immune biomarkers, one or more interferon (IFN) genes, one or more major histocompatibility complex, class II (MHCII) genes or a combination thereof


114. The method of embodiment 113, wherein the additional set of biomarkers comprises genes selected from Tables 6A, 6B, 7, 8, 9, or a combination thereof.


115. The method of embodiment 113, wherein the gene expression signatures of AICs are selected from Table 6A.


116. The method of embodiment 113, wherein the gene expression signature of IICs are selected from Table 6B.


117. The method of embodiment 113, wherein the one or more individual immune biomarkers are selected from Table 7.


118. The method of embodiment 113, wherein the one or more IFN genes are selected from Table 8.


119. The method of embodiment 113, wherein the one or more MHCII genes are selected from Table 9.


120. The method of any of embodiments 106-119, wherein the measuring the expression level is conducted using an amplification, hybridization and/or sequencing assay.


121. The method of embodiment 120, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.


122. The method of embodiment 121, wherein the expression level is detected by performing qRT-PCR.


123. The method of any of embodiments 106-122, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.


124. The method of embodiment 123, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.


125. The method of any one of embodiments 106-124, wherein the subject's HNSCC subtype is selected from basal, mesenchymal, atypical or classical.


126. The method of embodiment 106, wherein the at least one biomarker nucleic acid is a plurality of biomarker nucleic acids, wherein the plurality of biomarker nucleic acids comprises at least one biomarker nucleic acid listed in Table 1 or Table 3 in combination with one or more biomarker nucleic acids from a publically available HNSCC dataset, wherein the presence, absence and/or level of the plurality of biomarker nucleic acids indicates a subtype of the HNSCC.


127. The method of embodiment 106, wherein the at least one biomarker nucleic acid is a plurality of biomarker nucleic acids, wherein the plurality of biomarker nucleic acids comprises all of the biomarker nucleic acids listed in Table 1 or Table 3 in combination with one or more biomarker nucleic acids from a publically available HNSCC dataset, wherein the presence, absence and/or level of the plurality of biomarker nucleic acids indicates a subtype of the HNSCC.


128. The method of embodiment 126 or 127, wherein the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset.


129. The method of embodiments 106-128, wherein the method further comprises determining the HPV status of the patient.


130. The method of embodiment 129, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.


131. The method of embodiment 130, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.


The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent application, foreign patents, foreign patent application and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, application and publications to provide yet further embodiments.


These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure.

Claims
  • 1. A method for determining a head and neck squamous cell carcinoma (HNSCC) subtype of a head and neck tissue sample obtained from a patient, the method comprising detecting an expression level of at least one classifier biomarker of Table 1 or Table 3, wherein the detection of the expression level of the classifier biomarker specifically identifies a basal (BA), mesenchymal (MS), atypical (AT) or classical (CL) HNSCC subtype.
  • 2. The method of claim 1, wherein the method further comprises comparing the detected levels of expression of the at least one classifier biomarkers of Table 1 or Table 3 to the expression of the at least one classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the sample as BA, MS, AT or CL subtype based on the results of the comparing step.
  • 3. The method of claim 2, wherein the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm.
  • 4. The method of any of the above claims, wherein the expression level of the classifier biomarker is detected at the nucleic acid level.
  • 5. The method of claim 4, wherein the nucleic acid level is RNA or cDNA.
  • 6. The method claim 4, wherein the detecting an expression level comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.
  • 7. The method of claim 6, wherein the expression level is detected by performing qRT-PCR.
  • 8. The method of claim 7, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers specific for at least one classifier biomarker of Table 1 or Table 3.
  • 9. The method of claim 1, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.
  • 10. The method of claim 9, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 11. The method of claim 1, wherein the at least one classifier biomarker comprises a plurality of classifier biomarkers.
  • 12. The method of claim 11, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1.
  • 13. The method of claim 1, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 1.
  • 14. The method of claim 11, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 5 classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3.
  • 15. The method of claim 1, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 3.
  • 16. The method of claim 1, wherein the method further comprises determining the HPV status of the patient.
  • 17. The method of claim 16, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 18. The method of claim 17, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
  • 19. A method for determining a HNSCC subtype of a head and neck tissue sample obtained from a patient comprising detecting an expression level of at least one nucleic acid molecule that encodes a classifier biomarker having a specific expression pattern in head and neck cancer cells, wherein the classifier biomarker is selected from the group consisting of the classifier genes set forth in Table 1 or Table 3, the method comprising: (a) isolating nucleic acid material from a head and neck tissue sample from a patient; (b) mixing the nucleic acid material with oligonucleotides that are substantially complementary to portions of nucleic acid molecule of the classifier biomarker; and (c) detecting expression of the classifier biomarker.
  • 20. The method of claim 19, wherein the method further comprises comparing the detected levels of expression of the at least one classifier biomarkers of Table 1 or Table 3 to the expression of the at least one classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the at least one classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the sample as BA, MS, AT or CL subtype based on the results of the comparing step.
  • 21. The method of claim 20, wherein the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm.
  • 22. The method of any of claims 19-21, wherein the detecting the expression level comprises performing qRT-PCR or any hybridization-based gene assays.
  • 23. The method of claim 22, wherein the expression level is detected by performing qRT-PCR.
  • 24. The method of claim 23, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers specific for at least one classifier biomarker of Table 1 or Table 3.
  • 25. The method of claim 19, further comprising predicting the response to a therapy for treating a subtype of HNSCC based on the detected expression level of the classifier biomarker.
  • 26. The method of claim 25, wherein the therapy is radiotherapy, surgical intervention, chemotherapy, angiogenesis inhibitors and/or immunotherapy.
  • 27. The method of claim 19, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets or a bodily fluid obtained from the patient.
  • 28. The method of claim 27, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 29. The method of claim 19, wherein the at least one nucleic acid molecule that encodes a classifier biomarker comprises a plurality of nucleic acid molecules that encode a plurality of classifier biomarkers.
  • 30. The method of claim 29, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 9 classifier biomarkers, at least 18 classifier biomarkers, at least 36 classifier biomarkers, at least 54 classifier biomarkers, at least 72 classifier biomarkers, at least 90 classifier biomarkers, at least 108 classifier biomarkers, at least 126 classifier biomarkers or at least 144 classifier biomarkers of Table 1.
  • 31. The method of claim 19, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 1.
  • 32. The method of claim 29, wherein the plurality of classifier biomarkers comprises at least two classifier biomarkers, at least 5 classifier biomarkers, at least 10 classifier biomarkers, at least 20 classifier biomarkers, at least 30 classifier biomarkers, at least 40 classifier biomarkers, at least 50 classifier biomarkers, at least 60 classifier biomarkers, at least 70 classifier biomarkers or at least 80 classifier biomarkers of Table 3.
  • 33. The method of claim 19, wherein the at least one classifier biomarker comprises all the classifier biomarkers of Table 3.
  • 34. The method of claim 19, wherein the method further comprises determining the HPV status of the patient.
  • 35. The method of claim 34, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 36. The method of claim 35, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
  • 37. A method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method comprising measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay.
  • 38. The method of claim 37, wherein the head neck tissue sample was previously diagnosed as being squamous cell carcinoma.
  • 39. The method of claim 38, wherein the previous diagnosis was by histological examination.
  • 40. The method of any of claims 37-39, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.
  • 41. The method of claim 40, wherein the expression level is detected by performing qRT-PCR.
  • 42. The method of claim 41, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3.
  • 43. The method of claim 37, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.
  • 44. The method of claim 43, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 45. The method of claim 37, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.
  • 46. The method of claim 37, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1.
  • 47. The method of claim 37, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.
  • 48. The method of claim 37, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3.
  • 49. The method of claim 37, wherein the method further comprises determining the HPV status of the patient.
  • 50. The method of claim 49, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 51. The method of claim 50, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
  • 52. A method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method consisting essentially of measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay.
  • 53. The method of claim 52, wherein the head and neck tissue sample was previously diagnosed as being squamous cell carcinoma.
  • 54. The method of claim 53, wherein the previous diagnosis was by histological examination.
  • 55. The method of claim 52, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.
  • 56. The method of claim 55, wherein the expression level is detected by performing qRT-PCR.
  • 57. The method of claim 56, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3.
  • 58. The method of claim 52, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.
  • 59. The method of claim 58, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 60. The method of claim 52, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.
  • 61. The method of claim 52, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1.
  • 62. The method of claim 52, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.
  • 63. The method of claim 52, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3.
  • 64. The method of claim 52, wherein the method further comprises determining the HPV status of the patient.
  • 65. The method of claim 64, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 66. The method of claim 65, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
  • 67. A method of detecting a biomarker in a head and neck tissue sample obtained from a patient, the method consisting of measuring the expression level of a plurality of biomarker nucleic acids selected from Table 1 or Table 3 using an amplification, hybridization and/or sequencing assay.
  • 68. The method of claim 67, wherein the head and neck tissue sample was previously diagnosed as being squamous cell carcinoma.
  • 69. The method of claim 68, wherein the previous diagnosis was by histological examination.
  • 70. The method of claim 67, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.
  • 71. The method of claim 70, wherein the expression level is detected by performing qRT-PCR.
  • 72. The method of claim 71, wherein the detection of the expression level comprises using at least one pair of oligonucleotide primers per each of the plurality of biomarker nucleic acids selected from Table 1 or Table 3.
  • 73. The method of claim 67, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.
  • 74. The method of claim 73, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 75. The method of claim 67, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.
  • 76. The method of claim 67, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 1.
  • 77. The method of claim 67, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.
  • 78. The method of claim 67, wherein the plurality of biomarker nucleic acids comprises, consists essentially of or consists of all the classifier biomarker nucleic acids of Table 3.
  • 79. The method of claim 67, wherein the method further comprises determining the HPV status of the patient.
  • 80. The method of claim 79, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 81. The method of claim 80, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
  • 82. A method of determining whether a HNSCC patient is likely to respond to immunotherapy, the method comprising, determining the HNSCC subtype of a head and neck tissue sample from the patient, wherein the HNSCC subtype is selected from the group consisting of basal, mesenchymal, atypical and classical; andbased on the subtype, assessing whether the patient is likely to respond to immunotherapy.
  • 83. A method for selecting a HNSCC patient for immunotherapy, the method comprising, determining a HNSCC subtype of a head and neck tissue sample from the patient, based on the subtype; and selecting the patient for immunotherapy.
  • 84. The method of claim 82 or 83, wherein the immunotherapy comprises checkpoint inhibitor therapy.
  • 85. The method of claim 84, wherein the checkpoint inhibitor targets PD-1 or PD-L1.
  • 86. The method of claim 84, wherein the checkpoint inhibitor targets CTLA-4.
  • 87. The method of claim 85, wherein the checkpoint inhibitor is Pembrolizumab, Nivolumab or an antigen fragment binding fragment thereof.
  • 88. The method of claim 86, wherein the checkpoint inhibitor is Ipilimumab or an antigen binding fragment thereof.
  • 89. The method of claim 82 or 83, wherein the patient is initially determined to have HNSCC via a histological analysis of a sample.
  • 90. The method of claim 82 or 83, wherein the patient's HNSCC molecular subtype is selected from basal, mesenchymal, atypical or classical and is determined via a histological analysis of a sample obtained from the patient.
  • 91. The method of claim 89, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, or a bodily fluid obtained from the patient.
  • 92. The method of claim 91, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 93. The method of claim 82 or 83, wherein the determining the HNSCC subtype comprises determining expression levels of a plurality of classifier biomarkers.
  • 94. The method of claim 93, wherein the determining the expression levels of the plurality of classifier biomarkers is at a nucleic acid level by performing RNA sequencing, reverse transcriptase polymerase chain reaction (RT-PCR) or hybridization based analyses.
  • 95. The method of claim 93, wherein the plurality of classifier biomarkers for determining the HNSCC subtype is selected from a publically available HNSCC dataset.
  • 96. The method of claim 95, wherein the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset.
  • 97. The method of claim 94, wherein the plurality of classifier biomarkers for determining the HNSCC subtype is selected from Table 1 or Table 3.
  • 98. The method of claim 97, wherein the RT-PCR is quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR).
  • 99. The method of claim 98, wherein the RT-PCR is performed with primers specific to the plurality of classifier biomarkers of Table 1 or Table 3.
  • 100. The method of claim 93, further comprising comparing the detected levels of expression of the plurality of classifier biomarkers of Table 1 or Table 3 to the expression of the plurality of classifier biomarkers of Table 1 or Table 3 in at least one sample training set(s), wherein the at least one sample training set comprises expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC BA sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC MS sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC AT sample, expression data of the plurality of classifier biomarkers of Table 1 or Table 3 from a reference HNSCC CL sample or a combination thereof; and classifying the first sample as BA, MS, AT or CL based on the results of the comparing step.
  • 101. The method of claim 100, wherein the comparing step comprises applying a statistical algorithm which comprises determining a correlation between the expression data obtained from the sample and the expression data from the at least one training set(s); and classifying the sample as a BA, MS, AT or CL subtype based on the results of the statistical algorithm.
  • 102. The method of claim 93, wherein the plurality of the classifier biomarkers comprise each of the classifier biomarkers set forth in Table 1 or Table 3.
  • 103. The method of claim 82 or 83, wherein the method further comprises determining the HPV status of the patient.
  • 104. The method of claim 103, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 105. The method of claim 104, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
  • 106. A method of treating HNSCC in a subject, the method comprising: measuring the expression level of at least one biomarker nucleic acid in a HNSCC sample obtained from the subject, wherein the at least one biomarker nucleic acid is selected from a set of biomarkers listed in Table 1 or Table 3, wherein the presence, absence and/or level of the at least one biomarker indicates a subtype of the HNSCC; andadministering an immunotherapeutic agent based on the subtype of the HNSCC.
  • 107. The method of claim 106, wherein the head and neck sample is a HNSCC sample.
  • 108. The method of claim 107, wherein the at least one biomarker nucleic acid selected from the set of biomarkers comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 18 biomarker nucleic acids, at least 36 biomarker nucleic acids, at least 54 biomarker nucleic acids, at least 72 biomarker nucleic acids, at least 90 biomarker nucleic acids, at least 108 biomarker nucleic acids, at least 126 biomarker nucleic acids, or at least 144 biomarker nucleic acids of Table 1.
  • 109. The method of claim 107, wherein the at least one biomarker nucleic acid selected from the set of biomarkers comprises, consists essentially of or consists of at least two biomarker nucleic acids, at least 10 biomarker nucleic acids, at least 20 biomarker nucleic acids, at least 30 biomarker nucleic acids, at least 40 biomarker nucleic acids, at least 50 biomarker nucleic acids, at least 60 biomarker nucleic acids, at least 70 biomarker nucleic acids, or at least 80 biomarker nucleic acids of Table 3.
  • 110. The method of claim 106, wherein the head and neck tissue sample was previously diagnosed as HNSCC.
  • 111. The method of claim 109, wherein the previous diagnosis was by histological examination.
  • 112. The method of any one of claims 106-110, further comprising measuring the expression of at least one biomarker from an additional set of biomarkers.
  • 113. The method of claim 112, wherein the additional set of biomarkers comprise gene expression signatures of Innate Immune Cells (IIC), Adaptive Immune Cells (AIC), one or more individual immune biomarkers, one or more interferon (IFN) genes, one or more major histocompatibility complex, class II (MHCII) genes or a combination thereof.
  • 114. The method of claim 113, wherein the additional set of biomarkers comprises genes selected from Tables 6A, 6B, 7, 8, 9, or a combination thereof.
  • 115. The method of claim 113, wherein the gene expression signatures of AICs are selected from Table 6A.
  • 116. The method of claim 113, wherein the gene expression signature of IICs are selected from Table 6B.
  • 117. The method of claim 113, wherein the one or more individual immune biomarkers are selected from Table 7.
  • 118. The method of claim 113, wherein the one or more IFN genes are selected from Table 8.
  • 119. The method of claim 113, wherein the one or more MHCII genes are selected from Table 9.
  • 120. The method of claim 106, wherein the measuring the expression level is conducted using an amplification, hybridization and/or sequencing assay.
  • 121. The method of claim 120, wherein the amplification, hybridization and/or sequencing assay comprises performing quantitative real time reverse transcriptase polymerase chain reaction (qRT-PCR), RNAseq, microarrays, gene chips, nCounter Gene Expression Assay, Serial Analysis of Gene Expression (SAGE), Rapid Analysis of Gene Expression (RAGE), nuclease protection assays, Northern blotting, or any other equivalent gene expression detection techniques.
  • 122. The method of claim 121, wherein the expression level is detected by performing qRT-PCR.
  • 123. The method of claim 106, wherein the sample is a formalin-fixed, paraffin-embedded (FFPE) head and neck tissue sample, fresh or a frozen tissue sample, an exosome, wash fluids, cell pellets, or a bodily fluid obtained from the patient.
  • 124. The method of claim 123, wherein the bodily fluid is blood or fractions thereof, urine, saliva, or sputum.
  • 125. The method of claim 106, wherein the subject's HNSCC subtype is selected from basal, mesenchymal, atypical or classical.
  • 126. The method of claim 106, wherein the at least one biomarker nucleic acid is a plurality of biomarker nucleic acids, wherein the plurality of biomarker nucleic acids comprises at least one biomarker nucleic acid listed in Table 1 or Table 3 in combination with one or more biomarker nucleic acids from a publically available HNSCC dataset, wherein the presence, absence and/or level of the plurality of biomarker nucleic acids indicates a subtype of the HNSCC.
  • 127. The method of claim 106, wherein the at least one biomarker nucleic acid is a plurality of biomarker nucleic acids, wherein the plurality of biomarker nucleic acids comprises all of the biomarker nucleic acids listed in Table 1 or Table 3 in combination with one or more biomarker nucleic acids from a publically available HNSCC dataset, wherein the presence, absence and/or level of the plurality of biomarker nucleic acids indicates a subtype of the HNSCC.
  • 128. The method of claim 126 or 127, wherein the publically available HNSCC dataset is TCGA HNSCC RNAseq dataset.
  • 129. The method of claim 106, wherein the method further comprises determining the HPV status of the patient.
  • 130. The method of claim 129, wherein the determining the HPV status of the patient comprises measuring the expression of one or more HPV genes in the tissue sample obtained from the patient.
  • 131. The method of claim 130, wherein the one or more HPV genes is the E6 gene, the E7 gene, the E6 and E7 genes or the E6 and E7 genes in combination with one or more additional genes from the HPV genome.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of priority to U.S. Provisional Application No. 62/541,960 filed Aug. 7, 2017, U.S. Provisional Application No. 62/608,218 filed Dec. 20, 2017, and U.S. Provisional Application No. 62/629,934 filed Feb. 13, 2018, each of which are incorporated by reference herein in their entireties for all purposes.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/045522 8/7/2018 WO 00
Provisional Applications (3)
Number Date Country
62629934 Feb 2018 US
62608218 Dec 2017 US
62541960 Aug 2017 US