COMPOSITIONS AND METHODS FOR ASSESSING IMMUNE RESPONSE

The present invention provides methods and compositions that are useful for assessing gene expression for tumor immune response profile of a sample. In particular, a target-specific primer panel is provided that allows for selective amplification of immune response target sequences in a sample. In one aspect, the invention relates to target-specific primers useful for selective amplification of one or more target sequences associated with immune response. In some aspects, amplified target sequences obtained using the disclosed methods, and compositions can be used in various processes including nucleic acid sequencing and used to detect the presence of genetic variants and/or expression levels of one or more targeted sequences associated with immune response.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

This application claims priority to and the benefit of U.S. Provisional Application No. 62/398,756 filed Sep. 23, 2016 and U.S. Provisional Application No. 62/419,091 filed Nov. 8, 2016. The entire contents of each of the aforementioned applications are incorporated herein by reference.

SEQUENCE LISTING

This application hereby incorporates by reference the material of the electronic Sequence Listing filed concurrently herewith. The material in the electronic Sequence Listing is submitted as a text (.txt) file entitled “LT01193_ST25.txt” created on Sep. 22, 2017, and is herein incorporated by reference in its entirety.

BACKGROUND

Advances in cancer immunotherapies have started to provide promising results across oncology. Immune checkpoint inhibitors, cancer vaccines and T-cell therapies have shown sustainable results in responsive populations over conventional or targeted therapies. However, effective identification of responsive candidates and/or monitoring response has proven challenging. The need of a better understanding of the tumor microenvironment, tumor-lymphocyte interactions and drug response biomarkers is immediate. In particular, for example, while the presence of PD-L1 has been reported to be a promising marker to predict positive response to anti-PD-L1 therapy, current methods using immunohistochemistry to measure PD-L1 protein levels are inefficient and highly variable. Higher-throughput, systematic and standardized solutions are a more desirable alternative.

BRIEF SUMMARY OF THE INVENTION

In one aspect of the invention compositions are provided for a single stream multiplex determination of an immune response in a sample. In some embodiments the composition consists of a plurality of primer pair reagents directed to a plurality of target sequences to measure the expression levels of the targets in the sample. Provided compositions target immune response genes and housekeeping gene sequences wherein the plurality of immune response target gene sequences are selected from targets among the following function: checkpoint pathways, T cell related signaling pathways, markers of tumor infiltrating lymphocytes (TILS), tumor markers, and housekeeping genes. In some embodiments the plurality of immune response target genes are selected from targets among immune checkpoint pathways and targets; T and B cell signaling genes, markers of lymphocyte subsets, interferon signaling genes, cytokine signaling genes; tumor markers, tumor antigens, and proliferation markers. In particular embodiments, provided compositions include a plurality of primer pair reagents selected from Table 2. In some embodiments a multiplex assay comprising compositions of the invention is provided. In some embodiments a test kit comprising compositions of the invention is provided.

In another aspect of the invention, methods are provided for determining immune response activity in a biological sample. Such methods comprise performing multiplex amplification of a plurality of target expression sequences from a biological sample containing target sequences. Amplification comprises contacting at least a portion of the sample comprising multiple target sequences of interest using a plurality of target-specific primer pairs in the presence of a polymerase under amplification conditions to produce a plurality of amplified target expression sequences. The methods further comprise detecting the level of expression of each of the plurality of target immune response sequences, wherein a change in the level of expression of one or more target immune response markers as compared with a control sample determines a change in immune response activity in the sample. The methods described herein utilize compositions of the invention provided herein.

BRIEF DESCRIPTION OF THE DRAWINGS

The accompanying drawings, which are incorporated into and form a part of the specification, illustrate one or more exemplary embodiments and serve to explain the principles of various exemplary embodiments. The drawings are exemplary and explanatory only and are not to be construed as limiting or restrictive in any way.

FIG. 1 depicts exemplary repeatibility results of immune response research assay. Immune responses assay produced correlation greater than 0.98 between replicates of FFPE research samples of NSCLC, melanoma, and normal liver tissue.

FIG. 2A-2C depicts exemplary result of immune response assay to determine limits of detection and dynamic range: FIG. 2A depicts ratios of samples of HL-60 and human lung total RNA measured individually and mixed at 4 different ratios to evaluate dynamic range; FIG. 2B depicts exemplary results of correlation of fold changes between pure and mixed samples; and FIG. 2C depicts exemplary results of correlations among rank order of fold change between pure and mixed samples.

FIG. 3 depicts exemplary results of correlations between quantitative RT-PCR detection and the immune response NGS assay of the invention. Twenty two genes were selected and tested by quantitative RT-PCR in NSCLC and melanoma FFPE samples. Results demonstrated correlations between quantitative RT-PCR and NGS are 0.85 to 0.95.

FIG. 4 depicts exemplary results of correlation among fresh frozen and FFPE samples. NSCLC fresh frozen and FFPE samples showed greater than 0.96 correlation.

FIG. 5A-5D depicts exemplary results of the immune response assay. FIG. 5A is gene expression distribution plot; FIG. 5B is a bar graph depicting housekeeping gene expression; FIG. 5C is a sample correlation heatmap; and FIG. 5D depicts results of two principle component analysis for samples tested.

FIG. 6 depicts exemplary results of sample correlation of the immune response assay.

FIG. 7 depicts exemplary heatmap results of all targets of the immune response assay.

FIG. 8 depicts results of a simulated CPM normalization applied to measure the expression of genes and calculate fold change. Using endogenous controls accurately estimates fold change, because while most genes do not change between samples, fold change estimate is negligible error; however, when many genes do change, fold change estimate is wildly inaccurate.

FIG. 9A-B depicts results suggesting housekeeping gene control allow for better expression estimates. FIG. 9A depicts results of correlation of estimated and true expression among various genes, including an assay made of only the 5 genes associated with the leukocyte inhibition category. Assuming only interest in a leukocyte inhibition assay, and using just those genes, we do not get a high correlation with true expression; adding leukocyte migration only helps mildly (21 genes); adding stemness and tumor makers (uncorrelated categories; 27 genes) increases the correlation. However, using only targets and 11 housekeeping (HK) genes works just as well as including non-informative genes and true expression. FIG. 9B depicts results comparing performance of HK normalization, CPM and CPM without housekeeping, demonstrating housekeeping normalization increases correlation with true expression as expression decreases.

DESCRIPTION OF THE INVENTION

The following description of various exemplary embodiments is exemplary and explanatory only and is not to be construed as limiting or restrictive in any way. Other embodiments, features, objects, and advantages of the present teachings will be apparent from the description and accompanying drawings, and from the claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art to which these inventions belong.

As used herein, the terms “comprises,” “comprising,” “includes,” “including,” “has,” “having” or any other variation thereof, are intended to cover a non-exclusive inclusion. For example, a process, method, article, or apparatus that comprises a list of features is not necessarily limited only to those features but may include other features not expressly listed or inherent to such process, method, article, or apparatus. Further, unless expressly stated to the contrary, “or” refers to an inclusive-or and not to an exclusive-or.

As used herein, “amplify”, “amplifying” or “amplification reaction” and their derivatives, refer to any action or process whereby at least a portion of a nucleic acid molecule (referred to as a template nucleic acid molecule) is replicated or copied into at least one additional nucleic acid molecule. The additional nucleic acid molecule optionally includes sequence that is substantially identical or substantially complementary to at least some portion of the template nucleic acid molecule. The template nucleic acid molecule can be single-stranded or double-stranded and the additional nucleic acid molecule can independently be single-stranded or double-stranded. In some embodiments, amplification includes a template-dependent in vitro enzyme-catalyzed reaction for the production of at least one copy of at least some portion of the nucleic acid molecule or the production of at least one copy of a nucleic acid sequence that is complementary to at least some portion of the nucleic acid molecule. Amplification optionally includes linear or exponential replication of a nucleic acid molecule. In some embodiments, such amplification is performed using isothermal conditions; in other embodiments, such amplification can include thermocycling. In some embodiments, the amplification is a multiplex amplification that includes the simultaneous amplification of a plurality of target sequences in a single amplification reaction. At least some of the target sequences can be situated on the same nucleic acid molecule or on different target nucleic acid molecules included in the single amplification reaction. In some embodiments, “amplification” includes amplification of at least some portion of DNA- and RNA-based nucleic acids alone, or in combination. The amplification reaction can include single or double-stranded nucleic acid substrates and can further including any of the amplification processes known to one of ordinary skill in the art. In some embodiments, the amplification reaction includes polymerase chain reaction (PCR).

As used herein, “amplification conditions” and its derivatives, refers to conditions suitable for amplifying one or more nucleic acid sequences. Such amplification can be linear or exponential. In some embodiments, the amplification conditions can include isothermal conditions or alternatively can include thermocyling conditions, or a combination of isothermal and thermocycling conditions. In some embodiments, the conditions suitable for amplifying one or more nucleic acid sequences includes polymerase chain reaction (PCR) conditions. Typically, the amplification conditions refer to a reaction mixture that is sufficient to amplify nucleic acids such as one or more target sequences, or to amplify an amplified target sequence ligated to one or more adapters, e.g., an adapter-ligated amplified target sequence. Amplification conditions include a catalyst for amplification or for nucleic acid synthesis, for example a polymerase; a primer that possesses some degree of complementarity to the nucleic acid to be amplified; and nucleotides, such as deoxyribonucleotide triphosphates (dNTPs) to promote extension of the primer once hybridized to the nucleic acid. The amplification conditions can require hybridization or annealing of a primer to a nucleic acid, extension of the primer and a denaturing step in which the extended primer is separated from the nucleic acid sequence undergoing amplification. Typically, but not necessarily, amplification conditions can include thermocycling; in some embodiments, amplification conditions include a plurality of cycles where the steps of annealing, extending and separating are repeated. Typically, the amplification conditions include cations such as Mg++ or Mn++ (e.g., MgCl2, etc) and can also include various modifiers of ionic strength.

As used herein, “target sequence” or “target sequence of interest” and its derivatives, refers to any single or double-stranded nucleic acid sequence that can be amplified or synthesized according to the disclosure, including any nucleic acid sequence suspected or expected to be present in a sample. In some embodiments, the target sequence is present in double-stranded form and includes at least a portion of the particular nucleotide sequence to be amplified or synthesized, or its complement, prior to the addition of target-specific primers or appended adapters. Target sequences can include the nucleic acids to which primers useful in the amplification or synthesis reaction can hybridize prior to extension by a polymerase. In some embodiments, the term refers to a nucleic acid sequence whose sequence identity, ordering or location of nucleotides is determined by one or more of the methods of the disclosure.

As defined herein, “sample” and its derivatives, is used in its broadest sense and includes any specimen, culture and the like that is suspected of including a target. In some embodiments, the sample comprises cDNA, RNA, PNA, LNA, chimeric, hybrid, or multiplex-forms of nucleic acids. The sample can include any biological, clinical, surgical, agricultural, atmospheric or aquatic-based specimen containing one or more nucleic acids. The term also includes any isolated nucleic acid sample such a expressed RNA, fresh-frozen or formalin-fixed paraffin-embedded nucleic acid specimen.

As used herein, “contacting” and its derivatives, when used in reference to two or more components, refers to any process whereby the approach, proximity, mixture or commingling of the referenced components is promoted or achieved without necessarily requiring physical contact of such components, and includes mixing of solutions containing any one or more of the referenced components with each other. The referenced components may be contacted in any particular order or combination and the particular order of recitation of components is not limiting. For example, “contacting A with B and C” encompasses embodiments where A is first contacted with B then C, as well as embodiments where C is contacted with A then B, as well as embodiments where a mixture of A and C is contacted with B, and the like. Furthermore, such contacting does not necessarily require that the end result of the contacting process be a mixture including all of the referenced components, as long as at some point during the contacting process all of the referenced components are simultaneously present or simultaneously included in the same mixture or solution. Where one or more of the referenced components to be contacted includes a plurality (e.g., “contacting a target sequence with a plurality of target-specific primers and a polymerase”), then each member of the plurality can be viewed as an individual component of the contacting process, such that the contacting can include contacting of any one or more members of the plurality with any other member of the plurality and/or with any other referenced component (e.g., some but not all of the plurality of target specific primers can be contacted with a target sequence, then a polymerase, and then with other members of the plurality of target-specific primers) in any order or combination.

As used herein, the term “primer” and its derivatives refer to any polynucleotide that can hybridize to a target sequence of interest. In some embodiments, the primer can also serve to prime nucleic acid synthesis. Typically, the primer functions as a substrate onto which nucleotides can be polymerized by a polymerase; in some embodiments, however, the primer can become incorporated into the synthesized nucleic acid strand and provide a site to which another primer can hybridize to prime synthesis of a new strand that is complementary to the synthesized nucleic acid molecule. The primer may be comprised of any combination of nucleotides or analogs thereof, which may be optionally linked to form a linear polymer of any suitable length. In some embodiments, the primer is a single-stranded oligonucleotide or polynucleotide. (For purposes of this disclosure, the terms ‘polynucleotide” and “oligonucleotide” are used interchangeably herein and do not necessarily indicate any difference in length between the two). In some embodiments, the primer is single-stranded but it can also be double-stranded. The primer optionally occurs naturally, as in a purified restriction digest, or can be produced synthetically. In some embodiments, the primer acts as a point of initiation for amplification or synthesis when exposed to amplification or synthesis conditions; such amplification or synthesis can occur in a template-dependent fashion and optionally results in formation of a primer extension product that is complementary to at least a portion of the target sequence. Exemplary amplification or synthesis conditions can include contacting the primer with a polynucleotide template (e.g., a template including a target sequence), nucleotides and an inducing agent such as a polymerase at a suitable temperature and pH to induce polymerization of nucleotides onto an end of the target-specific primer. If double-stranded, the primer can optionally be treated to separate its strands before being used to prepare primer extension products. In some embodiments, the primer is an oligodeoxyribonucleotide or an oligoribonucleotide. In some embodiments, the primer can include one or more nucleotide analogs. The exact length and/or composition, including sequence, of the target-specific primer can influence many properties, including melting temperature (Tm), GC content, formation of secondary structures, repeat nucleotide motifs, length of predicted primer extension products, extent of coverage across a nucleic acid molecule of interest, number of primers present in a single amplification or synthesis reaction, presence of nucleotide analogs or modified nucleotides within the primers, and the like. In some embodiments, a primer can be paired with a compatible primer within an amplification or synthesis reaction to form a primer pair consisting or a forward primer and a reverse primer. In some embodiments, the forward primer of the primer pair includes a sequence that is substantially complementary to at least a portion of a strand of a nucleic acid molecule, and the reverse primer of the primer of the primer pair includes a sequence that is substantially identical to at least of portion of the strand. In some embodiments, the forward primer and the reverse primer are capable of hybridizing to opposite strands of a nucleic acid duplex. Optionally, the forward primer primes synthesis of a first nucleic acid strand, and the reverse primer primes synthesis of a second nucleic acid strand, wherein the first and second strands are substantially complementary to each other, or can hybridize to form a double-stranded nucleic acid molecule. In some embodiments, one end of an amplification or synthesis product is defined by the forward primer and the other end of the amplification or synthesis product is defined by the reverse primer. In some embodiments, where the amplification or synthesis of lengthy primer extension products is required, such as amplifying an exon, coding region, or gene, several primer pairs can be created than span the desired length to enable sufficient amplification of the region. In some embodiments, a primer can include one or more cleavable groups. In some embodiments, primer lengths are in the range of about 10 to about 60 nucleotides, about 12 to about 50 nucleotides and about 15 to about 40 nucleotides in length. Typically, a primer is capable of hybridizing to a corresponding target sequence and undergoing primer extension when exposed to amplification conditions in the presence of dNTPS and a polymerase. In some instances, the particular nucleotide sequence or a portion of the primer is known at the outset of the amplification reaction or can be determined by one or more of the methods disclosed herein. In some embodiments, the primer includes one or more cleavable groups at one or more locations within the primer.

As used herein, “target-specific primer” and its derivatives, refers to a single stranded or double-stranded polynucleotide, typically an oligonucleotide, that includes at least one sequence that is at least 50% complementary, typically at least 75% complementary or at least 85% complementary, more typically at least 90% complementary, more typically at least 95% complementary, more typically at least 98% or at least 99% complementary, or identical, to at least a portion of a nucleic acid molecule that includes a target sequence. In such instances, the target-specific primer and target sequence are described as “corresponding” to each other. In some embodiments, the target-specific primer is capable of hybridizing to at least a portion of its corresponding target sequence (or to a complement of the target sequence); such hybridization can optionally be performed under standard hybridization conditions or under stringent hybridization conditions. In some embodiments, the target-specific primer is not capable of hybridizing to the target sequence, or to its complement, but is capable of hybridizing to a portion of a nucleic acid strand including the target sequence, or to its complement. In some embodiments, the target-specific primer includes at least one sequence that is at least 75% complementary, typically at least 85% complementary, more typically at least 90% complementary, more typically at least 95% complementary, more typically at least 98% complementary, or more typically at least 99% complementary, to at least a portion of the target sequence itself; in other embodiments, the target-specific primer includes at least one sequence that is at least 75% complementary, typically at least 85% complementary, more typically at least 90% complementary, more typically at least 95% complementary, more typically at least 98% complementary, or more typically at least 99% complementary, to at least a portion of the nucleic acid molecule other than the target sequence. In some embodiments, the target-specific primer is substantially non-complementary to other target sequences present in the sample; optionally, the target-specific primer is substantially non-complementary to other nucleic acid molecules present in the sample. In some embodiments, nucleic acid molecules present in the sample that do not include or correspond to a target sequence (or to a complement of the target sequence) are referred to as “non-specific” sequences or “non-specific nucleic acids”. In some embodiments, the target-specific primer is designed to include a nucleotide sequence that is substantially complementary to at least a portion of its corresponding target sequence. In some embodiments, a target-specific primer is at least 95% complementary, or at least 99% complementary, or identical, across its entire length to at least a portion of a nucleic acid molecule that includes its corresponding target sequence. In some embodiments, a target-specific primer can be at least 90%, at least 95% complementary, at least 98% complementary or at least 99% complementary, or identical, across its entire length to at least a portion of its corresponding target sequence. In some embodiments, a forward target-specific primer and a reverse target-specific primer define a target-specific primer pair that can be used to amplify the target sequence via template-dependent primer extension. Typically, each primer of a target-specific primer pair includes at least one sequence that is substantially complementary to at least a portion of a nucleic acid molecule including a corresponding target sequence but that is less than 50% complementary to at least one other target sequence in the sample. In some embodiments, amplification can be performed using multiple target-specific primer pairs in a single amplification reaction, wherein each primer pair includes a forward target-specific primer and a reverse target-specific primer, each including at least one sequence that substantially complementary or substantially identical to a corresponding target sequence in the sample, and each primer pair having a different corresponding target sequence. In some embodiments, the target-specific primer can be substantially non-complementary at its 3′ end or its 5′ end to any other target-specific primer present in an amplification reaction. In some embodiments, the target-specific primer can include minimal cross hybridization to other target-specific primers in the amplification reaction. In some embodiments, target-specific primers include minimal cross-hybridization to non-specific sequences in the amplification reaction mixture. In some embodiments, the target-specific primers include minimal self-complementarity. In some embodiments, the target-specific primers can include one or more cleavable groups located at the 3′ end. In some embodiments, the target-specific primers can include one or more cleavable groups located near or about a central nucleotide of the target-specific primer. In some embodiments, one of more targets-specific primers includes only non-cleavable nucleotides at the 5′ end of the target-specific primer. In some embodiments, a target specific primer includes minimal nucleotide sequence overlap at the 3′end or the 5′ end of the primer as compared to one or more different target-specific primers, optionally in the same amplification reaction. In some embodiments 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more, target-specific primers in a single reaction mixture include one or more of the above embodiments. In some embodiments, substantially all of the plurality of target-specific primers in a single reaction mixture includes one or more of the above embodiments.

As used herein, “polymerase” and its derivatives, refers to any enzyme that can catalyze the polymerization of nucleotides (including analogs thereof) into a nucleic acid strand. Typically but not necessarily, such nucleotide polymerization can occur in a template-dependent fashion. Such polymerases can include without limitation naturally occurring polymerases and any subunits and truncations thereof, mutant polymerases, variant polymerases, recombinant, fusion or otherwise engineered polymerases, chemically modified polymerases, synthetic molecules or assemblies, and any analogs, derivatives or fragments thereof that retain the ability to catalyze such polymerization. Optionally, the polymerase can be a mutant polymerase comprising one or more mutations involving the replacement of one or more amino acids with other amino acids, the insertion or deletion of one or more amino acids from the polymerase, or the linkage of parts of two or more polymerases. Typically, the polymerase comprises one or more active sites at which nucleotide binding and/or catalysis of nucleotide polymerization can occur. Some exemplary polymerases include without limitation DNA polymerases and RNA polymerases. The term “polymerase” and its variants, as used herein, also refers to fusion proteins comprising at least two portions linked to each other, where the first portion comprises a peptide that can catalyze the polymerization of nucleotides into a nucleic acid strand and is linked to a second portion that comprises a second polypeptide. In some embodiments, the second polypeptide can include a reporter enzyme or a processivity-enhancing domain. Optionally, the polymerase can possess 5′ exonuclease activity or terminal transferase activity. In some embodiments, the polymerase can be optionally reactivated, for example through the use of heat, chemicals or re-addition of new amounts of polymerase into a reaction mixture. In some embodiments, the polymerase can include a hot-start polymerase or an aptamer based polymerase that optionally can be reactivated.

As used herein, the term “nucleotide” and its variants comprises any compound, including without limitation any naturally occurring nucleotide or analog thereof, which can bind selectively to, or can be polymerized by, a polymerase. Typically, but not necessarily, selective binding of the nucleotide to the polymerase is followed by polymerization of the nucleotide into a nucleic acid strand by the polymerase; occasionally however the nucleotide may dissociate from the polymerase without becoming incorporated into the nucleic acid strand, an event referred to herein as a “non-productive” event. Such nucleotides include not only naturally occurring nucleotides but also any analogs, regardless of their structure, that can bind selectively to, or can be polymerized by, a polymerase. While naturally occurring nucleotides typically comprise base, sugar and phosphate moieties, the nucleotides of the present disclosure can include compounds lacking any one, some or all of such moieties. In some embodiments, the nucleotide can optionally include a chain of phosphorus atoms comprising three, four, five, six, seven, eight, nine, ten or more phosphorus atoms. In some embodiments, the phosphorus chain can be attached to any carbon of a sugar ring, such as the 5′ carbon. The phosphorus chain can be linked to the sugar with an intervening O or S. In one embodiment, one or more phosphorus atoms in the chain can be part of a phosphate group having P and O. In another embodiment, the phosphorus atoms in the chain can be linked together with intervening O, NH, S, methylene, substituted methylene, ethylene, substituted ethylene, CNH2, C(O), C(CH2), CH2CH2, or C(OH)CH2R (where R can be a 4-pyridine or 1-imidazole). In one embodiment, the phosphorus atoms in the chain can have side groups having O, BH3, or S. In the phosphorus chain, a phosphorus atom with a side group other than O can be a substituted phosphate group. In the phosphorus chain, phosphorus atoms with an intervening atom other than O can be a substituted phosphate group. Some examples of nucleotide analogs are described in Xu, U.S. Pat. No. 7,405,281. In some embodiments, the nucleotide comprises a label and referred to herein as a “labeled nucleotide”; the label of the labeled nucleotide is referred to herein as a “nucleotide label”. In some embodiments, the label can be in the form of a fluorescent dye attached to the terminal phosphate group, i.e., the phosphate group most distal from the sugar. Some examples of nucleotides that can be used in the disclosed methods and compositions include, but are not limited to, ribonucleotides, deoxyribonucleotides, modified ribonucleotides, modified deoxyribonucleotides, ribonucleotide polyphosphates, deoxyribonucleotide polyphosphates, modified ribonucleotide polyphosphates, modified deoxyribonucleotide polyphosphates, peptide nucleotides, modified peptide nucleotides, metallonucleosides, phosphonate nucleosides, and modified phosphate-sugar backbone nucleotides, analogs, derivatives, or variants of the foregoing compounds, and the like. In some embodiments, the nucleotide can comprise non-oxygen moieties such as, for example, thio- or borano-moieties, in place of the oxygen moiety bridging the alpha phosphate and the sugar of the nucleotide, or the alpha and beta phosphates of the nucleotide, or the beta and gamma phosphates of the nucleotide, or between any other two phosphates of the nucleotide, or any combination thereof. “Nucleotide 5′-triphosphate” refers to a nucleotide with a triphosphate ester group at the 5′ position, and are sometimes denoted as “NTP”, or “dNTP” and “ddNTP” to particularly point out the structural features of the ribose sugar. The triphosphate ester group can include sulfur substitutions for the various oxygens, e.g. .alpha.-thio-nucleotide 5′-triphosphates. For a review of nucleic acid chemistry, see: Shabarova, Z. and Bogdanov, A. Advanced Organic Chemistry of Nucleic Acids, VCH, New York, 1994.

The term “extension” and its variants, as used herein, when used in reference to a given primer, comprises any in vivo or in vitro enzymatic activity characteristic of a given polymerase that relates to polymerization of one or more nucleotides onto an end of an existing nucleic acid molecule. Typically but not necessarily such primer extension occurs in a template-dependent fashion; during template-dependent extension, the order and selection of bases is driven by established base pairing rules, which can include Watson-Crick type base pairing rules or alternatively (and especially in the case of extension reactions involving nucleotide analogs) by some other type of base pairing paradigm. In one non-limiting example, extension occurs via polymerization of nucleotides on the 3′OH end of the nucleic acid molecule by the polymerase.

The term “portion” and its variants, as used herein, when used in reference to a given nucleic acid molecule, for example a primer or a template nucleic acid molecule, comprises any number of contiguous nucleotides within the length of the nucleic acid molecule, including the partial or entire length of the nucleic acid molecule.

The terms “identity” and “identical” and their variants, as used herein, when used in reference to two or more nucleic acid sequences, refer to similarity in sequence of the two or more sequences (e.g., nucleotide or polypeptide sequences). In the context of two or more homologous sequences, the percent identity or homology of the sequences or subsequences thereof indicates the percentage of all monomeric units (e.g., nucleotides or amino acids) that are the same (i.e., about 70% identity, preferably 75%, 80%, 85%, 90%, 95%, 98% or 99% identity). The percent identity can be over a specified region, when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using a BLAST or BLAST 2.0 sequence comparison algorithms with default parameters described below, or by manual alignment and visual inspection. Sequences are said to be “substantially identical” when there is at least 85% identity at the amino acid level or at the nucleotide level. Preferably, the identity exists over a region that is at least about 25, 50, or 100 residues in length, or across the entire length of at least one compared sequence. A typical algorithm for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al, Nuc. Acids Res. 25:3389-3402 (1977). Other methods include the algorithms of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), and Needleman & Wunsch, J. Mol. Biol. 48:443 (1970), etc. Another indication that two nucleic acid sequences are substantially identical is that the two molecules or their complements hybridize to each other under stringent hybridization conditions.

The terms “complementary” and “complement” and their variants, as used herein, refer to any two or more nucleic acid sequences (e.g., portions or entireties of template nucleic acid molecules, target sequences and/or primers) that can undergo cumulative base pairing at two or more individual corresponding positions in antiparallel orientation, as in a hybridized duplex. Such base pairing can proceed according to any set of established rules, for example according to Watson-Crick base pairing rules or according to some other base pairing paradigm. Optionally there can be “complete” or “total” complementarity between a first and second nucleic acid sequence where each nucleotide in the first nucleic acid sequence can undergo a stabilizing base pairing interaction with a nucleotide in the corresponding antiparallel position on the second nucleic acid sequence. “Partial” complementarity describes nucleic acid sequences in which at least 20%, but less than 100%, of the residues of one nucleic acid sequence are complementary to residues in the other nucleic acid sequence. In some embodiments, at least 50%, but less than 100%, of the residues of one nucleic acid sequence are complementary to residues in the other nucleic acid sequence. In some embodiments, at least 70%, 80%, 90%, 95% or 98%, but less than 100%, of the residues of one nucleic acid sequence are complementary to residues in the other nucleic acid sequence. Sequences are said to be “substantially complementary” when at least 85% of the residues of one nucleic acid sequence are complementary to residues in the other nucleic acid sequence. In some embodiments, two complementary or substantially complementary sequences are capable of hybridizing to each other under standard or stringent hybridization conditions. “Non-complementary” describes nucleic acid sequences in which less than 20% of the residues of one nucleic acid sequence are complementary to residues in the other nucleic acid sequence. Sequences are said to be “substantially non-complementary” when less than 15% of the residues of one nucleic acid sequence are complementary to residues in the other nucleic acid sequence. In some embodiments, two non-complementary or substantially non-complementary sequences cannot hybridize to each other under standard or stringent hybridization conditions. A “mismatch” is present at any position in the two opposed nucleotides are not complementary. Complementary nucleotides include nucleotides that are efficiently incorporated by DNA polymerases opposite each other during DNA replication under physiological conditions. In a typical embodiment, complementary nucleotides can form base pairs with each other, such as the A-T/U and G-C base pairs formed through specific Watson-Crick type hydrogen bonding, or base pairs formed through some other type of base pairing paradigm, between the nucleobases of nucleotides and/or polynucleotides in positions antiparallel to each other. The complementarity of other artificial base pairs can be based on other types of hydrogen bonding and/or hydrophobicity of bases and/or shape complementarity between bases.

As used herein, “amplified target sequences” and its derivatives, refers to a nucleic acid sequence produced by the amplification of/amplifying the target sequences using target-specific primers and the methods provided herein. The amplified target sequences may be either of the same sense (the positive strand produced in the second round and subsequent even-numbered rounds of amplification) or antisense (i.e., the negative strand produced during the first and subsequent odd-numbered rounds of amplification) with respect to the target sequences. For the purposes of this disclosure, the amplified target sequences are typically less than 50% complementary to any portion of another amplified target sequence in the reaction.

As used herein, the terms “ligating”, “ligation” and their derivatives refer to the act or process for covalently linking two or more molecules together, for example, covalently linking two or more nucleic acid molecules to each other. In some embodiments, ligation includes joining nicks between adjacent nucleotides of nucleic acids. In some embodiments, ligation includes forming a covalent bond between an end of a first and an end of a second nucleic acid molecule. In some embodiments, for example embodiments wherein the nucleic acid molecules to be ligated include conventional nucleotide residues, the litigation can include forming a covalent bond between a 5′ phosphate group of one nucleic acid and a 3′ hydroxyl group of a second nucleic acid thereby forming a ligated nucleic acid molecule. In some embodiments, any means for joining nicks or bonding a 5′phosphate to a 3′ hydroxyl between adjacent nucleotides can be employed. In an exemplary embodiment, an enzyme such as a ligase can be used. For the purposes of this disclosure, an amplified target sequence can be ligated to an adapter to generate an adapter-ligated amplified target sequence.

As used herein, “ligase” and its derivatives, refers to any agent capable of catalyzing the ligation of two substrate molecules. In some embodiments, the ligase includes an enzyme capable of catalyzing the joining of nicks between adjacent nucleotides of a nucleic acid. In some embodiments, the ligase includes an enzyme capable of catalyzing the formation of a covalent bond between a 5′ phosphate of one nucleic acid molecule to a 3′ hydroxyl of another nucleic acid molecule thereby forming a ligated nucleic acid molecule. Suitable ligases may include, but not limited to, T4 DNA ligase, T4 RNA ligase, and E. coli DNA ligase.

As used herein, “ligation conditions” and its derivatives, refers to conditions suitable for ligating two molecules to each other. In some embodiments, the ligation conditions are suitable for sealing nicks or gaps between nucleic acids. As defined herein, a “nick” or “gap” refers to a nucleic acid molecule that lacks a directly bound 5′ phosphate of a mononucleotide pentose ring to a 3′ hydroxyl of a neighboring mononucleotide pentose ring within internal nucleotides of a nucleic acid sequence. As used herein, the term nick or gap is consistent with the use of the term in the art. Typically, a nick or gap can be ligated in the presence of an enzyme, such as ligase at an appropriate temperature and pH. In some embodiments, T4 DNA ligase can join a nick between nucleic acids at a temperature of about 70-72° C.

As used herein, “blunt-end ligation” and its derivatives, refers to ligation of two blunt-end double-stranded nucleic acid molecules to each other. A “blunt end” refers to an end of a double-stranded nucleic acid molecule wherein substantially all of the nucleotides in the end of one strand of the nucleic acid molecule are base paired with opposing nucleotides in the other strand of the same nucleic acid molecule. A nucleic acid molecule is not blunt ended if it has an end that includes a single-stranded portion greater than two nucleotides in length, referred to herein as an “overhang”. In some embodiments, the end of nucleic acid molecule does not include any single stranded portion, such that every nucleotide in one strand of the end is based paired with opposing nucleotides in the other strand of the same nucleic acid molecule. In some embodiments, the ends of the two blunt ended nucleic acid molecules that become ligated to each other do not include any overlapping, shared or complementary sequence. Typically, blunted-end ligation excludes the use of additional oligonucleotide adapters to assist in the ligation of the double-stranded amplified target sequence to the double-stranded adapter, such as patch oligonucleotides as described in Mitra and Varley, US2010/0129874, published May 27, 2010. In some embodiments, blunt-ended ligation includes a nick translation reaction to seal a nick created during the ligation process.

As used herein, the terms “adapter” or “adapter and its complements” and their derivatives, refers to any linear oligonucleotide which can be ligated to a nucleic acid molecule of the disclosure. Optionally, the adapter includes a nucleic acid sequence that is not substantially complementary to the 3′ end or the 5′ end of at least one target sequences within the sample. In some embodiments, the adapter is substantially non-complementary to the 3′ end or the 5′ end of any target sequence present in the sample. In some embodiments, the adapter includes any single stranded or double-stranded linear oligonucleotide that is not substantially complementary to an amplified target sequence. In some embodiments, the adapter is substantially non-complementary to at least one, some or all of the nucleic acid molecules of the sample. In some embodiments, suitable adapter lengths are in the range of about 10-100 nucleotides, about 12-60 nucleotides and about 15-50 nucleotides in length. An adapter can include any combination of nucleotides and/or nucleic acids. In some aspects, the adapter can include one or more cleavable groups at one or more locations. In another aspect, the adapter can include a sequence that is substantially identical, or substantially complementary, to at least a portion of a primer, for example a universal primer. In some embodiments, the adapter can include a barcode or tag to assist with downstream cataloguing, identification or sequencing. In some embodiments, a single-stranded adapter can act as a substrate for amplification when ligated to an amplified target sequence, particularly in the presence of a polymerase and dNTPs under suitable temperature and pH.

As used herein, “reamplifying” or “reamplification” and their derivatives refer to any process whereby at least a portion of an amplified nucleic acid molecule is further amplified via any suitable amplification process (referred to in some embodiments as a “secondary” amplification or “reamplification”, thereby producing a reamplified nucleic acid molecule. The secondary amplification need not be identical to the original amplification process whereby the amplified nucleic acid molecule was produced; nor need the reamplified nucleic acid molecule be completely identical or completely complementary to the amplified nucleic acid molecule; all that is required is that the reamplified nucleic acid molecule include at least a portion of the amplified nucleic acid molecule or its complement. For example, the reamplification can involve the use of different amplification conditions and/or different primers, including different target-specific primers than the primary amplification.

As defined herein, a “cleavable group” refers to any moiety that once incorporated into a nucleic acid can be cleaved under appropriate conditions. For example, a cleavable group can be incorporated into a target-specific primer, an amplified sequence, an adapter or a nucleic acid molecule of the sample. In an exemplary embodiment, a target-specific primer can include a cleavable group that becomes incorporated into the amplified product and is subsequently cleaved after amplification, thereby removing a portion, or all, of the target-specific primer from the amplified product. The cleavable group can be cleaved or otherwise removed from a target-specific primer, an amplified sequence, an adapter or a nucleic acid molecule of the sample by any acceptable means. For example, a cleavable group can be removed from a target-specific primer, an amplified sequence, an adapter or a nucleic acid molecule of the sample by enzymatic, thermal, photo-oxidative or chemical treatment. In one aspect, a cleavable group can include a nucleobase that is not naturally occurring. For example, an oligodeoxyribonucleotide can include one or more RNA nucleobases, such as uracil that can be removed by a uracil glycosylase. In some embodiments, a cleavable group can include one or more modified nucleobases (such as 7-methylguanine, 8-oxo-guanine, xanthine, hypoxanthine, 5,6-dihydrouracil or 5-methylcytosine) or one or more modified nucleosides (i.e., 7-methylguanosine, 8-oxo-deoxyguanosine, xanthosine, inosine, dihydrouridine or 5-methylcytidine). The modified nucleobases or nucleotides can be removed from the nucleic acid by enzymatic, chemical or thermal means. In one embodiment, a cleavable group can include a moiety that can be removed from a primer after amplification (or synthesis) upon exposure to ultraviolet light (i.e., bromodeoxyuridine). In another embodiment, a cleavable group can include methylated cytosine. Typically, methylated cytosine can be cleaved from a primer for example, after induction of amplification (or synthesis), upon sodium bisulfite treatment. In some embodiments, a cleavable moiety can include a restriction site. For example, a primer or target sequence can include a nucleic acid sequence that is specific to one or more restriction enzymes, and following amplification (or synthesis), the primer or target sequence can be treated with the one or more restriction enzymes such that the cleavable group is removed. Typically, one or more cleavable groups can be included at one or more locations with a target-specific primer, an amplified sequence, an adapter or a nucleic acid molecule of the sample.

As used herein, “cleavage step” and its derivatives, refers to any process by which a cleavable group is cleaved or otherwise removed from a target-specific primer, an amplified sequence, an adapter or a nucleic acid molecule of the sample. In some embodiments, the cleavage steps involves a chemical, thermal, photo-oxidative or digestive process.

As used herein, the term “hybridization” is consistent with its use in the art, and refers to the process whereby two nucleic acid molecules undergo base pairing interactions. Two nucleic acid molecule molecules are said to be hybridized when any portion of one nucleic acid molecule is base paired with any portion of the other nucleic acid molecule; it is not necessarily required that the two nucleic acid molecules be hybridized across their entire respective lengths and in some embodiments, at least one of the nucleic acid molecules can include portions that are not hybridized to the other nucleic acid molecule. The phrase “hybridizing under stringent conditions” and its variants refers to conditions under which hybridization of a target-specific primer to a target sequence occurs in the presence of high hybridization temperature and low ionic strength. In one exemplary embodiment, stringent hybridization conditions include an aqueous environment containing about 30 mM magnesium sulfate, about 300 mM Tris-sulfate at pH 8.9, and about 90 mM ammonium sulfate at about 60-68° C., or equivalents thereof. As used herein, the phrase “standard hybridization conditions” and its variants refers to conditions under which hybridization of a primer to an oligonucleotide (i.e., a target sequence), occurs in the presence of low hybridization temperature and high ionic strength. In one exemplary embodiment, standard hybridization conditions include an aqueous environment containing about 100 mM magnesium sulfate, about 500 mM Tris-sulfate at pH 8.9, and about 200 mM ammonium sulfate at about 50-55° C., or equivalents thereof.

As used herein, “GC content” and its derivatives, refers to the cytosine and guanine content of a nucleic acid molecule. The GC content of a target-specific primer (or adapter) of the disclosure is 85% or lower. More typically, the GC content of a target-specific primer or adapter of the disclosure is between 15-85%.

As used herein, the term “end” and its variants, when used in reference to a nucleic acid molecule, for example a target sequence or amplified target sequence, can include the terminal 30 nucleotides, the terminal 20 and even more typically the terminal 15 nucleotides of the nucleic acid molecule. A linear nucleic acid molecule comprised of linked series of contiguous nucleotides typically includes at least two ends. In some embodiments, one end of the nucleic acid molecule can include a 3′ hydroxyl group or its equivalent, and can be referred to as the “3′ end” and its derivatives. Optionally, the 3′ end includes a 3′ hydroxyl group that is not linked to a 5′ phosphate group of a mononucleotide pentose ring. Typically, the 3′ end includes one or more 5′ linked nucleotides located adjacent to the nucleotide including the unlinked 3′ hydroxyl group, typically the nucleotides located adjacent to the 3′ hydroxyl, typically the terminal 20 and even more typically the terminal 15 nucleotides. One or more linked nucleotides can be represented as a percentage of the nucleotides present in the oligonucleotide or can be provided as a number of linked nucleotides adjacent to the unlinked 3′ hydroxyl. For example, the 3′ end can include less than 50% of the nucleotide length of the oligonucleotide. In some embodiments, the 3′ end does not include any unlinked 3′ hydroxyl group but can include any moiety capable of serving as a site for attachment of nucleotides via primer extension and/or nucleotide polymerization. In some embodiments, the term “3′ end” for example when referring to a target-specific primer, can include the terminal 10 nucleotides, the terminal 5 nucleotides, the terminal 4, 3, 2 or fewer nucleotides at the 3′end. In some embodiments, the term “3′ end” when referring to a target-specific primer can include nucleotides located at nucleotide positions 10 or fewer from the 3′ terminus.

As used herein, “5′ end”, and its derivatives, refers to an end of a nucleic acid molecule, for example a target sequence or amplified target sequence, which includes a free 5′ phosphate group or its equivalent. In some embodiments, the 5′ end includes a 5′ phosphate group that is not linked to a 3′ hydroxyl of a neighboring mononucleotide pentose ring. Typically, the 5′ end includes to one or more linked nucleotides located adjacent to the 5′ phosphate, typically the 30 nucleotides located adjacent to the nucleotide including the 5′ phosphate group, typically the terminal 20 and even more typically the terminal 15 nucleotides. One or more linked nucleotides can be represented as a percentage of the nucleotides present in the oligonucleotide or can be provided as a number of linked nucleotides adjacent to the 5′ phosphate. For example, the 5′ end can be less than 50% of the nucleotide length of an oligonucleotide. In another exemplary embodiment, the 5′ end can include about 15 nucleotides adjacent to the nucleotide including the terminal 5′ phosphate. In some embodiments, the 5′ end does not include any unlinked 5′ phosphate group but can include any moiety capable of serving as a site of attachment to a a 3′ hydroxyl group, or to the 3′end of another nucleic acid molecule. In some embodiments, the term “5′ end” for example when referring to a target-specific primer, can include the terminal 10 nucleotides, the terminal 5 nucleotides, the terminal 4, 3, 2 or fewer nucleotides at the 5′end. In some embodiments, the term “5′ end” when referring to a target-specific primer can include nucleotides located at positions 10 or fewer from the 5′ terminus. In some embodiments, the 5′ end of a target-specific primer can include only non-cleavable nucleotides, for example nucleotides that do not contain one or more cleavable groups as disclosed herein, or a cleavable nucleotide as would be readily determined by one of ordinary skill in the art.

As used herein, “DNA barcode” or “DNA tagging sequence” and its derivatives, refers to a unique short (6-14 nucleotide) nucleic acid sequence within an adapter that can act as a ‘key’ to distinguish or separate a plurality of amplified target sequences in a sample. For the purposes of this disclosure, a DNA barcode or DNA tagging sequence can be incorporated into the nucleotide sequence of an adapter.

As used herein, the phrases “two rounds of target-specific hybridization” or “two rounds of target-specific selection” and their derivatives refers to any process whereby the same target sequence is subjected to two consecutive rounds of hybridization-based target-specific selection, wherein a target sequence is hybridized to a target-specific sequence. Each round of hybridization based target-specific selection can include multiple target-specific hybridizations to at least some portion of a target-specific sequence. In one exemplary embodiment, a round of target-specific selection includes a first target-specific hybridization involving a first region of the target sequence and a second target-specific hybridization involving a second region of the target sequence. The first and second regions can be the same or different. In some embodiments, each round of hybridization-based target-specific selection can include use of two target specific oligonucleotides (e.g., a forward target-specific primer and a reverse target-specific primer), such that each round of selection includes two target-specific hybridizations.

As used herein, “comparable maximal minimum melting temperatures” and its derivatives, refers to the melting temperature (Tm) of each nucleic acid fragment for a single adapter or target-specific primer after cleavage of the cleavable groups. The hybridization temperature of each nucleic acid fragment generated by a single adapter or target-specific primer is compared to determine the maximal minimum temperature required preventing hybridization of any nucleic acid fragment from the target-specific primer or adapter to the target sequence. Once the maximal hybridization temperature is known, it is possible to manipulate the adapter or target-specific primer, for example by moving the location of the cleavable group along the length of the primer, to achieve a comparable maximal minimum melting temperature with respect to each nucleic acid fragment.

As used herein, “addition only” and its derivatives, refers to a series of steps in which reagents and components are added to a first or single reaction mixture. Typically, the series of steps excludes the removal of the reaction mixture from a first vessel to a second vessel in order to complete the series of steps. An addition only process excludes the manipulation of the reaction mixture outside the vessel containing the reaction mixture. Typically, an addition-only process is amenable to automation and high-throughput.

As used herein, “synthesizing” and its derivatives, refers to a reaction involving nucleotide polymerization by a polymerase, optionally in a template-dependent fashion. Polymerases synthesize an oligonucleotide via transfer of a nucleoside monophosphate from a nucleoside triphosphate (NTP), deoxynucleoside triphosphate (dNTP) or dideoxynucleoside triphosphate (ddNTP) to the 3′ hydroxyl of an extending oligonucleotide chain. For the purposes of this disclosure, synthesizing includes to the serial extension of a hybridized adapter or a target-specific primer via transfer of a nucleoside monophosphate from a deoxynucleoside triphosphate.

As used herein, “polymerizing conditions” and its derivatives, refers to conditions suitable for nucleotide polymerization. In typical embodiments, such nucleotide polymerization is catalyzed by a polymerase. In some embodiments, polymerizing conditions include conditions for primer extension, optionally in a template-dependent manner, resulting in the generation of a synthesized nucleic acid sequence. In some embodiments, the polymerizing conditions include polymerase chain reaction (PCR). Typically, the polymerizing conditions include use of a reaction mixture that is sufficient to synthesize nucleic acids and includes a polymerase and nucleotides. The polymerizing conditions can include conditions for annealing of a target-specific primer to a target sequence and extension of the primer in a template dependent manner in the presence of a polymerase. In some embodiments, polymerizing conditions can be practiced using thermocycling. Additionally, polymerizing conditions can include a plurality of cycles where the steps of annealing, extending, and separating the two nucleic strands are repeated. Typically, the polymerizing conditions include a cation such as MgCl2. Polymerization of one or more nucleotides to form a nucleic acid strand includes that the nucleotides be linked to each other via phosphodiester bonds, however, alternative linkages may be possible in the context of particular nucleotide analogs.

As used herein, the term “nucleic acid” refers to natural nucleic acids, artificial nucleic acids, analogs thereof, or combinations thereof, including polynucleotides and oligonucleotides. As used herein, the terms “polynucleotide” and “oligonucleotide” are used interchangeably and mean single-stranded and double-stranded polymers of nucleotides including, but not limited to, 2′-deoxyribonucleotides (nucleic acid) and ribonucleotides (RNA) linked by internucleotide phosphodiester bond linkages, e.g. 3′-5′ and 2′-5′, inverted linkages, e.g. 3′-3′ and 5′-5′, branched structures, or analog nucleic acids. Polynucleotides have associated counter ions, such as H+, NH4+, trialkylammonium, Mg2+, Na+ and the like. An oligonucleotide can be composed entirely of deoxyribonucleotides, entirely of ribonucleotides, or chimeric mixtures thereof. Oligonucleotides can be comprised of nucleobase and sugar analogs. Polynucleotides typically range in size from a few monomeric units, e.g. 5-40, when they are more commonly frequently referred to in the art as oligonucleotides, to several thousands of monomeric nucleotide units, when they are more commonly referred to in the art as polynucleotides; for purposes of this disclosure, however, both oligonucleotides and polynucleotides may be of any suitable length. Unless denoted otherwise, whenever a oligonucleotide sequence is represented, it will be understood that the nucleotides are in 5′ to 3′ order from left to right and that “A” denotes deoxyadenosine, “C” denotes deoxycytidine, “G” denotes deoxyguanosine, “T” denotes thymidine, and “U” denotes deoxyuridine. Oligonucleotides are said to have “5′ ends” and “3′ ends” because mononucleotides are typically reacted to form oligonucleotides via attachment of the 5′ phosphate or equivalent group of one nucleotide to the 3′ hydroxyl or equivalent group of its neighboring nucleotide, optionally via a phosphodiester or other suitable linkage.

As defined herein, the term “nick translation” and its variants comprise the translocation of one or more nicks or gaps within a nucleic acid strand to a new position along the nucleic acid strand. In some embodiments, a nick can be formed when a double stranded adapter is ligated to a double stranded amplified target sequence. In one example, the primer can include at its 5′ end, a phosphate group that can ligate to the double stranded amplified target sequence, leaving a nick between the adapter and the amplified target sequence in the complementary strand. In some embodiments, nick translation results in the movement of the nick to the 3′ end of the nucleic acid strand. In some embodiments, moving the nick can include performing a nick translation reaction on the adapter-ligated amplified target sequence. In some embodiments, the nick translation reaction can be a coupled 5′ to 3′ DNA polymerization/degradation reaction, or coupled to a 5′ to 3′ DNA polymerization/strand displacement reaction. In some embodiments, moving the nick can include performing a DNA strand extension reaction at the nick site. In some embodiments, moving the nick can include performing a single strand exonuclease reaction on the nick to form a single stranded portion of the adapter-ligated amplified target sequence and performing a DNA strand extension reaction on the single stranded portion of the adapter-ligated amplified target sequence to a new position. In some embodiments, a nick is formed in the nucleic acid strand opposite the site of ligation.

As used herein, the term “polymerase chain reaction” (“PCR”) refers to the method of K. B. Mullis U.S. Pat. Nos. 4,683,195 and 4,683,202, hereby incorporated by reference, which describe a method for increasing the concentration of a segment of a polynucleotide of interest in a mixture of expressed RNA or cDNA without cloning or purification. This process for amplifying the polynucleotide of interest consists of introducing a large excess of two oligonucleotide primers to the DNA mixture containing the desired polynucleotide of interest, followed by a precise sequence of thermal cycling in the presence of a DNA polymerase. The two primers are complementary to their respective strands of the double stranded polynucleotide of interest. To effect amplification, the mixture is denatured and the primers then annealed to their complementary sequences within the polynucleotide of interest molecule. Following annealing, the primers are extended with a polymerase to form a new pair of complementary strands. The steps of denaturation, primer annealing and polymerase extension can be repeated many times (i.e., denaturation, annealing and extension constitute one “cycle”; there can be numerous “cycles”) to obtain a high concentration of an amplified segment of the desired polynucleotide of interest. The length of the amplified segment of the desired polynucleotide of interest (amplicon) is determined by the relative positions of the primers with respect to each other, and therefore, this length is a controllable parameter. By virtue of repeating the process, the method is referred to as the “polymerase chain reaction” (hereinafter “PCR”). Because the desired amplified segments of the polynucleotide of interest become the predominant nucleic acid sequences (in terms of concentration) in the mixture, they are said to be “PCR amplified”. As defined herein, target nucleic acid molecules within a sample including a plurality of target nucleic acid molecules are amplified via PCR. In a modification to the method discussed above, the target nucleic acid molecules can be PCR amplified using a plurality of different primer pairs, in some cases, one or more primer pairs per target nucleic acid molecule of interest, thereby forming a multiplex PCR reaction. Using multiplex PCR, it is possible to simultaneously amplify multiple nucleic acid molecules of interest from a sample to form amplified target sequences. It is also possible to detect the amplified target sequences by several different methodologies (e.g., quantitation with a bioanalyzer or qPCR, hybridization with a labeled probe; incorporation of biotinylated primers followed by avidin-enzyme conjugate detection; incorporation of 32P-labeled deoxynucleotide triphosphates, such as dCTP or dATP, into the amplified target sequence). Any oligonucleotide sequence can be amplified with the appropriate set of primers, thereby allowing for the amplification of target nucleic acid molecules from RNA, cDNA, formalin-fixed paraffin-embedded DNA, fine-needle biopsies and various other sources. In particular, the amplified target sequences created by the multiplex PCR process as disclosed herein, are themselves efficient substrates for subsequent PCR amplification or various downstream assays or manipulations.

As defined herein “multiplex amplification” refers to selective and non-random amplification of two or more target sequences within a sample using at least one target-specific primer. In some embodiments, multiplex amplification is performed such that some or all of the target sequences are amplified within a single reaction vessel. The “plexy” or “plex” of a given multiplex amplification refers to the number of different target-specific sequences that are amplified during that single multiplex amplification. In some embodiments, the plexy can be about 12-plex, 24-plex, 48-plex, 74-plex, 96-plex, 120-plex, 144-plex, 168-plex, 192-plex, 216-plex, 240-plex, 264-plex, 288-plex, 312-plex, 336-plex, 360-plex, 384-plex, or 398-plex.

The practice of the present subject matter may employ, unless otherwise indicated, conventional techniques and descriptions of organic chemistry, molecular biology (including recombinant techniques), cell biology, and biochemistry, which are within the skill of the art. Such conventional techniques include, but are not limited to, preparation of synthetic polynucleotides, polymerization techniques, chemical and physical analysis of polymer particles, preparation of nucleic acid libraries, nucleic acid sequencing and analysis, and the like. Specific illustrations of suitable techniques can be used by reference to the examples provided herein. Other equivalent conventional procedures can also be used. Such conventional techniques and descriptions can be found in standard laboratory manuals such as Genome Analysis: A Laboratory Manual Series (Vols. I-IV), PCR Primer: A Laboratory Manual, and Molecular Cloning: A Laboratory Manual (all from Cold Spring Harbor Laboratory Press), Hermanson, Bioconjugate Techniques, Second Edition (Academic Press, 2008); Merkus, Particle Size Measurements (Springer, 2009); Rubinstein and Colby, Polymer Physics (Oxford University Press, 2003); and the like.

We have developed a multiplex next generation sequencing workflow to measure expression of genes involved in immune response, e.g., checkpoint pathways, T cell related signaling pathways, markers of different tumor infiltrating lymphocyte (TIL) subsets, and tumor markers, in order to determine immune response in a sample. The immune response assay compositions and methods of the invention offer a specific and robust solution for biomarker screening and for understanding mechanisms involved with tumor immune response. Thus, provided are multiplex gene expression compositions for multiplex library preparation and use in conjunction with next generation sequencing technologies and workflow solutions (e.g., Ion Torrent™ NGS workflow), manual or automated, to evaluate pathways that shield tumors from the immune response.

Thus, provided are compositions for a single stream multiplex determination of an immune response in a sample. In some embodiments, the composition consists of a plurality of sets of primer pair reagents directed to a plurality of target sequences to measure the expression levels of the targets in the sample, wherein the target genes are selected from immune response genes consisting of the following function: checkpoint pathways, T cell related signaling pathways, markers of tumor infiltrating lymphocytes (TILS), tumor markers, and housekeeping genes. In some embodiments, the target genes are selected from immune response genes consisting of one or more function of Table A. In some embodiments, the target genes are selected from immune response genes consisting of the following function: immune checkpoint pathways and targets; T and B cell signaling genes, markers of lymphocyte subsets, interferon signaling genes, cytokine signaling genes; tumor markers, tumor antigens, proliferation markers; and housekeeping genes. In some embodiments, the target genes are selected from immune response genes that elucidate T and B lymphocyte functions, from activation to antigen processing, including, e.g., genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation and TCR coexpression. In some embodiments, the target genes are selected from immune response genes that elucidate the level of inflammation and activation and co-op by helper cells, including, e.g., genes consisting of the following function: chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, and Type II interferon signaling. In some embodiments, the target genes identify the presence of various relevant cell types, including, e.g., genes selected from immune response genes consisting of the following function: B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, and T cell differentiation. In some embodiments, the target genes elucidate the mechanism of action, including, e.g., genes selected from immune response genes consisting of the following function: checkpoint pathway, PD-signaling, and drug target. In some embodiments, the target genes demonstrate the proliferative activity and/or stemness characteristics of tumor cells, including, e.g., genes selected from immune response genes consisting of the following function: adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In total, the various functions of genes comprising the provided multiplex panel of the invention provide a comprehensive picture of complex activities of the tumor microenvironment.

In some embodiments, immune response target sequences are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker.

TABLE A Immune Response Genes NCBI Accession No Gene ID Gene Function NM_001025091 ABCF1 Housekeeping NM_078481 ADGRE5 Adhesion, migration NM_000675 ADORA2A Checkpoint pathway NM_001623 AIF1 Macrophage NM_001014431 AKT1 Tumor marker NM_001141 ALOX15B Macrophage NM_000045 ARG1 Myeloid marker NM_021913 AXL Innate immune response NM_018644 B3GAT1 NK activation NM_182482 BAGE Tumor antigen NM_006399 BATF Helper T cells NM_000633 BCL2 Apoptosis NM_138621 BCL2L11 Apoptosis NM_001706 BCL6 Type II interferon signaling NM_007300 BRCA1 Tumor marker NM_000059 BRCA2 Tumor marker NM_004335 BST2 Type I interferon signaling NM_181780 BTLA Checkpoint pathway NM_004336 BUB1 Proliferation NM_022153 C10orf54 Checkpoint pathway NM_015991 C1QA Innate immune response NM_000491 C1QB Innate immune response NM_000717 CA4 Neutrophil NM_170662 CBLB T cell receptor signaling NM_002987 CCL17 Chemokine signaling NM_002988 CCL18 Lymphocyte infiltrate NM_002982 CCL2 Lymphocyte infiltrate NM_004591 CCL20 Chemokine signaling NM_002989 CCL21 Lymphocyte infiltrate NM_002990 CCL22 Chemokine signaling NM_002983 CCL3 Lymphocyte infiltrate NM_002984 CCL4 Lymphocyte infiltrate NM_002985 CCL5 Lymphocyte infiltrate NM_004701 CCNB2 Proliferation NM_001295 CCR1 Cytokine signaling NM_001123396 CCR2 Helper T cells NM_005508 CCR4 Chemokine signaling NM_001100168 CCR5 Lymphocyte infiltrate NM_004367 CCR6 Chemokine signaling NM_001838 CCR7 TCR coexpression NM_000591 CD14 Dendridic cell, macrophage NM_007053 CD160 Checkpoint pathway NM_004244 CD163 Macrophage NM_001178098 CD19 B cell marker NM_001765 CD1C Antigen presentation NM_001766 CD1D Antigen presentation NM_001767 CD2 Lymphocyte infiltrate NM_021155 CD209 Dendridic cell, macrophage NM_001771 CD22 B cell marker NM_006566 CD226 Adhesion, migration NM_001166663 CD244 Checkpoint pathway NM_198053 CD247 TCR coexpression NM_001242 CD27 Drug target NM_014143 CD274 Checkpoint pathway NM_001024736 CD276 Checkpoint pathway NM_006139 CD28 Checkpoint pathway NM_001772 CD33 Myeloid marker NM_001774 CD37 Lymphocyte infiltrate NM_001775 CD38 Adhesion, migration NM_000732 CD3D TCR coexpression NM_000733 CD3E TCR coexpression NM_000073 CD3G TCR coexpression NM_000616 CD4 Helper T cells NM_001250 CD40 Drug target NM_000074 CD40LG T cell receptor signaling NM_000610 CD44 Adhesion, migration NM_001777 CD47 Adhesion, migration NM_001778 CD48 Checkpoint pathway NM_001803 CD52 Lymphocyte infiltrate NM_001040033 CD53 Adhesion, migration NM_006725 CD6 TCR coexpression NM_001780 CD63 Lymphocyte infiltrate NM_001251 CD68 Macrophage NM_001781 CD69 Checkpoint pathway NM_001252 CD70 Drug target NM_001025159 CD74 Antigen processing NM_001783 CD79A B cell receptor signaling NM_001039933 CD79B B cell receptor signaling NM_005191 CD80 Checkpoint pathway NM_004233 CD83 Antigen presentation NM_175862 CD86 Checkpoint pathway NM_171827 CD8A TCR coexpression NM_172213 CD8B TCR coexpression NM_001786 CDK1 Proliferation NM_000077 CDKN2A Tumor marker NM_005192 CDKN3 Proliferation NM_001712 CEACAM1 Checkpoint pathway NM_001816 CEACAM8 Myeloid marker NM_000246 CIITA Type II interferon signaling NM_130441 CLEC4C Dendridic cell NM_001142345 CMKLR1 Dendridic cell, macrophage NM_007074 CORO1A Lymphocyte infiltrate NM_019604 CRTAM TCR coexpression NM_005211 CSF1R Cytokine signaling NM_000395 CSF2RB Cytokine signaling NM_001327 CTAG1B Tumor antigen ENST00000369585 CTAG2 Tumor antigen NM_005214 CTLA4 Drug target NM_004079 CTSS Lymphocyte infiltrate NM_002996 CX3CL1 Type II interferon signaling ENST00000399220 CX3CR1 Lymphocyte infiltrate ENST00000435290 CX3CR1 Lymphocyte infiltrate ENST00000541347 CX3CR1 Lymphocyte infiltrate NM_001171174 CX3CR1 Lymphocyte infiltrate NM_001511 CXCL1 Chemokine signaling NM_001565 CXCL10 Type II interferon signaling NM_005409 CXCL11 Type II interferon signaling NM_006419 CXCL13 Type II interferon signaling NM_000584 CXCL8 Cytokine signaling NM_002416 CXCL9 Type II interferon signaling NM_001557 CXCR2 Chemokine signaling NM_001504 CXCR3 Chemokine signaling NM_003467 CXCR4 Lymphocyte infiltrate NM_001716 CXCR5 Type II interferon signaling NM_006564 CXCR6 Lymphocyte infiltrate NM_000397 CYBB Type II interferon signaling NM_014314 DDX58 Interferon signaling NM_032564 DGAT2 Neutrophil NM_007329 DMBT1 Innate immune response NM_005755 EBI3 T cell regulation NM_005227 EFNA4 Tumor marker NM_005228 EGFR Tumor marker NM_000399 EGR2 T cell differentiation NM_004430 EGR3 Tumor marker NM_001135651 EIF2AK2 Type II interferon signaling NM_001098175 ENTPD1 Checkpoint pathway NM_005442 EOMES Checkpoint pathway NM_000043 FAS B cell receptor signaling NM_000639 FASLG Type II interferon signaling NM_004106 FCER1G Lymphocyte infiltrate NM_000566 FCGR1A B cell marker NM_004001 FCGR2B B cell marker NM_000569 FCGR3A Macrophage NM_000570 FCGR3B NK activation NM_001184866 FCRLA B cell marker NM_021953 FOXM1 Proliferation NM_002015 FOXO1 PD-1 signaling, tumor marker NM_014009 FOXP3 T cell regulation NM_002033 FUT4 Myeloid marker, stem cell NM_001465 FYB Lymphocyte infiltrate NM_000402 G6PD Housekeeping NM_052850 GADD45GIP1 Apoptosis NM_001040663 GAGE1, Tumor antigen GAGE12I, GAGE12F NM_001098413 GAGE10 Tumor antigen NM_001098406 GAGE12J Tumor antigen NM_001098412 GAGE13 Tumor antigen NM_001472 GAGE2C, Tumor antigen GAGE2A, GAGE2E NM_001002295 GATA3 Helper T cells NM_002053 GBP1 Type II interferon signaling NM_006433 GNLY NK activation NM_001098200 GPR18 TCR coexpression NM_004810 GRAP2 TCR coexpression NM_000181 GUSB Housekeeping NM_006144 GZMA Lymphocyte infiltrate NM_004131 GZMB Lymphocyte infiltrate NM_033423 GZMH Lymphocyte infiltrate NM_002104 GZMK Lymphocyte infiltrate NM_032782 HAVCR2 Checkpoint pathway NM_017912 HERC6 Dendridic cell NM_000601 HGF Cytokine signaling NM_001530 HIF1A PD-1 signaling, tumor marker NM_002116 HLA-A Antigen processing NM_005514 HLA-B Antigen processing NM_002117 HLA-C Antigen processing NM_006120 HLA-DMA Antigen processing NM_002118 HLA-DMB Antigen processing NM_002119 HLA-DOA Antigen processing NM_002120 HLA-DOB Antigen processing NM_033554 HLA-DPA1 Antigen processing NM_002121 HLA-DPB1 Antigen processing NM_002122 HLA-DQA1 Antigen processing NM_020056 HLA-DQA2 Antigen processing NM_001198858 HLA-DQB2 Antigen processing NM_019111 HLA-DRA Antigen processing NM_002124 HLA-DRB1 Antigen processing NM_005516 HLA-E Antigen processing NM_001098479 HLA-F Antigen processing NR_026972 HLA-F-AS1 Antigen processing NM_002127 HLA-G Antigen processing NM_000190 HMBS Housekeeping NM_000201 ICAM1 Type II interferon signaling NM_012092 ICOS Checkpoint pathway NM_015259 ICOSLG Checkpoint pathway NM_002166 ID2 T cell regulation NM_002167 ID3 T cell regulation NM_002164 IDO1 Drug target NM_194294 IDO2 Checkpoint pathway NM_005532 IFI27 Type I interferon signaling NM_005533 IFI35 Interferon signaling NM_006820 IFI44L Interferon signaling NM_022873 IFI6 Interferon signaling NM_022168 IFIH1 Innate immune response NM_001548 IFIT1 Type I interferon signaling NM_001547 IFIT2 Cytokine signaling NM_001031683 IFIT3 Type I interferon signaling NM_003641 IFITM1 Type I interferon signaling NM_006435 IFITM2 Type I interferon signaling NM_021268 IFNA17 T cell receptor signaling NM_002176 IFNB1 Type II interferon signaling NM_000619 IFNG Type II interferon signaling NM_000875 IGF1R Adhesion, migration NM_005849 IGSF6 Lymphocyte infiltrate NM_006060 IKZF1 Lymphocyte development NM_016260 IKZF2 Lymphocyte development NM_012481 IKZF3 TCR coexpression NM_022465 IKZF4 Lymphocyte development NM_000572 IL10 Drug target NM_001558 IL10RA Lymphocyte infiltrate NM_000882 IL12A Drug target NM_002187 IL12B Drug target NM_002188 IL13 Cytokine signaling NM_000585 IL15 T cell regulation NM_002190 IL17A Helper T cells NM_052872 IL17F Dendridic cell, macrophage NM_001562 IL18 T cell regulation NM_000575 IL1A Cytokine signaling NM_000576 IL1B Type II interferon signaling NM_000586 IL2 Drug target NM_021803 IL21 Cytokine signaling NM_020525 IL22 T cell regulation NM_016584 IL23A Dendridic cell, macrophage NM_000417 IL2RA Cytokine signaling NM_000878 IL2RB TCR coexpression NM_000206 IL2RG Lymphocyte infiltrate NM_002183 IL3RA Dendridic cell NM_000589 IL4 Cytokine signaling NM_000600 IL6 Cytokine signaling NM_000880 IL7 Cytokine signaling NM_002185 IL7R TCR coexpression NM_002198 IRF1 Type II interferon signaling NM_002460 IRF4 Interferon signaling NM_006084 IRF9 Type II interferon signaling NM_005544 IRS1 Tumor marker NM_005101 ISG15 Type I interferon signaling NM_002201 ISG20 Type I interferon signaling NM_181501 ITGA1 Adhesion, migration NM_002208 ITGAE Adhesion, migration NM_002209 ITGAL Leukocyte migration NM_001145808 ITGAM Leukocyte migration NM_000887 ITGAX Dendridic cell NM_002211 ITGB1 Adhesion, migration NM_000211 ITGB2 Lymphocyte infiltrate NM_000889 ITGB7 Leukocyte migration NM_005546 ITK TCR coexpression NM_001098526 JAML Lymphocyte infiltrate NM_144646 JCHAIN B cell marker NM_014736 KIAA0101 Proliferation NM_014218 KIR2DL1 Drug target ENST00000344867 KIR2DL2 NK cell marker NM_015868 KIR2DL3 NK cell marker NM_016270 KLF2 T cell regulation, trafficking NM_002258 KLRB1 NK activation NM_002262 KLRD1 Drug target NM_016523 KLRF1 NK activation NM_005810 KLRG1 NK activation NM_007360 KLRK1 NK activation NM_032045 KREMEN1 Neutrophil NM_000424 KRT5 Tumor marker NM_005556 KRT7 Tumor marker NM_002286 LAG3 Drug target NM_005561 LAMP1 Lymphocyte infiltrate NM_014398 LAMP3 TCR coexpression NM_006762 LAPTM5 Lymphocyte infiltrate NM_001042771 LCK TCR coexpression NM_005564 LCN2 Innate immune response NM_001110533 LEXM T cell differentiation NM_001081637 LILRB1 Leukocyte inhibition NM_001080978 LILRB2 Lymphocyte infiltrate NM_170707 LMNA Housekeeping NM_052972 LRG1 Neutrophil NM_002332 LRP1 Housekeeping NM_007161 LST1 Leukocyte inhibition NM_002348 LY9 Lymphocyte infiltrate NM_000239 LYZ Innate immune response NM_002355 M6PR T cell regulation NM_002358 MAD2L1 Proliferation NM_130760 MADCAM1 Adhesion, migration NM_004988 MAGEA1 Tumor antigen NM_021048 MAGEA10 Tumor antigen NM_005367 MAGEA12 Tumor antigen NM_005362 MAGEA3 Tumor antigen NM_001011548 MAGEA4 Tumor antigen NM_016249 MAGEC2 Tumor antigen NM_002745 MAPK1 Tumor marker NM_139012 MAPK14 Innate immune response NM_014791 MELK Proliferation NM_002415 MIF Innate immune response NM_002417 MKI67 Proliferation NM_005511 MLANA Tumor antigen NM_004530 MMP2 Tumor marker NM_004994 MMP9 Tumor marker NM_000250 MPO Myeloid marker NM_002438 MRC1 Dendridic cell, macrophage NM_021950 MS4A1 Drug target NM_004958 MTOR PD-1 signaling, tumor marker NM_001178046 MX1 Interferon signaling NM_002467 MYC Tumor marker NM_181351 NCAM1 Adhesion, migration NM_000265 NCF1 Chemokine signaling NM_004829 NCR1 NK cell marker NM_147130 NCR3 NK cell marker NM_001042724 NECTIN2 Adhesion, migration NM_172387 NFATC1 PD-1 signaling NM_020529 NFKBIA T cell receptor signaling NM_005601 NKG7 Lymphocyte infiltrate NM_000625 NOS2 Innate immune response NM_000435 NOTCH3 Tumor marker NM_003873 NRP1 Dendridic cell NM_002526 NT5E Checkpoint pathway NM_006181 NTN3 B cell marker NM_016816 OAS1 Type II interferon signaling NM_016817 OAS2 Interferon signaling NM_006187 OAS3 Interferon signaling NM_005018 PDCD1 Drug target NM_025239 PDCD1LG2 Checkpoint pathway NM_000442 PECAM1 Adhesion, migration NM_002632 PGF Tumor marker NM_006218 PIK3CA PD-1 signaling, tumor marker NM_005026 PIK3CD PD-1 signaling, tumor marker NM_006928 PMEL Drug target NM_000937 POLR2A Housekeeping NM_006235 POU2AF1 B cell marker NM_001198 PRDM1 PD-1 signaling NM_005041 PRF1 NK activation NM_002800 PSMB9 Type II interferon signaling NM_000314 PTEN PD-1 signaling, tumor marker NM_000963 PTGS2 Tumor marker NM_002821 PTK7 Tumor marker NM_002834 PTPN11 PD-1 signaling, tumor marker NM_080548 PTPN6 T cell receptor signaling NM_001199797 PTPN7 Lymphocyte infiltrate NM_002838 PTPRC Lymphocyte infiltrate ENST00000326294 PTPRCAP TCR coexpression NM_006505 PVR Checkpoint pathway NM_002863 PYGL Neutrophil NM_000321 RB1 Tumor marker NM_005060 RORC Helper T cells NM_001010 RPS6 Tumor marker NM_002964 S100A8 Myeloid marker, MDSC NM_002965 S100A9 Myeloid marker, MDSC NM_015474 SAMHD1 Lymphocyte infiltrate NM_004168 SDHA Housekeeping NM_000655 SELL Leukocyte migration NM_002351 SH2D1A Lymphocyte activation NM_053282 SH2D1B Lymphocyte activation NM_014450 SIT1 Lymphocyte infiltrate NM_003930 SKAP2 B cell marker NM_021181 SLAMF7 Drug target NM_020125 SLAMF8 Lymphocyte infiltrate NM_005985 SNAI1 Tumor marker, stemness NM_003068 SNAI2 Tumor marker, stemness NM_002727 SRGN Lymphocyte infiltrate NM_003147 SSX2 Tumor antigen NM_007315 STAT1 Type II interferon signaling NM_139276 STAT3 Drug target NM_003151 STAT4 Helper T cells NM_003152 STAT5A Cytokine signaling NM_003153 STAT6 Helper T cells NM_054114 TAGAP Lymphocyte infiltrate NM_000593 TAP1 Type II interferon signaling NM_001003806 TARP Lymphocyte infiltrate NM_003194 TBP Housekeeping NM_013351 TBX21 Type II interferon signaling NM_003202 TCF7 Tumor marker NM_005651 TDO2 Checkpoint pathway NM_001128148 TFRC Housekeeping NM_000660 TGFB1 Checkpoint pathway NM_173799 TIGIT TCR coexpression NM_003265 TLR3 Dendridic cell NM_016562 TLR7 Innate immune response NM_138636 TLR8 Lymphocyte infiltrate NM_017442 TLR9 Drug target NM_000594 TNF Checkpoint pathway NM_014350 TNFAIP8 Lymphocyte infiltrate NM_003820 TNFRSF14 Checkpoint pathway NM_001192 TNFRSF17 B cell marker NM_004195 TNFRSF18 Drug target NM_003327 TNFRSF4 Drug target NM_001561 TNFRSF9 Drug target NM_003810 TNFSF10 Apoptosis NM_006573 TNFSF13B B cell marker NM_003807 TNFSF14 Checkpoint pathway NM_005092 TNFSF18 Checkpoint pathway NM_003326 TNFSF4 Checkpoint pathway NM_003811 TNFSF9 Cytokine signaling NM_001067 TOP2A Proliferation NM_003722 TP63 Tumor marker NM_012101 TRIM29 Tumor marker NM_178014 TUBB Housekeeping NM_000474 TWIST1 Tumor marker, stemness NM_198125 TYROBP Lymphocyte infiltrate NM_001078 VCAM1 Leukocyte migration NM_001171623 VEGFA Chemokine signaling NM_024626 VTCN1 Checkpoint pathway NM_001097594 XAGE1B Tumor antigen NM_001079 ZAP70 T cell receptor signaling NM_025224 ZBTB46 Dendridic cell NM_001174093 ZEB1 Tumor marker, stemness

In some embodiments, immune response target sequences are selected from immune response genes consisting of the following functions: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, and drug target. In some embodiments, immune response target sequences are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In some embodiments, immune response target sequences are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In some embodiments, immune response target sequences are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In some embodiments, immune response target sequences are selected from immune response genes consisting of the following function: chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker.

In certain embodiments, target immune response sequences are directed to sequences having aberrant expression associated with cancer. In some embodiments, the target sequences or amplified target sequences are directed to sequences having aberrant expression associated with one or more solid tumor cancers selected from the group consisting of head and neck cancers (e.g., HNSCC, nasopharyngeal, salivary gland), brain cancer (e.g., glioblastoma, glioma, gliosarcoma, glioblastoma multiforme, neuroblastoma), breast cancer (e.g., TNBC, trastuzumab resistant HER2+ breast cancer, ER+/HER− breast cancer), gynecological (e.g., uterine, ovarian cancer, cervical cancer, endometrial cancer, fallopian cancer), colorectal cancer, gallbladder cancer, esophageal cancer, gastrointestinal cancer, gastric cancer, bladder cancer, prostate cancer, testicular cancer, urothelial cancer, liver cancer (e.g., hepatocellular, HCC), lung cancer (e.g., non-small cell lung, small cell lung), kidney (renal cell) cancer, pancreatic cancer (e.g., adenocarcinoma, ductal), thyroid cancer, bile duct cancer, pituitary tumor, wilms tumor, kaposi sarcoma, hairy cell carcinoma, osteosarcoma, thymus cancer, skin cancer, melanoma, heart cancer, oral and larynx cancer, neuroblastoma, mesothelioma, and other solid tumors (thymic, bone, soft tissue, oral SCC, myelofibrosis, synovial sarcoma). In one embodiment, the mutations can include substitutions, insertions, inversions, point mutations, deletions, mismatches and translocations. In some embodiments, the target sequences or amplified target sequences are directed to sequences having aberrant expression associated with one or more blood/hematologic cancers selected from the group consisting of multiple myeloma, diffuse large B cell lymphoma (DLBCL), lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, follicular lymphoma, leukemia, acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), myelodisplastic syndrome. In one embodiment, the aberrant expression associated with cancer are located in at least one of the genes provided in Table 1.

In some embodiments, one or more aberrant expression immune response sequences are located in at least one of the genes selected from CD63, CD69, CXCL1, KLRD1, HLA-DOB, CXCR5, IL12B, PTK7, CEACAM1, CXCL9, IL13, NT5E, VEGFA, ABCF1, CD38, JAML, S100A8, MYC, IRF1, CCL22, CXCR2, IFIT1, IFIT2, CD68, M6PR, SH2D1A, ISG20, GBP1, TBP, STAT6, ID3, CX3CL1, KLRB1, TNFSF4, CD52, IL10RA, HLA-DOA, IFNB1, CCR5, IKZF3, STAT1, CD6, BRCA1, CORO1A, TBX21, KLRK1, CXCR6, PTEN, PMEL, DMBT1, IFI44L, LAPTM5, CD226, TNFSF13B, ICOS, CD160, TRIM29, LST1, ZBTB46, VTCN1, KREMEN1, PDCD1LG2, TUBB, CLEC4C, CD86, HAVCR2, GZMH, NFATC1, CD8B, BCL2, GADD45GIP1, CBLB, ITGA1, CD8A, IL2RA, EIF2AK2, MADCAM1, PTPN6, LRG1, ADGRE5, SH2D1B, ITGB2, HLA-DPA1, DGAT2, IGF1R, TAGAP, LMNA, NCAM1, TIGIT, IL17F, HLA-F-AS1, CD247, CD79B, IDO2, IL4, TYROBP, BTLA, AKT1, IL2RG, POLR2A, ITGAX, IL1B, CSF2RB, DDX58, KIAA0101, CD274, LAMP3, TNFAIP8, FOXP3, IL12A, SAMHD1, SIT1, CD3E, ICOSLG, HGF, MELK, IGSF6, GNLY, TDO2, KRT7, HLA-E, HLA-DM, LAMP1, NTN3, CD28, TARP, EGFR, CCR4, MAGEA3, BATF, KLRG1, IRS1, CSF1R, CTLA4, TNFSF18, POU2AF1, GZMA, PIK3CA, ITK, IFI27, EOMES, LCN2, CD80, CD83, CXCL13, MTOR, FCER1G, TFRC, RORC, MMP9, BST2, PIK3CD, FCGR2B, TNFRSF14, OAS3, GRAP2, CCNB2, MLANA, MAGEA12, VCAM1, CDKN3, NCR1, FAS, GZMB, IRF9, IFITM2, TNFSF14, HLA-B, SDHA, NRP1, EBI3, EFNA4, PVR, BUB1, SKAP2, PRF1, CCL20, TNFRSF18, CTSS, NKG7, ISG15, PDCD1, SNAI1, CXCL11, CIITA, IFI35, TNFSF9, TNFSF10, MMP2, EGR3, MAGEA1, CD163, IL6, KLRF1, B3GAT1, C1QA, OAS1, IKZF2, TLR9, KLF2, GUSB, NFKBIA, IL23A, HERC6, SLAMF8, IL15, TLR7, OAS2, HLA-DR, CRTAM, MAGEC2, ICAM1, CD4, MAPK14, C1QB, NOTCH3, NCR3, STAT3, TLR8, CYBB, IKZF4, IFIH1, LCK, BCL2L11, ITGAM, ITGB7, JCHAIN, CD209, SLAMF7, IL10, IL1A, FCGR3A, IFNA17, EGR2, TOP2A, C10orf54, FOXM1, AXL, MS4A1, IFI6, CD3D, GPR18, CD3G, ZAP70, HMBS, IL7, IFIT3, RB1, PTGS2, TGFB1, NCF1, TWIST1, CA4, SELL, LILRB1, CD14, ALOX15B, PECAM1, NOS2, FASLG, CD44, ENTPD1, CMKLR1, CD53, TNF, CXCL8, CD40LG, HLA-F, GATA3, LYZ, ARG1, IL2RB, NECTIN2, MPO, CCR2, BRCA2, ADORA2A, G6PD, TAP1, MX1, HLA-DQB2, CD27, CD276, STAT4, PTPN7, PTPRC, PSMB9, CD244, CXCR4, MAPK1, TP63, IRF4, CCL3, CCL18, IL7R, HLA-DRB1, CEACAM8, CXCL10, CCL2, SRGN, CD19, ITGB1, IFITM1, CCL21, MRC1, PGF, ITGAL, ID2, CD22, CCL17, ITGAE, IL3RA, CCR7, CD1C, MAD2L1, PYGL, CD40, LY9, HLA-G, TLR3, CD48, STAT5A, FCRLA, BCL6, ZEB1, CCL5, IDO1, IL18, TNFRSF9, HIF1A, HLA-DPB1, FOXO1, CD33, S100A9, HLA-DMB, HLA-A, SNAI2, TNFRSF17, LRP1, MAGEA4, HLA-DQA1, CD1D, RPS6, MKI67, GZMK, CD79A, CD37, FUT4, AIF1, CCR1, PRDM1, CD47, CD74, LAG3, TNFRSF4, CD2, CCL4, BAGE, LEXM, CCR6, CD70, CDK1, CTAG1B, CTAG2, CX3CR1, CX3CR1, CX3CR1, CX3CR1, GAGE1, GAGE12I, GAGE12F, GAGE12J, GAGE2C, GAGE2A, GAGE2E, GAGE10, GAGE13, IKZF1, IL17A, IL2, IL21, IL22, KIR2DL2, KIR2DL3, MAGEA10, MIF, PTPRCAP, SSX2, TCF7, XAGE1B, CEACAM8, CXCR3, FCGR1A, FCGR3B, FYB, HLA-C, HLA-DQA2, IFNG, KIR2DL1, KRT5, LMNA, and PTPN11. In some embodiments the one or more aberrant expression sequences indicate cancer activity.

In some embodiments the one or more aberrant expression sequences indicate a patient's likelihood to response to a therapeutic agent. In some embodiments, the one or more aberrant expression sequences indication a patient's likelihood to not be responsive to a therapeutic agent. In certain embodiments, relevant therapeutic agents can be immunotherapies including but not limited to checkpoint blockades, T cell therapies, and therapeutic vaccines. In some embodiments a therapeutic agent may modify an immune response gene selected from PD1 (e.g., nivolumab, pembrolizumab, AMP-244, MEDI0680, AMP-514, pidlizumab), CTLA4 (e.g., ipilimumab, tremelimumab), PD-L1 (e.g., atezolizumab, MDX1105-01, MEDI4736, avelumab), MR (e.g., lirilumab, NCC0141-0000-0100), CD9/NKG2A (e.g., IPH2201), LAG3 (e.g., BMS986016), GITR (e.g., TRX518), OX40 (e.g., MEDI6383, MEDI6489, MOXR0916), IDO (e.g., indoximod (NLG8189), INCB024360, F001287, NLG919), TGFbeta (e.g., sotaracept, fresolumimab, trabedersen, lucanix), TGFbetaR (e.g., LY2157299, ACE536), CD137 (e.g., urelumab), CD137/41BB (e.g., pf05082566), CD289/TLR9 (e.g., MGN1703), MUC1/CD227 (e.g., OINT-10, ASN-004), CD27 (e.g., varlilumab), CD27L (e.g., AMG172), SLAMF7/CD1 (e.g., elotuzumab), CD20 (e.g., DI-Leu16-IL2), talimogene laherparepvec, CD70 (e.g., ARGX110), IL10 (e.g., AM0010), PSA (e.g., PROSTVAC), GP100 (e.g., MDX1379), STAT3 (e.g., AZD9150), CVAC, IL12 (e.g., veledimex, INXN2001, MSB0010360N, IMMUNOPULSE, GEN-1, INO-9012; IL2: MSB0010445, RG7813/R06895882), IL33 (e.g., alarmin IL33), ICT140, CNDO-109, hTERT (e.g., INO-1400), dysplastic tissues (e.g., ADXS-HPV), SMAC-mimetic (e.g., birinapant), ImmTACs (e.g., IMCgp100), and CD40 (e.g., RO7009789. In certain embodiments, relevant therapeutic agent is an immunotherapy selected from checkpoint blockade, T cell therapy, and therapeutic vaccine. In some embodiments a therapeutic agent modifies an immune response gene selected from any of PD1 (e.g., nivolumab, pembrolizumab, AMP-244, MEDI0680, AMP-514, pidlizumab), CTLA4 (e.g., ipilimumab, tremelimumab), PD-L1 (e.g., atezolizumab, MDX1105-01, MEDI4736, avelumab), MR (e.g., lirilumab, NCC0141-0000-0100), CD9/NKG2A (e.g., IPH2201), LAG3 (e.g., BMS986016), GITR (e.g., TRX518), OX40 (e.g., MEDI6383, MEDI6489, MOXR0916), IDO (e.g., indoximod (NLG8189), INCB024360, F001287, NLG919), TGFbeta (e.g., sotaracept, fresolumimab, trabedersen, lucanix), TGFbetaR (e.g., LY2157299, ACE536), CD137 (e.g., urelumab), CD137/41BB (e.g., pf05082566), CD289/TLR9 (e.g., MGN1703), MUC1/CD227 (e.g., OINT-10, ASN-004), CD27 (e.g., varlilumab), CD27L (e.g., AMG172), SLAMF7/CD1 (e.g., elotuzumab), CD20 (e.g., DI-Leu16-IL2), talimogene laherparepvec, CD70 (e.g., ARGX110), IL10 (e.g., AM0010), PSA (e.g., PROSTVAC), GP100 (e.g., MDX1379), STAT3 (e.g., AZD9150), CVAC, IL12 (e.g., veledimex, INXN2001, MSB0010360N, IMMUNOPULSE, GEN-1, INO-9012; IL2: MSB0010445, RG7813/R06895882), IL33 (e.g., alarmin IL33), ICT140, CNDO-109, hTERT (e.g., INO-1400), dysplastic tissues (e.g., ADXS-HPV), SMAC-mimetic (e.g., birinapant), ImmTACs (e.g., IMCgp100), and CD40 (e.g., RO7009789.

In some embodiments, target sequences or amplified target sequences are directed to mutations associated with cancer. In some embodiments, the target sequences or amplified target sequences are directed to mutations associated with one or more solid tumor cancers selected from the group consisting of head and neck cancers (e.g., HNSCC, nasopharyngeal, salivary gland), brain cancer (e.g., glioblastoma, glioma, gliosarcoma, glioblastoma multiforme, neuroblastoma), breast cancer (e.g., TNBC, trastuzumab resistant HER2+ breast cancer, ER+/HER− breast cancer), gynecological (e.g., uterine, ovarian cancer, cervical cancer, endometrial cancer, fallopian cancer), colorectal cancer, gallbladder cancer, esophageal cancer, gastrointestinal cancer, gastric cancer, bladder cancer, prostate cancer, testicular cancer, urothelial cancer, liver cancer (e.g., hepatocellular, HCC), lung cancer (e.g., non-small cell lung, small cell lung), kidney (renal cell) cancer, pancreatic cancer (e.g., adenocarcinoma, ductal), thyroid cancer, bile duct cancer, pituitary tumor, wilms tumor, kaposi sarcoma, hairy cell carcinoma, osteosarcoma, thymus cancer, skin cancer, melanoma, heart cancer, oral and larynx cancer, neuroblastoma, mesothelioma, and other solid tumors (thymic, bone, soft tissue, oral SCC, myelofibrosis, synovial sarcoma). In one embodiment, the mutations can include substitutions, insertions, inversions, point mutations, deletions, mismatches and translocations. In one embodiment, the mutations can include variation in copy number. In one embodiment, the mutations can include germline or somatic mutations. In some embodiments, the target sequences or amplified target sequences are directed to sequences having mutations associated with one or more blood/hematologic cancers selected from the group consisting of multiple myeloma, diffuse large B cell lymphoma (DLBCL), lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, follicular lymphoma, leukemia, acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), myelodisplastic syndrome. In one embodiment, the mutations associated with cancer are located in at least one of the genes provided in Table 1.

In some embodiments, one or more mutations in immune response sequences associated with cancer are located in at least one of the genes selected from CD63, CD69, CXCL1, KLRD1, HLA-DOB, CXCR5, IL12B, PTK7, CEACAM1, CXCL9, IL13, NT5E, VEGFA, ABCF1, CD38, JAML, S100A8, MYC, IRF1, CCL22, CXCR2, IFIT1, IFIT2, CD68, M6PR, SH2D1A, ISG20, GBP1, TBP, STAT6, ID3, CX3CL1, KLRB1, TNFSF4, CD52, IL10RA, HLA-DOA, IFNB1, CCR5, IKZF3, STAT1, CD6, BRCA1, CORO1A, TBX21, KLRK1, CXCR6, PTEN, PMEL, DMBT1, IFI44L, LAPTM5, CD226, TNFSF13B, ICOS, CD160, TRIM29, LST1, ZBTB46, VTCN1, KREMEN1, PDCD1LG2, TUBB, CLEC4C, CD86, HAVCR2, GZMH, NFATC1, CD8B, BCL2, GADD45GIP1, CBLB, ITGA1, CD8A, IL2RA, EIF2AK2, MADCAM1, PTPN6, LRG1, ADGRE5, SH2D1B, ITGB2, HLA-DPA1, DGAT2, IGF1R, TAGAP, LMNA, NCAM1, TIGIT, IL17F, HLA-F-AS1, CD247, CD79B, IDO2, IL4, TYROBP, BTLA, AKT1, IL2RG, POLR2A, ITGAX, IL1B, CSF2RB, DDX58, KIAA0101, CD274, LAMP3, TNFAIP8, FOXP3, IL12A, SAMHD1, SIT1, CD3E, ICOSLG, HGF, MELK, IGSF6, GNLY, TDO2, KRT7, HLA-E, HLA-DM, LAMP1, NTN3, CD28, TARP, EGFR, CCR4, MAGEA3, BATF, KLRG1, IRS1, CSF1R, CTLA4, TNFSF18, POU2AF1, GZMA, PIK3CA, ITK, IFI27, EOMES, LCN2, CD80, CD83, CXCL13, MTOR, FCER1G, TFRC, RORC, MMP9, BST2, PIK3CD, FCGR2B, TNFRSF14, OAS3, GRAP2, CCNB2, MLANA, MAGEA12, VCAM1, CDKN3, NCR1, FAS, GZMB, IRF9, IFITM2, TNFSF14, HLA-B, SDHA, NRP1, EBI3, EFNA4, PVR, BUB1, SKAP2, PRF1, CCL20, TNFRSF18, CTSS, NKG7, ISG15, PDCD1, SNAI1, CXCL11, CIITA, IFI35, TNFSF9, TNFSF10, MMP2, EGR3, MAGEA1, CD163, IL6, KLRF1, B3GAT1, C1QA, OAS1, IKZF2, TLR9, KLF2, GUSB, NFKBIA, IL23A, HERC6, SLAMF8, IL15, TLR7, OAS2, HLA-DR, CRTAM, MAGEC2, ICAM1, CD4, MAPK14, C1QB, NOTCH3, NCR3, STAT3, TLR8, CYBB, IKZF4, IFIH1, LCK, BCL2L11, ITGAM, ITGB7, JCHAIN, CD209, SLAMF7, IL10, IL1A, FCGR3A, IFNA17, EGR2, TOP2A, C10orf54, FOXM1, AXL, MS4A1, IFI6, CD3D, GPR18, CD3G, ZAP70, HMBS, IL7, IFIT3, RB1, PTGS2, TGFB1, NCF1, TWIST1, CA4, SELL, LILRB1, CD14, ALOX15B, PECAM1, NOS2, FASLG, CD44, ENTPD1, CMKLR1, CD53, TNF, CXCL8, CD40LG, HLA-F, GATA3, LYZ, ARG1, IL2RB, NECTIN2, MPO, CCR2, BRCA2, ADORA2A, G6PD, TAP1, MX1, HLA-DQB2, CD27, CD276, STAT4, PTPN7, PTPRC, PSMB9, CD244, CXCR4, MAPK1, TP63, IRF4, CCL3, CCL18, IL7R, HLA-DRB1, CEACAM8, CXCL10, CCL2, SRGN, CD19, ITGB1, IFITM1, CCL21, MRC1, PGF, ITGAL, ID2, CD22, CCL17, ITGAE, IL3RA, CCR7, CD1C, MAD2L1, PYGL, CD40, LY9, HLA-G, TLR3, CD48, STAT5A, FCRLA, BCL6, ZEB1, CCL5, IDO1, IL18, TNFRSF9, HIF1A, HLA-DPB1, FOXO1, CD33, S100A9, HLA-DMB, HLA-A, SNAI2, TNFRSF17, LRP1, MAGEA4, HLA-DQA1, CD1D, RPS6, MKI67, GZMK, CD79A, CD37, FUT4, AIF1, CCR1, PRDM1, CD47, CD74, LAG3, TNFRSF4, CD2, CCL4, BAGE, LEXM, CCR6, CD70, CDK1, CTAG1B, CTAG2, CX3CR1, CX3CR1, CX3CR1, CX3CR1, GAGE1, GAGE12I, GAGE12F, GAGE12J, GAGE2C, GAGE2A, GAGE2E, GAGE10, GAGE13, IKZF1, IL17A, IL2, IL21, IL22, KIR2DL2, KIR2DL3, MAGEA10, MIF, PTPRCAP, SSX2, TCF7, XAGE1B, CEACAM8, CXCR3, FCGR1A, FCGR3B, FYB, HLA-C, HLA-DQA2, IFNG, KIR2DL1, KRT5, LMNA, and PTPN11

In some embodiments, amplified target sequences are directed to any one of more of the genes provided in Table 1. In some embodiments, amplified target sequences comprise any one or more amplicon sequences provided in Table 1. In some embodiments, amplified target sequences consist of any one or more amplicon sequences provided in Table 1. In some embodiments, amplified target sequences include each amplicon sequence provided in Table 1.

In some embodiments, compositions comprise any one or more of the immune response target-specific primer pairs provided in Table 2. In some embodiments, compositions comprise all of the immune response target-specific primer pairs provided in Table 2. In some embodiments, any one or more of the immune response target-specific primer pairs provided in Table 2 can be used to amplify a target sequence present in a sample as disclosed by the methods described herein.

In some embodiments, the immune response target-specific primers from Table 2 can include 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 398 or more, target-specific primer pairs. In some embodiments, the amplified target sequences can include any one or more of the amplified target sequences provided in Table 1 (e.g., using amplicon ID target-specific primers provided in Table 2). In some embodiments, at least one of the target-specific primers associated with cancer is at least 90% identical to at least one nucleic acid sequence selected from SEQ ID NOs: 399-1194. In some embodiments, at least one of the target-specific primers associated with immune response is complementary across its entire length to at least one target sequence in a sample. In some embodiments, at least one of the target-specific primers associated with immune response includes a non-cleavable nucleotide at the 3′ end. In some embodiments, the non-cleavable nucleotide at the 3′ end includes the terminal 3′ nucleotide. In one embodiment, the amplified target sequences are directed to one or more individual exons having aberrant expression associated with cancer. In one embodiment, the amplified target sequences are directed to individual exons having a mutation associated with cancer. In some embodiments, provided methods comprise selective amplification of more than one target sequences in a sample and the detection and/or identification of mutations associated with cancer. In some embodiments, the amplified target sequences include two or more nucleotide sequences provided in Table 2. In some embodiments, the amplified target sequences can include any one or more the amplified target sequences generated using the target-specific primers provided in Table 2. In one embodiment, the amplified target sequences include 10, 50, 100, 150, 200, 250, 300, 350 or more amplicons from Table 1. In some embodiments, methods comprise detection and optionally, the identification of clinically actionable markers. As defined herein, the term “clinically actionable marker” includes clinically actionable mutations and/or clinically actionable expression patterns that are known or can be associated by one of ordinary skill in the art with, but not limited to, prognosis for the treatment of cancer. In one embodiment, prognosis for the treatment of cancer includes the identification of mutations and/or expression patterns associated with responsiveness or non-responsiveness of a cancer to a drug, drug combination, or treatment regime. In one embodiment, methods comprise amplification of a plurality of target sequences from a population of nucleic acid molecules linked to, or correlated with, the onset, progression or remission of cancer.

In some embodiments, housekeeping genes are comprised in the immune response assay. In certain embodiments, one or more housekeeping gene sequences are included in the assay. In particular embodiments the one or more housekeeping genes are selected from ABCF1, G6PD, GUSB, HMBS, LMNA, LRP1, POLR2A, SDHA, TBP, TFRC, and TUBB.

Provided herein are methods for determining immune response activity in a sample. In some embodiments, the method comprises multiplex amplification of a plurality of target expression sequences from a biological sample, wherein amplifying comprises contacting at least a portion of the sample with a plurality of sets of primer pair reagents directed to the plurality of target sequences, and a polymerase under amplification conditions, to thereby produce amplified target expression sequences. The method further comprises detecting the levels of expression of the target sequences in the sample, wherein a change in the level of expression of one or more immune response markers as compared with a control determines a change in immune response activity in the sample. In some embodiments the target expression sequences of the methods are selected from immune response genes consisting of the following function: checkpoint pathways, T cell related signaling pathways, markers of tumor infiltrating lymphocytes (TILS), tumor markers, and housekeeping genes. In some embodiments, the target genes are selected from immune response genes consisting of the following function: immune checkpoint pathways and targets; T and B cell signaling genes, markers of lymphocyte subsets, interferon signaling genes, cytokine signaling genes; tumor markers, tumor antigens, proliferation markers; and housekeeping genes. In some embodiments, the target genes of the methods are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation and TCR coexpression. In some embodiments, the target genes of the methods are selected from immune response genes consisting of the following function: chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, and Type II interferon signaling. In some embodiments, the target genes of the methods are selected from immune response genes consisting of the following function of markers: B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, and T cell differentiation. In some embodiments, the target genes of the methods are selected from immune response genes consisting of the following function of marker: checkpoint pathway, PD-signaling, and drug target. In some embodiments, the target genes of the methods are selected from immune response genes consisting of the following function of markers: adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker.

In some embodiments, immune response target sequences of the methods are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker.

In some embodiments, immune response target sequences of the methods are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, and drug target. In some embodiments, immune response target sequences of the methods are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In some embodiments, immune response target sequences of the methods are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In some embodiments, immune response target sequences of the methods are selected from immune response genes consisting of the following function: antigen presentation, antigen processing, innate immune response, leukocyte inhibition, leukocyte migration, lymphocyte activation, lymphocyte development, lymphocyte infiltrate, B cell receptor signaling, T cell receptor signaling, T cell regulation, TCR coexpression, chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker. In some embodiments, immune response target sequences of the methods are selected from immune response genes consisting of the following function: chemokine signaling, cytokine signaling, interferon signaling, Type I interferon signaling, Type II interferon signaling, B cell marker, dendritic cell, dendritic cell/macrophage, helper T cell, macrophage, myeloid, neutrophil, NK activation, NK cell, T cell differentiation, checkpoint pathway, PD-signaling, drug target, adhesion/migration, apoptosis, proliferation, tumor antigen, and tumor marker.

In certain embodiments, target immune response sequences of the methods are directed to sequences having aberrant expression associated with cancer. In some embodiments, the target sequences or amplified target sequences of the methods are directed to sequences having aberrant expression associated with associated with one or more solid tumor cancers selected from the group consisting of head and neck cancers (e.g., HNSCC, nasopharyngeal, salivary gland), brain cancer (e.g., glioblastoma, glioma, gliosarcoma, glioblastoma multiforme, neuroblastoma), breast cancer (e.g., TNBC, trastuzumab resistant HER2+ breast cancer, ER+/HER− breast cancer), gynecological (e.g., uterine, ovarian cancer, cervical cancer, endometrial cancer, fallopian cancer), colorectal cancer, gallbladder cancer, esophageal cancer, gastrointestinal cancer, gastric cancer, bladder cancer, prostate cancer, testicular cancer, urothelial cancer, liver cancer (e.g., hepatocellular, HCC), lung cancer (e.g., non-small cell lung, small cell lung), kidney (renal cell) cancer, pancreatic cancer (e.g., adenocarcinoma, ductal), thyroid cancer, bile duct cancer, pituitary tumor, wilms tumor, kaposi sarcoma, hairy cell carcinoma, osteosarcoma, thymus cancer, skin cancer, melanoma, heart cancer, oral and larynx cancer, neuroblastoma, mesothelioma, and other solid tumors (thymic, bone, soft tissue, oral SCC, myelofibrosis, synovial sarcoma). In certain embodiments, the mutations can include any of substitutions, insertions, inversions, point mutations, deletions, mismatches and translocations. In some embodiments, the target sequences or amplified target sequences are directed to sequences having aberrant expression associated with one or more blood/hematologic cancers selected from the group consisting of multiple myeloma, diffuse large B cell lymphoma (DLBCL), lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, follicular lymphoma, leukemia, acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), myelodisplastic syndrome. In certain embodiments, aberrant expression associated with cancer involves in at least one of the genes provided in Table 1.

In some embodiments, one or more aberrant expression immune response sequences of the methods are at least one of the genes selected from CD63, CD69, CXCL1, KLRD1, HLA-DOB, CXCR5, IL12B, PTK7, CEACAM1, CXCL9, IL13, NT5E, VEGFA, ABCF1, CD38, JAML, S100A8, MYC, IRF1, CCL22, CXCR2, IFIT1, IFIT2, CD68, M6PR, SH2D1A, ISG20, GBP1, TBP, STAT6, ID3, CX3CL1, KLRB1, TNFSF4, CD52, IL10RA, HLA-DOA, IFNB1, CCR5, IKZF3, STAT1, CD6, BRCA1, CORO1A, TBX21, KLRK1, CXCR6, PTEN, PMEL, DMBT1, IFI44L, LAPTM5, CD226, TNFSF13B, ICOS, CD160, TRIM29, LST1, ZBTB46, VTCN1, KREMEN1, PDCD1LG2, TUBB, CLEC4C, CD86, HAVCR2, GZMH, NFATC1, CD8B, BCL2, GADD45GIP1, CBLB, ITGA1, CD8A, IL2RA, EIF2AK2, MADCAM1, PTPN6, LRG1, ADGRE5, SH2D1B, ITGB2, HLA-DPA1, DGAT2, IGF1R, TAGAP, LMNA, NCAM1, TIGIT, IL17F, HLA-F-AS1, CD247, CD79B, IDO2, IL4, TYROBP, BTLA, AKT1, IL2RG, POLR2A, ITGAX, IL1B, CSF2RB, DDX58, KIAA0101, CD274, LAMP3, TNFAIP8, FOXP3, IL12A, SAMHD1, SIT1, CD3E, ICOSLG, HGF, MELK, IGSF6, GNLY, TDO2, KRT7, HLA-E, HLA-DM, LAMP1, NTN3, CD28, TARP, EGFR, CCR4, MAGEA3, BATF, KLRG1, IRS1, CSF1R, CTLA4, TNFSF18, POU2AF1, GZMA, PIK3CA, ITK, IFI27, EOMES, LCN2, CD80, CD83, CXCL13, MTOR, FCER1G, TFRC, RORC, MMP9, BST2, PIK3CD, FCGR2B, TNFRSF14, OAS3, GRAP2, CCNB2, MLANA, MAGEA12, VCAM1, CDKN3, NCR1, FAS, GZMB, IRF9, IFITM2, TNFSF14, HLA-B, SDHA, NRP1, EBI3, EFNA4, PVR, BUB1, SKAP2, PRF1, CCL20, TNFRSF18, CTSS, NKG7, ISG15, PDCD1, SNAI1, CXCL11, CIITA, IFI35, TNFSF9, TNFSF10, MMP2, EGR3, MAGEA1, CD163, IL6, KLRF1, B3GAT1, C1QA, OAS1, IKZF2, TLR9, KLF2, GUSB, NFKBIA, IL23A, HERC6, SLAMF8, IL15, TLR7, OAS2, HLA-DR, CRTAM, MAGEC2, ICAM1, CD4, MAPK14, C1QB, NOTCH3, NCR3, STAT3, TLR8, CYBB, IKZF4, IFIH1, LCK, BCL2L11, ITGAM, ITGB7, JCHAIN, CD209, SLAMF7, IL10, IL1A, FCGR3A, IFNA17, EGR2, TOP2A, C10orf54, FOXM1, AXL, MS4A1, IFI6, CD3D, GPR18, CD3G, ZAP70, HMBS, IL7, IFIT3, RB1, PTGS2, TGFB1, NCF1, TWIST1, CA4, SELL, LILRB1, CD14, ALOX15B, PECAM1, NOS2, FASLG, CD44, ENTPD1, CMKLR1, CD53, TNF, CXCL8, CD40LG, HLA-F, GATA3, LYZ, ARG1, IL2RB, NECTIN2, MPO, CCR2, BRCA2, ADORA2A, G6PD, TAP1, MX1, HLA-DQB2, CD27, CD276, STAT4, PTPN7, PTPRC, PSMB9, CD244, CXCR4, MAPK1, TP63, IRF4, CCL3, CCL18, IL7R, HLA-DRB1, CEACAM8, CXCL10, CCL2, SRGN, CD19, ITGB1, IFITM1, CCL21, MRC1, PGF, ITGAL, ID2, CD22, CCL17, ITGAE, IL3RA, CCR7, CD1C, MAD2L1, PYGL, CD40, LY9, HLA-G, TLR3, CD48, STAT5A, FCRLA, BCL6, ZEB1, CCL5, IDO1, IL18, TNFRSF9, HIF1A, HLA-DPB1, FOXO1, CD33, S100A9, HLA-DMB, HLA-A, SNAI2, TNFRSF17, LRP1, MAGEA4, HLA-DQA1, CD1D, RPS6, MKI67, GZMK, CD79A, CD37, FUT4, AIF1, CCR1, PRDM1, CD47, CD74, LAG3, TNFRSF4, CD2, CCL4, BAGE, LEXM, CCR6, CD70, CDK1, CTAG1B, CTAG2, CX3CR1, CX3CR1, CX3CR1, CX3CR1, GAGE1, GAGE12I, GAGE12F, GAGE12J, GAGE2C, GAGE2A, GAGE2E, GAGE10, GAGE13, IKZF1, IL17A, IL2, IL21, IL22, KIR2DL2, KIR2DL3, MAGEA10, MIF, PTPRCAP, SSX2, TCF7, XAGE1B, CEACAM8, CXCR3, FCGR1A, FCGR3B, FYB, HLA-C, HLA-DQA2, IFNG, KIR2DL1, KRT5, LMNA, and PTPN11. In some embodiments the one or more aberrant expression sequences indicate cancer activity. In some embodiments the one or more aberrant expression sequences indicate a patient's likelihood to response to a therapeutic agent. In some embodiments, the one or more aberrant expression sequences indication a patient's likelihood to not be responsive to a therapeutic agent. In certain embodiments, relevant therapeutic agents can be immunotherapies including but not limited to checkpoint blockades, T cell therapies, and therapeutic vaccines. In some embodiments a therapeutic agent may modify an immune response gene selected from PD1 (e.g., nivolumab, pembrolizumab, AMP-244, MEDI0680, AMP-514, pidlizumab), CTLA4 (e.g., ipilimumab, tremelimumab), PD-L1 (e.g., atezolizumab, MDX1105-01, MEDI4736, avelumab), MR (e.g., lirilumab, NCC0141-0000-0100), CD9/NKG2A (e.g., IPH2201), LAG3 (e.g., BMS986016), GITR (e.g., TRX518), OX40 (e.g., MEDI6383, MEDI6489, MOXR0916), IDO (e.g., indoximod (NLG8189), INCB024360, F001287, NLG919), TGFbeta (e.g., sotaracept, fresolumimab, trabedersen, lucanix), TGFbetaR (e.g., LY2157299, ACE536), CD137 (e.g., urelumab), CD137/41BB (e.g., pf05082566), CD289/TLR9 (e.g., MGN1703), MUC1/CD227 (e.g., OINT-10, ASN-004), CD27 (e.g., varlilumab), CD27L (e.g., AMG172), SLAMF7/CD1 (e.g., elotuzumab), CD20 (e.g., DI-Leu16-IL2), talimogene laherparepvec, CD70 (e.g., ARGX110), IL10 (e.g., AM0010), PSA (e.g., PROSTVAC), GP100 (e.g., MDX1379), STAT3 (e.g., AZD9150), CVAC, IL12 (e.g., veledimex, INXN2001, MSB0010360N, IMMUNOPULSE, GEN-1, INO-9012; IL2: MSB0010445, RG7813/R06895882), IL33 (e.g., alarmin IL33), ICT140, CNDO-109, hTERT (e.g., INO-1400), dysplastic tissues (e.g., ADXS-HPV), SMAC-mimetic (e.g., birinapant), ImmTACs (e.g., IMCgp100), and CD40 (e.g., RO7009789. In certain embodiments, relevant therapeutic agent is an immunotherapy selected from checkpoint blockade, T cell therapy, and therapeutic vaccine. In some embodiments a therapeutic agent modifies an immune response gene selected from any of PD1 (e.g., nivolumab, pembrolizumab, AMP-244, MEDI0680, AMP-514, pidlizumab), CTLA4 (e.g., ipilimumab, tremelimumab), PD-L1 (e.g., atezolizumab, MDX1105-01, MEDI4736, avelumab), MR (e.g., lirilumab, NCC0141-0000-0100), CD9/NKG2A (e.g., IPH2201), LAG3 (e.g., BMS986016), GITR (e.g., TRX518), OX40 (e.g., MEDI6383, MEDI6489, MOXR0916), IDO (e.g., indoximod (NLG8189), INCB024360, F001287, NLG919), TGFbeta (e.g., sotaracept, fresolumimab, trabedersen, lucanix), TGFbetaR (e.g., LY2157299, ACE536), CD137 (e.g., urelumab), CD137/41BB (e.g., pf05082566), CD289/TLR9 (e.g., MGN1703), MUC1/CD227 (e.g., OINT-10, ASN-004), CD27 (e.g., varlilumab), CD27L (e.g., AMG172), SLAMF7/CD1 (e.g., elotuzumab), CD20 (e.g., DI-Leu16-IL2), talimogene laherparepvec, CD70 (e.g., ARGX110), IL10 (e.g., AM0010), PSA (e.g., PROSTVAC), GP100 (e.g., MDX1379), STAT3 (e.g., AZD9150), CVAC, IL12 (e.g., veledimex, INXN2001, MSB0010360N, IMMUNOPULSE, GEN-1, INO-9012; IL2: MSB0010445, RG7813/R06895882), IL33 (e.g., alarmin IL33), ICT140, CNDO-109, hTERT (e.g., INO-1400), dysplastic tissues (e.g., ADXS-HPV), SMAC-mimetic (e.g., birinapant), ImmTACs (e.g., IMCgp100), and CD40 (e.g., RO7009789.

In some embodiments, target sequences or amplified target sequences of the methods are directed to mutations associated with cancer. In some embodiments, the target sequences or amplified target sequences are directed to mutations associated with one or more solid tumor cancers selected from the group consisting of head and neck cancers (e.g., HNSCC, nasopharyngeal, salivary gland), brain cancer (e.g., glioblastoma, glioma, gliosarcoma, glioblastoma multiforme, neuroblastoma), breast cancer (e.g., TNBC, trastuzumab resistant HER2+ breast cancer, ER+/HER− breast cancer), gynecological (e.g., uterine, ovarian cancer, cervical cancer, endometrial cancer, fallopian cancer), colorectal cancer, gallbladder cancer, esophageal cancer, gastrointestinal cancer, gastric cancer, bladder cancer, prostate cancer, testicular cancer, urothelial cancer, liver cancer (e.g., hepatocellular, HCC), lung cancer (e.g., non-small cell lung, small cell lung), kidney (renal cell) cancer, pancreatic cancer (e.g., adenocarcinoma, ductal), thyroid cancer, bile duct cancer, pituitary tumor, wilms tumor, kaposi sarcoma, hairy cell carcinoma, osteosarcoma, thymus cancer, skin cancer, melanoma, heart cancer, oral and larynx cancer, neuroblastoma, mesothelioma, and other solid tumors (thymic, bone, soft tissue, oral SCC, myelofibrosis, synovial sarcoma). In one embodiment, the mutations can include substitutions, insertions, inversions, point mutations, deletions, mismatches and translocations. In one embodiment, the mutations can include variation in copy number. In one embodiment, mutations can include germline or somatic mutations. In some embodiments, the target sequences or amplified target sequences are directed to sequences having mutations associated with one or more blood/hematologic cancers selected from the group consisting of multiple myeloma, diffuse large B cell lymphoma (DLBCL), lymphoma, Hodgkins lymphoma, non-Hodgkins lymphoma, follicular lymphoma, leukemia, acute myeloid leukemia (AML), chronic lymphocytic leukemia (CLL), myelodisplastic syndrome. In one embodiment, mutations associated with cancer are located in at least one of the genes provided in Table 1.

In some embodiments, one or more mutations in immune response sequences associated with cancer are located in at least one of the genes selected from CD63, CD69, CXCL1, KLRD1, HLA-DOB, CXCR5, IL12B, PTK7, CEACAM1, CXCL9, IL13, NT5E, VEGFA, ABCF1, CD38, JAML, S100A8, MYC, IRF1, CCL22, CXCR2, IFIT1, IFIT2, CD68, M6PR, SH2D1A, ISG20, GBP1, TBP, STAT6, ID3, CX3CL1, KLRB1, TNFSF4, CD52, IL10RA, HLA-DOA, IFNB1, CCR5, IKZF3, STAT1, CD6, BRCA1, CORO1A, TBX21, KLRK1, CXCR6, PTEN, PMEL, DMBT1, IFI44L, LAPTM5, CD226, TNFSF13B, ICOS, CD160, TRIM29, LST1, ZBTB46, VTCN1, KREMEN1, PDCD1LG2, TUBB, CLEC4C, CD86, HAVCR2, GZMH, NFATC1, CD8B, BCL2, GADD45GIP1, CBLB, ITGA1, CD8A, IL2RA, EIF2AK2, MADCAM1, PTPN6, LRG1, ADGRE5, SH2D1B, ITGB2, HLA-DPA1, DGAT2, IGF1R, TAGAP, LMNA, NCAM1, TIGIT, IL17F, HLA-F-AS1, CD247, CD79B, IDO2, IL4, TYROBP, BTLA, AKT1, IL2RG, POLR2A, ITGAX, IL1B, CSF2RB, DDX58, KIAA0101, CD274, LAMP3, TNFAIP8, FOXP3, IL12A, SAMHD1, SIT1, CD3E, ICOSLG, HGF, MELK, IGSF6, GNLY, TDO2, KRT7, HLA-E, HLA-DM, LAMP1, NTN3, CD28, TARP, EGFR, CCR4, MAGEA3, BATF, KLRG1, IRS1, CSF1R, CTLA4, TNFSF18, POU2AF1, GZMA, PIK3CA, ITK, IFI27, EOMES, LCN2, CD80, CD83, CXCL13, MTOR, FCER1G, TFRC, RORC, MMP9, BST2, PIK3CD, FCGR2B, TNFRSF14, OAS3, GRAP2, CCNB2, MLANA, MAGEA12, VCAM1, CDKN3, NCR1, FAS, GZMB, IRF9, IFITM2, TNFSF14, HLA-B, SDHA, NRP1, EBI3, EFNA4, PVR, BUB1, SKAP2, PRF1, CCL20, TNFRSF18, CTSS, NKG7, ISG15, PDCD1, SNAI1, CXCL11, CIITA, IFI35, TNFSF9, TNFSF10, MMP2, EGR3, MAGEA1, CD163, IL6, KLRF1, B3GAT1, C1QA, OAS1, IKZF2, TLR9, KLF2, GUSB, NFKBIA, IL23A, HERC6, SLAMF8, IL15, TLR7, OAS2, HLA-DR, CRTAM, MAGEC2, ICAM1, CD4, MAPK14, C1QB, NOTCH3, NCR3, STAT3, TLR8, CYBB, IKZF4, IFIH1, LCK, BCL2L11, ITGAM, ITGB7, JCHAIN, CD209, SLAMF7, IL10, IL1A, FCGR3A, IFNA17, EGR2, TOP2A, C10orf54, FOXM1, AXL, MS4A1, IFI6, CD3D, GPR18, CD3G, ZAP70, HMBS, IL7, IFIT3, RB1, PTGS2, TGFB1, NCF1, TWIST1, CA4, SELL, LILRB1, CD14, ALOX15B, PECAM1, NOS2, FASLG, CD44, ENTPD1, CMKLR1, CD53, TNF, CXCL8, CD40LG, HLA-F, GATA3, LYZ, ARG1, IL2RB, NECTIN2, MPO, CCR2, BRCA2, ADORA2A, G6PD, TAP1, MX1, HLA-DQB2, CD27, CD276, STAT4, PTPN7, PTPRC, PSMB9, CD244, CXCR4, MAPK1, TP63, IRF4, CCL3, CCL18, IL7R, HLA-DRB1, CEACAM8, CXCL10, CCL2, SRGN, CD19, ITGB1, IFITM1, CCL21, MRC1, PGF, ITGAL, ID2, CD22, CCL17, ITGAE, IL3RA, CCR7, CD1C, MAD2L1, PYGL, CD40, LY9, HLA-G, TLR3, CD48, STAT5A, FCRLA, BCL6, ZEB1, CCL5, ID01, IL18, TNFRSF9, HIF1A, HLA-DPB1, FOXO1, CD33, S100A9, HLA-DMB, HLA-A, SNAI2, TNFRSF17, LRP1, MAGEA4, HLA-DQA1, CD1D, RPS6, MKI67, GZMK, CD79A, CD37, FUT4, AIF1, CCR1, PRDM1, CD47, CD74, LAG3, TNFRSF4, CD2, CCL4, BAGE, LEXM, CCR6, CD70, CDK1, CTAG1B, CTAG2, CX3CR1, CX3CR1, CX3CR1, CX3CR1, GAGE1, GAGE12I, GAGE12F, GAGE12J, GAGE2C, GAGE2A, GAGE2E, GAGE10, GAGE13, IKZF1, IL17A, IL2, IL21, IL22, KIR2DL2, KIR2DL3, MAGEA10, MIF, PTPRCAP, SSX2, TCF7, XAGE1B, CEACAM8, CXCR3, FCGR1A, FCGR3B, FYB, HLA-C, HLA-DQA2, IFNG, KIR2DL1, KRT5, LMNA, and PTPN11.

In one aspect, compositions and methods of the invention are used to identify biomarkers of response, sensitivity and/or non-response. In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes between samples derived from a patients responding to treatment of immunotherapy and samples derived from non-responding patients. Any one or more gene(s) that demonstrate significantly different expression levels (e.g., high in responders group, low in non-responders group) is considered biomarker(s) predictive of drug response. In certain embodiments, such biomarkers of response are used for stratification of patients for immune response therapy. In some embodiments, provided compositions and methods are used to identify biomarkers of immune response monitoring after treatment. In certain embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with immunohistochemistry readouts of an established clinical makers (e.g., PD-L1, NY-ESO-1, MAGE). In certain embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with changes in any therapeutic target, pharmacodynamics markers, or immunogenic antigen. Any genes or combination of genes that show correlated up or down expression with these readouts could be considered as predictive markers of drug response. In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with samples containing high level of tumor infiltrating lymphocytes (TIL) and samples containing low level of TIL, as reported by pathology review. Any genes or combination of genes significantly differ in expression between these two groups could be considered predictors of drug response. In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with samples of different molecular subtypes (e.g., examples for colon-rectal cancer include but are not limited to microsatellite instability immune; hypermutated, microsatellite unstable and strong immune activation; epithelial and evident metabolic dysregulation; mesenchymal [http://www.nature.com/nm/journal/vaop/ncurrent/full/nm.3967.html]). Any genes or combination of genes significantly differ in expression between any of these groups could be considered predictors of drug response. In certain embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with samples of different histopathology subtypes (e.g., examples for non-small cell lung include but are not limited to adenocarcinoma, squamous cell carcinoma, not-otherwise specified; examples for colon rectal). Any genes or combination of genes significantly differ in expression between these two groups could be considered predictors of drug response. In certain embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the T cell signaling pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response.

In certain embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the T cell regulation pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response. In certain embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as antigen processing and/or antigen presentation between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the cytokine signaling pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as type I or type II interferon pathways between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response. In certain embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as lymphocyte development and/or lymphocyte migration between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response. In certain embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the immune checkpoint pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers that are predictive of drug response. In certain embodiments, provided compositions and methods are used to evaluate the expression level of any genes contained on the panel or combination thereof (e.g., PD-1, PD-L1, CTLA4, etc.) between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers of drug response. In certain embodiments, provided compositions and methods are used to evaluate the expression level of genes or isoforms of antigens (e.g., cancer testis antigens (GAGE), melanoma antigens (MAGE)) between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers of drug response. In some embodiments, provided compositions and methods are used to evaluate different mathematical, quantitative or qualitative transformations of raw read count or normalized read count of any genes contained on the panel or combination thereof (for example, PD-1, PD-L1, CTLA4, etc.) between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. Any genes that show significantly different expression levels (high in one group, low in the other) could be considered potential biomarkers of drug response. In certain embodiments, provided compositions and methods are used to profile the inflammatory level and abundance of different immune subsets (for example: cytotoxic T cells, helper T cells, macrophages, neutrophils, natural killer cells, myeloid derived suppressor cells) in the tumor micro-environment. Any differences in expression levels of these markers between samples derived responders and non-responders of treatment could be considered biomarkers predictive of response. In certain embodiments, provided compositions and methods are used to determine HLA type and its correlation with antigen presentation, T cell and B cell activation and drug response. In certain embodiments, provided compositions and methods are used to classify tumor samples as likely responder or non-responders to a therapeutic agent based on a classifier defined by another assay. In particular embodiments, provided compositions and methods are used to identify a subset of genes from which another panel can be derived to be used for purposes described herein. In a particular embodiment, additional primer sets could be added to the current panel to create an additional panel which can be used for purposes described herein.

In another aspect, provided compositions and methods are used to evaluate the expression levels of genes up or down regulated by immune checkpoint modulators. See, e.g., https://en.wikipedia.org/wiki/Immune_checkpoint. The identity of one or more gene(s) modulated by any specific therapeutic agent could be used to elucidate the mechanism of action of that agent. In some embodiments, provided compositions and methods are used to evaluate the expression levels of genes involved in the immune checkpoint pathway (including, e.g., PD-L1 signaling pathway). The identities of one or more genes modulated by any specific therapeutic agent could be used to elucidate the mechanism of action of that agent. In certain embodiments, provided compositions and methods are used to evaluate the expression levels of genes involved chemokine signaling, cytokine signaling, and/or interferon signaling. The identities of one or more genes modulated by any specific therapeutic agent can be used to elucidate the mechanism of action of that agent. In certain embodiments, provided compositions and methods are used to evaluate the expression levels of genes involved tumor cell adhesion, tumor cell migration, tumor cell proliferation or cancer stemness. The pattern of expression associating with each therapeutic agent could be used to infer the mechanism of action of that agent. In particular embodiments, provided compositions and methods are used to detect post-treatment changes in expression of any one or more genes in the panel. The high or low expression levels of such genes would be used to elucidate the mechanism of action of treated agent. In some embodiments, provided compositions and methods are used to detect post-treatment changes in expression of any intracellular or extracellular protein encoding genes contained on the panel. The high or low expression levels of such genes could be used to elucidate the mechanism of action of that agent. In some embodiments, provided compositions and methods are used to study the changed expression of intracellular protein-encoding genes in response to treatment of therapeutic agent or agents modulating extracellular proteins. High or low expression levels of such genes could be used to elucidate the mechanism of action of such agents. In still additional embodiments, provided compositions and methods are used to study the expression of any genes contained on the panel at different time points or treatment conditions. High or low expression levels of such genes could be used to elucidate the mechanism of action of that agent. In particular embodiments, provided compositions and methods are used to study the expression of any genes contained on the panel at different time points or treatment conditions with different therapeutic agents. The patterns of expression that are considered complementary could provide rationales to combine the corresponding agents for greater benefit. In particular embodiments, provided compositions and methods are used to study the expression of any genes contained on the panel at different time points or treatment conditions with different therapeutic agents. Patterns of expression that are considered complementary could provide rationales to combine the corresponding agents for reduced risk of adverse events. In certain embodiments, provided compositions and methods are used to study the expression of any genes modulated by any radiotherapy, chemotherapy, targeted therapy or immunotherapy. Complementary patterns of expression complementary could provide rationales to combine the corresponding agents for greater benefit or reduced risk of adverse events. In still other embodiments, provided compositions and methods are used to study expression of any genes modulated by a T cell therapy and immunosuppressive therapy. Complementary patterns of expression could provide rationales to combine the corresponding agents for greater benefit or reduced risk of adverse events.

In some embodiments, amplified target sequences of the methods are directed to any one of more of the genes provided in Table 1. In some embodiments, amplified target sequences of the methods comprise any one or more amplicon sequences provided in Table 1. In some embodiments, amplified target sequences of the methods consist of any one or more amplicon sequences provided in Table 1. In some embodiments, amplified target sequences of the methods include each amplicon sequence provided in Table 1.

In some embodiments, compositions used in provided methods comprise any one or more of the immune response target-specific primer pairs provided in Table 2. In some embodiments, compositions used in connection with provided methods comprise all of the immune response target-specific primer pairs provided in Table 2. In some embodiments, any one or more of the immune response target-specific primer pairs provided in Table 2 can be used to amplify a target sequence present in a sample as disclosed by the methods described herein.

In some embodiments, the methods utilize immune response target-specific primers from Table 2 and include 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 40, 60, 80, 100, 150, 200, 250, 300, 350, 398 or more, target-specific primer pairs. In some embodiments, methods include targeting amplified target sequences including one or more of the amplified target sequences generated in Table 1 (e.g., using amplicon target-specific primers provided in Table 2). In some embodiments, methods include use of at least one target-specific primer pair wherein each primer is at least 90% identical to at least one nucleic acid sequence selected from SEQ ID NOs: 399-1194. In some embodiments, methods include use of at least one of the target-specific primers associated with immune response is complementary across its entire length to at least one target sequence in a sample. In some embodiments, methods include use of at least one of the target-specific primers associated with immune response includes a non-cleavable nucleotide at the 3′ end. In some embodiments, methods include use of target specific primers wherein non-cleavable nucleotide at the 3′ end includes the terminal 3′ nucleotide. In one embodiment, methods include use of amplified target sequences directed to one or more individual exons having aberrant expression associated with cancer. In one embodiment, methods include use of amplified target sequences directed to individual exons having a mutation associated with cancer. In some embodiments, provided methods comprise selective amplification of more than one target sequences in a sample and the detection and/or identification of mutations associated with cancer. In some embodiments, methods include use of amplified target sequences including two or more nucleotide sequences provided in Table 2. In some embodiments, methods include use of amplified target sequences including any one or more the amplified target sequences generated using the target-specific primers provided in Table 2. In one embodiment, methods include use of amplified target sequences including 10, 50, 100, 150, 200, 250, 300, 350 or more amplicons from Table 1. In some embodiments, methods comprise detection and optionally, the identification of clinically actionable marker(s). As defined herein, the term “clinically actionable marker” includes clinically actionable mutations and/or clinically actionable expression patterns that are known or can be associated by one of ordinary skill in the art with, but not limited to, prognosis for the treatment of cancer. In one embodiment, prognosis for the treatment of cancer includes the identification of mutations and/or expression patterns associated with responsiveness or non-responsiveness of a cancer to a drug, drug combination, or treatment regime. In one embodiment, methods comprise amplification of a plurality of target sequences from a population of nucleic acid molecules linked to, or correlated with, the onset, progression or remission of cancer.

In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes between samples derived from patients with good prognosis (defined by durable response, overall survival or 5-year survival) and those with poor prognosis. For example, gene(s) that show significantly different expression levels (e.g., high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with samples containing high level of tumor infiltrating lymphocytes (TIL) and samples containing low level of TIL, as reported by pathology review. For example, gene(s) or combination of genes significantly differ in expression between these two groups could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with samples of different histopathology subtypes molecular subtypes (examples for breast cancer include luminal A, luminal B, basal-like, HER2 positive). For example, gene(s) or combination of genes that significantly differ in expression between any of these groups could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to compare absolute or relative gene expression changes correlating with samples of different histopathology subtypes (e.g., for breast cancer include, for example, ER+, PR+, HER2+, triple negative). For example, gene(s) or combination of genes that significantly differ in expression between these two groups could be considered prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the T cell signaling pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (e.g., high in one group, low in another) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate gene expression levels of different pathways and functions such as the T cell regulation pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate gene expression levels of different pathways and functions such as the expression of genes in antigen processing and/or antigen presentation between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the expression of genes in cytokine signaling pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as type I or type II interferon pathways between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as lymphocyte development and migration between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate absolute or relative gene expression levels of different pathways and functions such as the immune checkpoint pathway between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate the expression level of any genes contained on the panel or combination thereof (e.g., for example, PD-1, PD-L1, CTLA4, etc.) between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to evaluate different mathematical, quantitative or qualitative transformations of raw read count or normalized read count of any genes contained on the panel or combination thereof (e.g., for example, PD-1, PD-L1, CTLA4, etc.) between samples derived from patients responding to treatment of immunotherapy and samples derived from non-responders. For example, gene(s) that show significantly different expression levels (high in one group, low in the other) could be considered potential prognostic biomarkers. In some embodiments, provided compositions and methods are used to profile the inflammatory level and presence of different immune subsets (for example: cytotoxic T cells, helper T cells, macrophages, neutrophils, natural killer cells, myeloid derived suppressor cells) in the tumor micro-environment. Any differences in expression levels of these markers between samples derived responder and non-responder of treatment could be considered potential prognostic biomarkers.

In some embodiments, housekeeping genes are utilized in conjunction with provided methods in the immune response assay. In certain embodiments, one or more housekeeping gene sequences are included in the assay methods. In particular method embodiments the one or more housekeeping genes are selected from ABCF1, G6PD, GUSB, HMBS, LMNA, LRP1, POLR2A, SDHA, TBP, TFRC, and TUBB.

In some embodiments, the disclosure provides for amplification of multiple target-specific sequences from a population of expressed nucleic acid molecules. In some embodiments, the method comprises hybridizing one or more target-specific primer pairs to the target sequence, extending a first primer of the primer pair, denaturing the extended first primer product from the population of nucleic acid molecules, hybridizing to the extended first primer product the second primer of the primer pair, extending the second primer to form a double stranded product, and digesting the target-specific primer pair away from the double stranded product to generate a plurality of amplified target sequences. In some embodiments, the digesting includes partial digesting of one or more of the target-specific primers from the amplified target sequence. In some embodiments, the amplified target sequences can be ligated to one or more adapters. In some embodiments, adapters can include one or more DNA barcodes or tagging sequences. In some embodiments, amplified target sequences once ligated to an adapter can undergo a nick translation reaction and/or further amplification to generate a library of adapter-ligated amplified target sequences.

In some embodiments, provided methods comprise preparation and formation of multiple immune response target-specific amplicons. In some embodiments, the method comprises hybridizing one or more target-specific primer pairs to a nucleic acid molecule, extending a first primer of the primer, pair, denaturing the extended first primer from the nucleic acid molecule, hybridizing to the extended first primer product, a second primer of the primer pair and extending the second primer, digesting the target-specific primer pairs to generate a plurality of target-specific amplicons. In some embodiments, adapters can be ligated to the ends of the target-specific amplicons prior to performing a nick translation reaction to generate a plurality of target-specific amplicons suitable for nucleic acid sequencing. In some embodiments, the one or more target specific amplicons can be amplified using bridge amplification or emPCR to generate a plurality of clonal templates suitable for nucleic acid sequencing. In some embodiments, the disclosure provides methods for preparing a target-specific amplicon library, for use in a variety of downstream processes or assays such as nucleic acid sequencing or clonal amplification. In one embodiment, the disclosure provides a method of performing target-specific multiplex PCR on a nucleic acid sample having a plurality of expressed target sequences using primers having a cleavable group.

In certain embodiments, library and/or template preparation to be sequenced can be prepared automatically from a population of nucleic acid samples using the compositions provided herein using an automated systems, e.g., the Ion Chef™ system.

In one embodiment, nucleic acid templates to be sequenced using the Ion Torrent PGM 318™ or Ion Torrent S5 520™ or Ion Torrent S5 530™ system can be prepared from a population of nucleic acid molecules using the target-specific amplification techniques as outlined herein. Optionally, following target-specific amplification a secondary and/or tertiary amplification process including, but not limited to, a library amplification step and/or a clonal amplification step such as emPCR can be performed.

In some embodiments, provided are compositions comprising a plurality of target-specific primer pairs, each containing a forward primer and a reverse primer having at least one cleavable group located at either a) the 3′ end or the 5′ end, and/or b) at about the central nucleotide position of the target-specific primer, and wherein the target-specific primer pairs can be substantially non-complementary to other primer pairs in the composition. In some embodiments, the composition comprises at least 50, 100, 150, 200, 250, 300, 350, 398, or more target-specific primer pairs. In some embodiments, the target-specific primer pairs comprise about 15 nucleotides to about 40 nucleotides in length, wherein at least one nucleotide is replaced with a cleavable group. In some embodiments the cleavable group can be a uridine nucleotide. In some embodiments, the target-specific primer sets are designed to amplify an exon, gene, exome or region of the genome associated with a clinical or pathological condition, e.g., the amplification of one or more expressed sequences associated with cancer, wherein an increase and/or decrease in expression is associated with a change in immune response. In some embodiments, the target-specific primer sets are designed to amplify an exon, gene, exome or region of the genome associated with a change in immune response wherein an increase and/or decrease in expression is indicative of a cancer patient's likelihood to respond to one or more therapeutic agent(s). In some embodiments, the target-specific primer sets are designed to amplify an exon, gene, exome or region of the genome associated with a clinical or pathological condition, e.g., the amplification of one or more mutations associated with cancer. In some embodiments, the target-specific primer pairs when hybridized to a target sequence and amplified as outlined herein can generate a library of adapter-ligated amplified target sequences that are about 100 to about 500 base pairs in length. In some embodiments, no one adapter-ligated amplified target sequence is overexpressed in the library by more than 30% as compared to the remainder of the adapter-ligated amplified target sequences in the library. In some embodiments, the adapter-ligated amplified target sequence library is substantially homogenous with respect to GC content, amplified target sequence length or melting temperature (Tm).

In some embodiments, a kit is provided for performing multiplex PCR comprising a plurality of target-specific primers having a cleavable group, a DNA polymerase, an adapter, dATP, dCTP, dGTP and dTTP. In some embodiments, the cleavable group can be a uracil nucleotide. The kit can further include one or more antibodies, nucleic acid barcodes, purification solutions or columns.

In some embodiments, provided is a kit for generating a target-specific amplicon library comprising a plurality of target-specific primers having a cleavable group, a DNA polymerase, an adapter, dATP, dCTP, dGTP, dTTP, and a cleaving reagent. In some embodiments, the kit further comprises one or more antibodies, nucleic acid barcodes, purification solutions or columns.

In one embodiment, methods are provided for amplification of multiple target-specific sequences from a single nucleic acid source or sample. In another embodiment, methods are provided for target-specific amplification of two or more target sequences from two or more nucleic acid sources, samples or species. For example, it is envisioned by the disclosure that a single nucleic acid sample can include expressed RNA or fixed-formalin paraffin-embedded (FFPE) RNA. It is also envisioned that the sample can be from a single individual, a collection of nucleic acid samples from genetically related members, multiple nucleic acid samples from genetically unrelated members, multiple nucleic acid samples (matched) from a single individual such as a tumor sample and normal tissue sample, or genetic material from a single source that contains two distinct forms of genetic material such as maternal and fetal RNA obtained from a maternal subject, or the presence of contaminating bacteria RNA in a sample that contains plant or animal nucleic acid. In some embodiments, the source of nucleic acid material can include nucleic acids obtained from a newborn, for example as typically procured as a blood sample for newborn screening.

The nucleic acid sample can include high molecular weight material such as expressed RNA or cDNA. The sample can include low molecular weight material such as nucleic acid molecules obtained from FFPE or archived RNA samples. In another embodiment, low molecular weight material includes enzymatically or mechanically sheared nucleic acid sample. The sample can include cell-free circulating RNA such as material obtained from a maternal subject. In some embodiments, the sample can include nucleic acid molecules obtained from biopsies, tumors, scrapings, swabs, blood, mucus, urine, plasma, semen, hair, laser capture micro-dissections, surgical resections, and other clinical or laboratory obtained samples. In some embodiments, the sample can include nucleic acid molecules obtained from an animal such as a human or mammalian source. In another embodiment, the sample can include nucleic acid molecules obtained from a non-mammalian source such as a plant, bacteria, virus or fungus. In some embodiments, the source of the nucleic acid molecules may be an archived or extinct sample or species.

In some embodiments, provided methods comprise selective amplification of at least one target sequence in a normal or diseased containing tissue, biopsy, core, tumor, fine needle aspirate, fine needle biopsy, tumor microenvironment, blood, serum, or other sample. In some embodiments, provided methods comprise selective amplification of at least one target sequence and the detection and/or identification of mutations in the diseased tissue, biopsy, core, tumor, fine needle aspirate, fine needle biopsy, tumor microenvironment, blood, serum, or other sample. In some embodiments, the diseased or normal sample can include whole transcriptomic RNA, formalin-fixed paraffin-embedded tissue (FFPE), sheared or enzymatically treated RNA. In some embodiments, the disclosure is directed to the selective amplification of at least one target sequence and detection and/or identification of clinically actionable mutations or expression patterns. In some embodiments, the disclosure is directed to the detection and/or identification of mutations or expression associated with drug resistance or drug susceptibility.

In some embodiments, provided methods comprise selective amplification of at least one target sequence in cell-free circulating RNA. In some embodiments, the selective amplification of at least one target sequence in a sample includes a mixture of different nucleic acid molecules. The selective amplification can optionally be accompanied by detection and/or identification of mutations observed in circulating RNA. In some embodiments, selective amplification can optionally be accompanied by detection and/or identification of mutations associated with cancer or an inherited disease such as metabolic, neuromuscular, developmental, cardiovascular, autoimmune or other inherited disorder.

In some embodiments, the target-specific primers and primer pairs are target-specific sequences that can amplify specific regions of a nucleic acid molecule. In some embodiments, the target-specific primers can amplify expressed RNA or cDNA. In some embodiments, the target-specific primers can amplify mammalian RNA, such as human RNA or cDNA prepared therefrom. In some embodiments, the amount of RNA required for selective amplification can be from about 1 ng to 1 microgram. In some embodiments, the amount of RNA required for selective amplification of one or more target sequences can be about 1 ng, about 5 ng or about 10 ng. In some embodiments, the amount of RNA required for selective amplification of target sequence is about 10 ng to about ng.

In some embodiments, selective amplification of at least one target sequence further includes nucleic acid sequencing of the amplified target sequence. Optionally, the method further includes detecting and/or identifying mutations present in the sample identified through nucleic acid sequencing of the amplified target sequence.

In one embodiment, a sample containing one or more target sequences can be amplified using any one or more of the target-specific primers disclosed herein. In another embodiment, amplified target sequences obtained using the methods (and associated compositions, systems, apparatuses and kits) disclosed herein, can be coupled to a downstream process, such as but not limited to, nucleic acid sequencing. For example, once the nucleic acid sequence of an amplified target sequence is known, the nucleic acid sequence can be compared to one or more reference samples such as Hg19 genome. The Hg19 genome is commonly used in the genomics field as a reference genome sample for humans.

It is envisaged that one of ordinary skill in the art can readily prepare one or more target-specific primers using the primer criteria disclosed herein without undue experimentation. It is also envisaged that one of ordinary skill in the art can readily prepare one or more target-specific primers using the criteria disclosed herein, to identify at least one medically relevant polymorphism. In some instances, a medically relevant polymorphism can be used in forensic or human identification purposes. A medically relevant mutation includes a mutation that is associated with at least one disease state in multiple populations (e.g., a European Caucasian population). In some embodiments, a medically relevant polymorphism includes any one or more of the polymorphisms outlined below.

In some embodiments, provided are methods (and associated compositions, systems, apparatuses and kits) for reducing the formation of amplification artifacts in a multiplex PCR. In some embodiments, primer-dimers or non-specific amplification products are obtained in lower number or yield as compared to standard multiplex PCR of the prior art. In some embodiments, the reduction in amplification artifacts is in part, governed by the use of target-specific primer pairs in the multiplex PCR reaction. In one embodiment, the number of target-specific primer pairs in the multiplex PCR reaction can be greater than 50, 100, 150, 200, 250, 300 or more. In some embodiments, provided are methods (and associated compositions, systems, apparatuses and kits) for performing multiplex PCR using target-specific primers that contain a cleavable group. In one embodiment, target-specific primers containing a cleavable group can include one or more cleavable moieties per primer of each primer pair. In some embodiments, a target-specific primer containing a cleavable group includes a nucleotide neither normally present in a non-diseased sample nor native to the population of nucleic acids undergoing multiplex PCR. For example, a target-specific primer can include one or more non-native nucleic acid molecules such as, but not limited to thymine dimers, 8-oxo-2′-deoxyguanosine, inosine, deoxyuridine, bromodeoxyuridine, apurinic nucleotides, and the like.

In some embodiments, provided methods (and associated compositions, systems, etc.,) involve performing a primary amplification of target sequences from a population of nucleic acids, optionally using target-specific primers. In some embodiments, provided methods involve amplifying target sequences using target-specific forward and reverse primer pairs. The target-specific forward and reverse primer pairs can optionally include one or more intron-specific and/or exon specific forward and reverse primer pairs. In some embodiments, each primer pair is directed to a single or discrete exon. In some embodiments, provided methods involve amplifying target sequences using exon-specific forward and reverse primer pairs containing at least one cleavable group. In some embodiments, the target-specific forward and reverse primer pairs contain a uracil nucleotide as the one or more cleavable groups. In one embodiment, a target-specific primer pair can include a uracil nucleotide in each of the forward and reverse primers of each primer pair. In one embodiment, a target-specific forward or reverse primer contains one, two, three or more uracil nucleotides. In some embodiments, provided methods involve amplifying at least 10, 50, 100, 150, 200, 250, 300, 350, 398 or more, target sequences from a population of nucleic acids having a plurality of target sequences using target-specific forward and reverse primer pairs containing at least two uracil nucleotides.

In some embodiments, target-specific primers (including but not limited to intron-specific and exon-specific primers, which can be forward and/or reverse primers) can be designed de novo using algorithms that generate oligonucleotide sequences according to specified design criteria. For example, the primers may be selected according to any one or more of criteria specified herein. In some embodiments, one or more of the target-specific primers are selected or designed to satisfy any one or more of the following criteria: (1) inclusion of two or more modified nucleotides within the primer sequence, at least one of which is included near or at the termini of the primer and at least one of which is included at, or about the center nucleotide position of the primer sequence; (2) primer length of about 15 to about 40 bases in length; (3) Tm of from about 60° C. to about 70° C.; (4) low cross-reactivity with non-target sequences present in the target genome or sample of interest; (5) for each primer in a given reaction, the sequence of at least the first four nucleotides (going from 3′ to 5′ direction) are not complementary to any sequence within any other primer present in the same reaction; and (6) no amplicon includes any consecutive stretch of at least 5 nucleotides that is complementary to any sequence within any other amplicon.

In some embodiments, the target-specific primers include one or more primer pairs designed to amplify target sequences from the sample that are about 100 base pairs to about 500 base pairs in length. In some embodiments, the target-specific primers include a plurality of primer pairs designed to amplify target sequences, where the amplified target sequences are predicted to vary in length from each other by no more than 50%, typically no more than 25%, even more typically by no more than 10%, or 5%. For example, if one target-specific primer pair is selected or predicted to amplify a product that is 100 nucleotides in length, then other primer pairs are selected or predicted to amplify products that are between 50-150 nucleotides in length, typically between 75-125 nucleotides in length, even more typically between 90-110 nucleotides, or 95-105 nucleotides, or 99-nucleotides in length.

In some embodiments, at least one primer pair in the amplification reaction is not designed de novo according to any predetermined selection criteria. For example, at least one primer pair can be an oligonucleotide sequence selected or generated at random, or previously selected or generated for other applications. In one exemplary embodiment, the amplification reaction can include at least one primer pair selected from the TaqMan® probe reagents (Roche Molecular Systems). The TaqMan® reagents include labeled probes and can be useful, inter alia, for measuring the amount of target sequence present in the sample, optionally in real time. Some examples of TaqMan technology are disclosed in U.S. Pat. Nos. 5,210,015, 5,487,972, 5,804,375, 6,214,979, 7,141,377 and 7,445,900, hereby incorporated by reference in their entireties.

In some embodiments, at least one primer within the amplification reaction can be labeled, for example with an optically detectable label, to facilitate a particular application of interest. For example, labeling may facilitate quantification of target template and/or amplification product, isolation of the target template and/or amplification, product, and the like.

In some embodiments, one or more of the primers within the amplification reaction can be useful in genotyping of a nucleic acid sample.

In some embodiments, the target-specific primers can be provided as a set of target-specific primer pairs in a single amplification vessel. In some embodiments, the target-specific primers can be provided in one or more aliquots of target-specific primer pairs that can be pooled prior to performing the multiplex PCR reaction in a single amplification vessel or reaction chamber. In one embodiment, the target-specific primers can be provided as a pool of target-specific forward primers and a separate pool of target-specific reverse primers. In another embodiment, target-specific primer pairs can be pooled into subsets such as non-overlapping target-specific primer pairs. In some embodiments, the pool of target-specific primer pairs can be provided in a single reaction chamber or microwell, for example on a PCR plate to perform multiplex PCR using a thermocycler. In some embodiments, the target-specific forward and reverse primer pairs can be substantially complementary to the target sequences.

In some embodiments, the method of performing multiplex PCR amplification includes contacting a plurality of target-specific primer pairs having a forward and reverse primer, with a population of target sequences to form a plurality of template/primer duplexes; adding a DNA polymerase and a mixture of dNTPs to the plurality of template/primer duplexes for sufficient time and at sufficient temperature to extend either (or both) the forward or reverse primer in each target-specific primer pair via template-dependent synthesis thereby generating a plurality of extended primer product/template duplexes; denaturing the extended primer product/template duplexes; annealing to the extended primer product the complementary primer from the target-specific primer pair; and extending the annealed primer in the presence of a DNA polymerase and dNTPs to form a plurality of target-specific double-stranded nucleic acid molecules. In some embodiments, the steps of the amplification PCR method can be performed in any order. In some instances, the methods disclosed herein can be further optimized to remove one or more steps and still obtain sufficient amplified target sequences to be used in a variety of downstream processes. For example, the number of purification or clean-up steps can be modified to include more or less steps than disclose herein, providing the amplified target sequences are generated in sufficient yield.

In some embodiments, the target-specific primer pairs do not contain a common extension (tail) at the 3′ or 5′ end of the primer. In another embodiment, the target-specific primers do not contain a Tag or universal sequence. In some embodiments, the target-specific primer pairs are designed to eliminate or reduce interactions that promote the formation of non-specific amplification.

In one embodiment, the target-specific primer pairs comprise at least one cleavable group per forward and reverse target-specific primer. In one embodiment, the cleavable group can be a uracil nucleotide. In one embodiment, the target-specific primer pairs are partially or substantially removed after generation of the amplified target sequence. In one embodiment, the removal can include enzymatic, heat or alkali treatment of the target-specific primer pairs as part of the amplified target sequence. In some embodiments, the amplified target sequences are further treated to form blunt-ended amplification products, referred to herein as, blunt-ended amplified target sequences.

There is a need for new methods, computer readable media, and systems for identifying or designing products or kits that use PCR to enrich one or more genomic regions of interest (which may be, for example, cumulative regions of 1 kb to 1 Mb) for subsequent sequencing.

There is a need for new methods, computer readable media, and systems for identifying or designing products or kits including primers or assays that maximize coverage of one or more genomic regions or targets of interest while minimizing one or more of off-target hybridization, a number of primers, and a number of primer pools.

In accordance with the teachings and principles embodied in this application, new methods, computer readable media, and systems are provided that identify or design products or kits that use PCR to enrich one or more genomic regions or targets of interest for subsequent sequencing and/or that include primers or assays that maximize coverage of one or more genomic regions or targets of interest while minimizing one or more of off-target hybridization, a number of primers, and a number of primer pools.

According to an exemplary embodiment, there is provided a method comprising: (1) receiving or providing as inputs a genomic target region and a set of candidate amplicons for the genomic target region; (2) generating a graph comprising a source vertex, a set of amplicon vertices arranged in correspondence with the set of candidate amplicons, and a sink vertex; (3) determining a cost associated with one or more paths across the graph from the source vertex to the sink vertex via amplicon vertices; and (4) extracting the amplicon vertices from the one of the one or more paths across the graph having a least cost associated therewith.

In various embodiments, the one or more paths may comprise a sequence of amplicons wherein an ending portion of an insert of a first amplicon in the sequence of amplicons overlaps a beginning portion of an insert of a second amplicon in the sequence of amplicons. An ending portion of an insert of the second amplicon in the sequence of amplicons may overlap a beginning portion of an insert of a third amplicon in the sequence of amplicons. An ending portion of an insert of the third amplicon in the sequence of amplicons may overlap a beginning portion of an insert of a fourth amplicon in the sequence of amplicons.

In various embodiments, the one or more paths may comprise a sequence of N amplicons, N being a positive integer, wherein an ending portion of an insert of an amplicon amp in the sequence of amplicons overlaps a beginning portion of an insert of an amplicon amp+1 in the sequence of amplicons, wherein amp is an integer taking values 1, . . . , N−1. The one or more paths may comprise a sequence of L=N+M amplicons, N and M being positive integers, wherein an ending portion of an insert of an amplicon amp in the sequence of amplicons overlaps (which may include merely touching) a beginning portion of an insert of an amplicon amp+1 in the sequence of amplicons where amp is an integer taking values 1, . . . , N−1; wherein an ending portion of an insert of an amplicon amp in the sequence of amplicons overlaps (which may include merely touching) a beginning portion of an insert of an amplicon amp+1 in the sequence of amplicons where amp is an integer taking values N+1, . . . , N+M−1; and wherein there is a gap between an ending portion of an insert of amplicon amp=N and a beginning portion of an insert of amplicon amp=N+1.

In various embodiments, the cost associated with each of the one or more paths may be a sum of the cost of every edge of the path linking two amplicon vertices. The cost associated with every edge of the path linking two amplicon vertices may be a sum of a first term related to the cost of the edge's destination amplicon vertex and a second term related to the cost of an overlap between an insert of the edge's destination amplicon and an insert of the edge's origin amplicon. The first term and the second term may be weighed by a blending factor such that the first term is multiplied by the blending factor or a function thereof and the second term is multiplied by one minus the blending factor or a function thereof. The cost of an amplicon vertex may be a numerical value along a scale between a first value representing a lower level of one or more undesirable characteristics selected from a group comprising at least a level of off-target amplification and a level of primer-dimer propensity and a second value representing a higher level of the one or more undesirable characteristics. The cost of an overlap between an insert of the edge's destination amplicon and an insert of the edge's origin amplicon may be determined based on a redundancy introduced by overlapping inserts. The cost of an overlap between an insert of the edge's destination amplicon and an insert of the edge's origin amplicon may be a function of a quotient between a number of base pairs in an overlap between the insert of the edge's destination amplicon and the insert of the edge's origin amplicon and a number of base pairs in a union of the insert of the edge's destination amplicon and the insert of the edge's origin amplicon.

According to an exemplary embodiment, there is provided a non-transitory machine-readable storage medium comprising instructions which, when executed by a processor, cause the processor to perform a method comprising: (1) receiving or providing as inputs a genomic target region and a set of candidate amplicons for the genomic target region; (2) generating a graph comprising a source vertex, a set of amplicon vertices arranged in correspondence with the set of candidate amplicons, and a sink vertex; (3) determining a cost associated with one or more paths across the graph from the source vertex to the sink vertex via amplicon vertices; and (4) extracting the amplicon vertices from the one of the one or more paths across the graph having a least cost associated therewith. In some embodiments, such a method may be extended to a method for pooling amplicons across a plurality of pools by using as input the amplicons corresponding to the extracted vertices.

According to an exemplary embodiment, there is provided a system, comprising: (1) a machine-readable memory; and (2) a processor configured to execute machine-readable instructions, which, when executed by the processor, cause the system to perform steps including: (a) receiving or providing as inputs a genomic target region and a set of candidate amplicons for the genomic target region; (b) generating a graph comprising a source vertex, a set of amplicon vertices arranged in correspondence with the set of candidate amplicons, and a sink vertex; (c) determining a cost associated with one or more paths across the graph from the source vertex to the sink vertex via amplicon vertices; and (d) extracting the amplicon vertices from the one of the one or more paths across the graph having a least cost associated therewith. In some embodiments, such a system may be extended to a system for pooling amplicons across a plurality of pools by using as input the amplicons corresponding to the extracted vertices.

According to various exemplary embodiments, one or more features of any one or more of the above-discussed teachings and/or exemplary embodiments may be performed or implemented using appropriately configured and/or programmed hardware and/or software elements. Determining whether an embodiment is implemented using hardware and/or software elements may be based on any number of factors, such as desired computational rate, power levels, heat tolerances, processing cycle budget, input data rates, output data rates, memory resources, data bus speeds, etc., and other design or performance constraints.

Examples of hardware elements may include processors, microprocessors, input(s) and/or output(s) (I/O) device(s) (or peripherals) that are communicatively coupled via a local interface circuit, circuit elements (e.g., transistors, resistors, capacitors, inductors, and so forth), integrated circuits, application specific integrated circuits (ASIC), programmable logic devices (PLD), digital signal processors (DSP), field programmable gate array (FPGA), logic gates, registers, semiconductor device, chips, microchips, chip sets, and so forth. The local interface may include, for example, one or more buses or other wired or wireless connections, controllers, buffers (caches), drivers, repeaters and receivers, etc., to allow appropriate communications between hardware components. A processor is a hardware device for executing software, particularly software stored in memory. The processor can be any custom made or commercially available processor, a central processing unit (CPU), an auxiliary processor among several processors associated with the computer, a semiconductor based microprocessor (e.g., in the form of a microchip or chip set), a macroprocessor, or any device for executing software instructions. A processor can also represent a distributed processing architecture. The I/O devices can include input devices, for example, a keyboard, a mouse, a scanner, a microphone, a touch screen, an interface for various medical devices and/or laboratory instruments, a bar code reader, a stylus, a laser reader, a radio-frequency device reader, etc. Furthermore, the I/O devices also can include output devices, for example, a printer, a bar code printer, a display, etc. Finally, the I/O devices further can include devices that communicate as both inputs and outputs, for example, a modulator/demodulator (modem; for accessing another device, system, or network), a radio frequency (RF) or other transceiver, a telephonic interface, a bridge, a router, etc.

Examples of software may include software components, programs, applications, computer programs, application programs, system programs, machine programs, operating system software, middleware, firmware, software modules, routines, subroutines, functions, methods, procedures, software interfaces, application program interfaces (API), instruction sets, computing code, computer code, code segments, computer code segments, words, values, symbols, or any combination thereof. A software in memory may include one or more separate programs, which may include ordered listings of executable instructions for implementing logical functions. The software in memory may include a system for identifying data streams in accordance with the present teachings and any suitable custom made or commercially available operating system (O/S), which may control the execution of other computer programs such as the system, and provides scheduling, input-output control, file and data management, memory management, communication control, etc.

According to various exemplary embodiments, one or more features of any one or more of the above-discussed teachings and/or exemplary embodiments may be performed or implemented using appropriately configured and/or programmed non-transitory machine-readable medium or article that may store an instruction or a set of instructions that, if executed by a machine, may cause the machine to perform a method and/or operations in accordance with the exemplary embodiments. Such a machine may include, for example, any suitable processing platform, computing platform, computing device, processing device, computing system, processing system, computer, processor, scientific or laboratory instrument, etc., and may be implemented using any suitable combination of hardware and/or software. The machine-readable medium or article may include, for example, any suitable type of memory unit, memory device, memory article, memory medium, storage device, storage article, storage medium and/or storage unit, for example, memory, removable or non-removable media, erasable or non-erasable media, writeable or re-writeable media, digital or analog media, hard disk, floppy disk, read-only memory compact disc (CD-ROM), recordable compact disc (CD-R), rewriteable compact disc (CD-RW), optical disk, magnetic media, magneto-optical media, removable memory cards or disks, various types of Digital Versatile Disc (DVD), a tape, a cassette, etc., including any medium suitable for use in a computer. Memory can include any one or a combination of volatile memory elements (e.g., random access memory (RAM, such as DRAM, SRAM, SDRAM, etc.)) and nonvolatile memory elements (e.g., ROM, EPROM, EEROM, Flash memory, hard drive, tape, CDROM, etc.). Moreover, memory can incorporate electronic, magnetic, optical, and/or other types of storage media. Memory can have a distributed architecture where various components are situated remote from one another, but are still accessed by the processor. The instructions may include any suitable type of code, such as source code, compiled code, interpreted code, executable code, static code, dynamic code, encrypted code, etc., implemented using any suitable high-level, low-level, object-oriented, visual, compiled and/or interpreted programming language.

According to various exemplary embodiments, one or more features of any one or more of the above-discussed teachings and/or exemplary embodiments may be performed or implemented at least partly using a distributed, clustered, remote, or cloud computing resource.

According to various exemplary embodiments, one or more features of any one or more of the above-discussed teachings and/or exemplary embodiments may be performed or implemented using a source program, executable program (object code), script, or any other entity comprising a set of instructions to be performed. When a source program, the program can be translated via a compiler, assembler, interpreter, etc., which may or may not be included within the memory, so as to operate properly in connection with the O/S. The instructions may be written using (a) an object oriented programming language, which has classes of data and methods, or (b) a procedural programming language, which has routines, subroutines, and/or functions, which may include, for example, C, C++, Pascal, Basic, Fortran, Cobol, Perl, Java, and Ada.

According to various exemplary embodiments, one or more of the above-discussed exemplary embodiments may include transmitting, displaying, storing, printing or outputting to a user interface device, a computer readable storage medium, a local computer system or a remote computer system, information related to any information, signal, data, and/or intermediate or final results that may have been generated, accessed, or used by such exemplary embodiments. Such transmitted, displayed, stored, printed or outputted information can take the form of searchable and/or filterable lists of runs and reports, pictures, tables, charts, graphs, spreadsheets, correlations, sequences, and combinations thereof, for example.

Various additional exemplary embodiments may be derived by repeating, adding, or substituting any generically or specifically described features and/or components and/or substances and/or steps and/or operating conditions set forth in one or more of the above-described exemplary embodiments. Further, it should be understood that an order of steps or order for performing certain actions is immaterial so long as the objective of the steps or action remains achievable, unless specifically stated otherwise. Furthermore, two or more steps or actions can be conducted simultaneously so long as the objective of the steps or action remains achievable, unless specifically stated otherwise. Moreover, any one or more feature, component, aspect, step, or other characteristic mentioned in one of the above-discussed exemplary embodiments may be considered to be a potential optional feature, component, aspect, step, or other characteristic of any other of the above-discussed exemplary embodiments so long as the objective of such any other of the above-discussed exemplary embodiments remains achievable, unless specifically stated otherwise.

In some embodiments, the amplified target sequences generated by the methods disclosed herein represent at least 60%, 70%, 80%, 90%, or more, of one or more exons amplified from the plurality of target sequences. In one embodiment, amplified target sequences of the present invention are about 90 to about 140 base pairs in length, about 100 to about 200 base pairs in length, about 100 to about 300 base pairs in length, or about 100 to about 400 base pairs in length. In one embodiment, the amplified target sequence includes the length of the forward primer and the length of the complementary reverse primer for each primer pair. In another embodiment, the amplified target sequence length includes the length of the reverse primer and the length of the complementary forward primer. In some embodiments, the length of the amplified target sequence minus the forward and reverse primer lengths is about 40 base pairs to about 350 base pairs. In some embodiments, the length of the amplified target sequences generated in the multiplex PCR reaction is substantially the same. As defined herein, “substantially the same” with respect to length of amplified target sequences generated via the methods disclosed herein refers to no more than 30% deviation in nucleotide length across the total number of amplified target sequences. In one embodiment, the percent GC content of an amplicon is less than 85%, less than 75%, less than 65%, less than 60%, or less than 50%. In one embodiment, substantially all amplified target sequences within a reaction contain between 30% and less than 85% GC content. In one embodiment, where the nucleic acid molecules are obtained from an archived or FFPE RNA sample, the length of the amplified target sequence is typically about 100 to about 200 base pairs in length. In one embodiment, if the nucleic acid sample is derived or obtained from expressed RNA or cDNA, the length of the amplified target sequence can be about 100 to about 500 base pairs in length.

In some embodiments, the amplified target sequences of provided methods can be used in various downstream analysis or assays with, or without, further purification or manipulation. For example, the amplified target sequences can be clonally amplified by techniques known in the art, such a bridge amplification or emPCR to generate a template library that can be used in next generation sequencing. In some embodiments, the amplified target sequences of provided methods or the resulting template libraries can be used for single nucleotide polymorphism (SNP) analysis, genotyping or epigenetic analysis, copy number variation analysis, gene expression analysis, analysis of gene mutations including but not limited to detection, prognosis and/or diagnosis, detection and analysis of rare or low frequency allele mutations, nucleic acid sequencing including but not limited to de novo sequencing, targeted resequencing and synthetic assembly analysis. In one embodiment, amplified target sequences can be used to detect mutations at less than 5% allele frequency. In some embodiments, the methods disclosed herein can be used to detect mutations in a population of nucleic acids at less than 4%, 3%, 2% or at about 1% allele frequency. In another embodiment, amplified target sequences prepared as described herein can be sequenced to detect and/or identify germline or somatic mutations from a population of nucleic acid molecules.

In some embodiments, the forward and/or reverse target-specific primers in the target-specific primer pairs can be “complementary” or “substantially complementary” to the population of nucleic acid molecules. As termed herein “substantially complementary to the population of nucleic acid molecules” refers to percentage complementarity between the primer and the nucleic acid molecule to which the primer will hybridize. The term “substantially complementary” as used herein refers to at least 70% complementarity. Therefore, substantially complementary refers to a range of complementarity of at least 70% but less than 100% complementarity between the primer and the nucleic acid molecule. A complementary primer is one that possesses 100% complementarity to the nucleic acid molecule. In one embodiment, each target-specific primer pair is designed to minimize cross-hybridization to another primer (or primer pair) in the same multiple PCR reaction (i.e., reduce the prevalence of primer-dimers). In another embodiment, each target-specific primer pair is designed to minimize cross-hybridization to non-specific nucleic acid sequences in the population of nucleic acid molecules (i.e., minimize off-target hybridization). In one embodiment, each target-specific primer is designed to minimize self-complementarity, formation of hairpin structures or other secondary structures.

In some embodiments, the amplified target sequences are formed via polymerase chain reaction. Extension of target-specific primers can be accomplished using one or more DNA polymerases. In one embodiment, the polymerase can be any Family A DNA polymerase (also known as pol I family) or any Family B DNA polymerase. In some embodiments, the DNA polymerase can be a recombinant form capable of extending target-specific primers with superior accuracy and yield as compared to a non-recombinant DNA polymerase. For example, the polymerase can include a high-fidelity polymerase or thermostable polymerase. In some embodiments, conditions for extension of target-specific primers can include ‘Hot Start’ conditions, for example Hot Start polymerases, such as Amplitaq Gold® DNA polymerase (Applied Biosciences), Platinum® Taq DNA Polymerase High Fidelity (Invitrogen) or KOD Hot Start DNA polymerase (EMD Biosciences). A ‘Hot Start’ polymerase includes a thermostable polymerase and one or more antibodies that inhibit DNA polymerase and 3′-5′ exonuclease activities at ambient temperature. In some instances, ‘Hot Start’ conditions can include an aptamer.

In some embodiments, the polymerase can be an enzyme such as Taq polymerase (from Thermus aquaticus), Tfi polymerase (from Thermus filiformis), Bst polymerase (from Bacillus stearothermophilus), Pfu polymerase (from Pyrococcus furiosus), Tth polymerase (from Thermus thermophilus), Pow polymerase (from Pyrococcus woesei), Tli polymerase (from Thermococcus litoralis), Ultima polymerase (from Thermotoga maritima), KOD polymerase (from Thermococcus kodakaraensis), Pol I and II polymerases (from Pyrococcus abyssi) and Pab (from Pyrococcus abyssi). In some embodiments, the DNA polymerase can include at least one polymerase such as Amplitaq Gold® DNA polymerase (Applied Biosciences), Stoffel fragment of Amplitaq® DNA Polymerase (Roche), KOD polymerase (EMD Biosciences), KOD Hot Start polymerase (EMD Biosciences), Deep Vent™ DNA polymerase (New England Biolabs), Phusion polymerase (New England Biolabs), Klentaql polymerase (DNA Polymerase Technology, Inc), Klentaq Long Accuracy polymerase (DNA Polymerase Technology, Inc), Omni KlenTaq™ DNA polymerase (DNA Polymerase Technology, Inc), Omni KlenTaq™ LA DNA polymerase (DNA Polymerase Technology, Inc), Platinum® Taq DNA Polymerase (Invitrogen), Hemo Klentaq™ (New England Biolabs), Platinum® Taq DNA Polymerase High Fidelity (Invitrogen), Platinum® Pfx (Invitrogen), Accuprime™ Pfx (Invitrogen), or Accuprime™ Taq DNA Polymerase High Fidelity (Invitrogen).

In some embodiments, the DNA polymerase can be a thermostable DNA polymerase. In some embodiments, the mixture of dNTPs can be applied concurrently, or sequentially, in a random or defined order. In some embodiments, the amount of DNA polymerase present in the multiplex reaction is significantly higher than the amount of DNA polymerase used in a corresponding single plex PCR reaction. As defined herein, the term “significantly higher” refers to an at least 3-fold greater concentration of DNA polymerase present in the multiplex PCR reaction as compared to a corresponding single plex PCR reaction.

In some embodiments, the amplification reaction does not include a circularization of amplification product, for example as disclosed by rolling circle amplification.

In some embodiments, the methods of the disclosure include selectively amplifying target sequences in a sample containing a plurality of nucleic acid molecules and ligating the amplified target sequences to at least one adapter and/or barcode. Adapters and barcodes for use in molecular biology library preparation techniques are well known to those of skill in the art. The definitions of adapters and barcodes as used herein are consistent with the terms used in the art. For example, the use of barcodes allows for the detection and analysis of multiple samples, sources, tissues or populations of nucleic acid molecules per multiplex reaction. A barcoded and amplified target sequence contains a unique nucleic acid sequence, typically a short 6-15 nucleotide sequence, that identifies and distinguishes one amplified nucleic acid molecule from another amplified nucleic acid molecule, even when both nucleic acid molecules minus the barcode contain the same nucleic acid sequence. The use of adapters allows for the amplification of each amplified nucleic acid molecule in a uniformed manner and helps reduce strand bias. Adapters can include universal adapters or propriety adapters both of which can be used downstream to perform one or more distinct functions. For example, amplified target sequences prepared by the methods disclosed herein can be ligated to an adapter that may be used downstream as a platform for clonal amplification. The adapter can function as a template strand for subsequent amplification using a second set of primers and therefore allows universal amplification of the adapter-ligated amplified target sequence. In some embodiments, selective amplification of target nucleic acids to generate a pool of amplicons can further comprise ligating one or more barcodes and/or adapters to an amplified target sequence. The ability to incorporate barcodes enhances sample throughput and allows for analysis of multiple samples or sources of material concurrently. In one example, amplified target nucleic acid molecules prepared by provided methods can be ligated to Ion Torrent™ Sequencing Adapters (A and P1 adapters, sold as a component of the Ion Fragment Library Kit, Life Technologies, Part No. 4466464) or Ion Torrent™ DNA Barcodes (Life Technologies, Part No. 4468654).

The methods disclosed herein are directed to the amplification of multiple target sequences via polymerase chain reaction (PCR). In some embodiments the multiplex PCR comprises hybridizing one or more target-specific primer pairs to a nucleic acid molecule, extending the primers of the target-specific primer pairs via template dependent synthesis in the presence of a DNA polymerase and dNTPs; repeating the hybridization and extension steps for sufficient time and sufficient temperature there generating a plurality of amplified target sequences. In some embodiments, the steps of the multiplex amplification reaction method can be performed in any order.

The amount of nucleic acid material required for successful multiplex amplification can be about 1 ng. In some embodiments, the amount of nucleic acid material can be about 10 ng to about 50 ng, about 10 ng to about 100 ng, or about 1 ng to about 200 ng of nucleic acid material. Higher amounts of input material can be used, however one aspect of the disclosure is to selectively amplify a plurality of target sequence from a low (ng) about of starting material.

The multiplex PCR amplification reactions disclosed herein can include a plurality of “cycles” typically performed on a thermocycler. Each cycle includes at least one annealing step and at least one extension step. In one embodiment, a multiplex PCR amplification reaction is performed wherein target-specific primer pairs are hybridized to a target sequence; the hybridized primers are extended generating an extended primer product/nucleic acid duplex; the extended primer product/nucleic acid duplex is denatured allowing the complementary primer to hybridize to the extended primer product, wherein the complementary primer is extended to generate a plurality of amplified target sequences. In one embodiment, the methods disclosed herein have about 5 to about cycles per preamplification reaction. The annealing temperature and/or annealing duration per cycle can be identical; can include incremental increases or decreases, or a combination of both. The extension temperature and/or extension duration per cycle can be identical; can include incremental increases or decreases, or a combination of both. For example, the annealing temperature or extension temperature can remain constant per cycle. In some embodiments, the annealing temperature can remain constant each cycle and the extension duration can incrementally increase per cycle. In some embodiments, increases or decreases in duration can occur in 15 second, 30 second, 1 minute, 2 minute or 4 minute increments. In some embodiments, increases or decrease in temperature can occur as 0.5, 1, 2, 3, or 4 Celsius deviations. In some embodiments, the amplification reaction can be conducted using hot-start PCR techniques. These techniques include the use of a heating step (>60° C.) before polymerization begins to reduce the formation of undesired PCR products. Other techniques such as the reversible inactivation or physical separation of one or more critical reagents of the reaction, for example the magnesium or DNA polymerase can be sequestered in a wax bead, which melts as the reaction is heated during the denaturation step, releasing the reagent only at higher temperatures. The DNA polymerase can also be kept in an active state by binding to an aptamer or an antibody. This binding is disrupted at higher temperatures, releasing the functional DNA polymerase that can proceed with the PCR unhindered.

In some embodiments, the disclosed methods can optionally include destroying one or more primer-containing amplification artifacts, e.g., primer-dimers, dimer-dimers or superamplicons. In some embodiments, the destroying can optionally include treating the primer and/or amplification product so as to cleave specific cleavable groups present in the primer and/or amplification product. In some embodiments, the treating can include partial or complete digestion of one or more target-specific primers. In one embodiment, the treating can include removing at least 40% of the target specific primer from the amplification product. The cleavable treatment can include enzymatic, acid, alkali, thermal, photo or chemical activity. The cleavable treatment can result in the cleavage or other destruction of the linkages between one or more nucleotides of the primer, or between one or more nucleotides of the amplification product. The primer and/or the amplification product can optionally include one or more modified nucleotides or nucleobases. In some embodiments, the cleavage can selectively occur at these sites, or adjacent to the modified nucleotides or nucleobases. In some embodiments, the cleavage or treatment of the amplified target sequence can result in the formation of a phosphorylated amplified target sequence. In some embodiments, the amplified target sequence is phosphorylated at the 5′ terminus.

In some embodiments, the template, primer and/or amplification product includes nucleotides or nucleobases that can be recognized by specific enzymes. In some embodiments, the nucleotides or nucleobases can be bound by specific enzymes. Optionally, the specific enzymes can also cleave the template, primer and/or amplification product at one or more sites. In some embodiments, such cleavage can occur at specific nucleotides within the template, primer and/or amplification product. For example, the template, primer and/or amplification product can include one or more nucleotides or nucleobases including uracil, which can be recognized and/or cleaved by enzymes such as uracil DNA glycosylase (UDG, also referred to as UNG) or formamidopyrimidine DNA glycosylase (Fpg). The template, primer and/or amplification product can include one or more nucleotides or nucleobases including RNA-specific bases, which can be recognized and/or cleaved by enzymes such as RNAseH. In some embodiments, the template, primer and/or amplification product can include one or more abasic sites, which can be recognized and/or cleaved using various proofreading polymerases or apyrase treatments. In some embodiments, the template, primer and/or amplification product can include 7,8-dihydro-8-oxoguanine (8-oxoG) nucleobases, which can be recognized or cleaved by enzymes such as Fpg. In some embodiments, one or more amplified target sequences can be partially digested by a FuPa reagent.

In some embodiments, the primer and/or amplification product includes one or more modified nucleotides including bases that bind, e.g., base pair, with other nucleotides, for example nucleotides in a complementary nucleic acid strand, via chemical linkages. In some embodiments, the chemical linkages are subject to specific chemical attack that selectively cleaves the modified nucleotides (or selectively cleaves one or more covalent linkages between the modified nucleotides and adjacent nucleotides within the primer and/or amplification product) but leaves the other nucleotides unaffected. For example, in some embodiments modified nucleotides can form disulfite linkages with other nucleotides in a complementary strand. Such disulfite linkages can be oxidized via suitable treatments. Similarly, certain modified nucleotides can base pair with other nucleotides in a complementary nucleic acid strand through linkages that can be selectively disrupted via alkali treatment. In some embodiments, the primer and/or amplification product includes one or more modified nucleotides that bind, e.g., base pair, with other nucleotides in a complementary nucleic acid strand through linkages exhibiting decreased thermal stability relative to typical base pairing linkages formed between natural bases. Such reduced-thermal stability linkages can be selectively disrupted through exposure of the primer and/or amplification product to elevated temperatures following amplification.

In an exemplary embodiment amplification primers are bisulfite in design, with either a 5′ universal forward amplification sequence linked to a 3′ target-specific forward primer, or a 5′ universal reverse amplification sequence linked to a 3′ target-specific reverse primer. Both primers contain modified nucleotides.

In some embodiments, primers are synthesized that are complementary to, and can hybridize with, discrete segments of a nucleic acid template strand, including: a primer that can hybridize to the 5′ region of the template, which encompasses a sequence that is complementary to either the forward or reverse amplification primer. In some embodiments, the forward primers, reverse primers, or both, share no common nucleic acid sequence, such that they hybridize to distinct nucleic acid sequences. For example, target-specific forward and reverse primers can be prepared that do not compete with other primer pairs within the primer pool to amplify the same nucleic acid sequence. In this example, primer pairs that do not compete with other primer pairs in the primer pool assist in the reduction of non-specific or spurious amplification products. In some embodiments, the forward and reverse primers of each primer pair are unique, in that the nucleotide sequence for each primer is non-complementary and non-identical to the other primer in the primer pair. In some embodiments, the primer pair can differ by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90% nucleotide identity. In some embodiments, the forward and reverse primers in each primer pair are non-complementary or non-identical to other primer pairs in the primer pool or multiplex reaction. For example, the primer pairs within a primer pool or multiplex reaction can differ by at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, or at least 70% nucleotide identity to other primer pairs within the primer pool or multiplex reaction. Primers are designed to minimize the formation of primer-dimers, dimer-dimers or other non-specific amplification products. Typically, primers are optimized to reduce GC bias and low melting temperatures (Tm) during the amplification reaction. In some embodiments, the primers are designed to possess a Tm of about 55° C. to about 72° C. In some embodiments, the primers of a primer pool can possess a Tm of about 59° C. to about 70° C., 60° C. to about 68° C., or 60° C. to about 65° C. In some embodiments, the primer pool can possess a Tm that does not deviate by more than 5° C.

In some embodiments, the target-specific primers do not contain a carbon-spacer or terminal linker. In some embodiments, the target-specific primers or amplified target sequences do not contain an enzymatic, magnetic, optical or fluorescent label.

The template can include a 3′ region that contains the sequence for either the upstream or downstream regions surrounding a particular gene or region of interest, such that the region of interest is bracketed by a forward amplification/upstream gene-specific fusion, and a reverse amplification/downstream region of interest fusion primer. In some embodiments, an internal separator sequence can separate the template regions that can hybridize to the amplification and gene-specific primers, and this may act as a key or barcode for subsequent downstream applications such as sequencing, etc. In some embodiments, a barcode or key can be incorporated into each of the amplification products to assist with data analysis and for example, cataloging. In some embodiments, the barcodes can be Ion Torrent™ DNA barcodes (Life Technologies).

In some embodiments, the primer includes a sufficient number of modified nucleotides to allow functionally complete degradation of the primer by the cleavage treatment, but not so many as to interfere with the primer's specificity or functionality prior to such cleavage treatment, for example in the amplification reaction. In some embodiments, the primer includes at least one modified nucleotide, but no greater than 75% of nucleotides of the primer are modified.

In some embodiments, multiple different primers including at least one modified nucleotide can be used in a single amplification reaction. For example, multiplexed primers including modified nucleotides can be added to the amplification reaction mixture, where each primer (or set of primers) selectively hybridizes to, and promotes amplification of different target nucleic acid molecules within the nucleic acid population. In some embodiments, different primer combinations can be added to the amplification reaction at plexy of at least about about 12-plex, 24-plex, 48-plex, 74-plex, 96-plex, 120-plex, 144-plex, 168-plex, 192-plex, 216-plex, 240-plex, 264-plex, 288-plex, 312-plex, 336-plex, 360-plex, 384-plex, or 398-plex. In some embodiments, the modified primers contain at least one modified nucleotide near or at the termini of the primer. In some embodiments, the modified primers contain two or more modified nucleotides within the primer sequence. In an exemplary embodiment, the primer sequence contains a uracil near, or at, the termini of the primer sequence. For the purposes of this disclosure “near” or “at the termini” of the primer sequences refers up to 10 nucleotides from the termini of the primer sequence. In some embodiments, the primer sequence contains a uracil located at, or about, the center nucleotide position of the primer sequence. For the purposes of this disclosure “at, or about the center nucleotide position of the primer sequence” refers to the incorporation of a uracil moiety at the center nucleotide of the primer sequence or within eight nucleotides, in either a 3′ or 5′ direction flanking the center nucleotide. In one embodiment, the target-specific primer sequence can contain a modified nucleobase at or about the center nucleotide position and contain a modified nucleobase at the 3′ and/or 5′ terminus. In some embodiments, the length of the forward or reverse primer sequence can be about 15 to about 40 bases in length. In some embodiments, the Tm of the primer sequence used in the multiplex reaction can be about 55° C. to about 72° C. In some embodiments, the primer pairs are designed to amplify sequences from the target nucleic acid molecules or amplicons that are about 100 base pairs to about 500 base pairs in length.

In some embodiments, the amplification reactions are conducted in parallel within a single reaction phase (for example, within the same amplification reaction mixture within a single tube). In some instances, an amplification reaction can generate a mixture of products including both the intended amplicon product as well as unintended, unwanted, nonspecific amplification artifacts such as primer-dimers. Post amplification, the reactions are then treated with any suitable agent that will selectively cleave or otherwise selectively destroy the nucleotide linkages of the modified nucleotides within the excess unincorporated primers and the amplification artifacts without cleaving or destroying the specification amplification products. For example, the primers can include uracil-containing nucleobases that can be selectively cleaved using UNG/UDG (optionally with heat and/or alkali). In some embodiments, the primers can include uracil-containing nucleotides that can be selectively cleaved using UNG and Fpg. In some embodiments, the cleavage treatment includes exposure to oxidizing conditions for selective cleavage of dithiols, treatment with RNAseH for selective cleavage of modified nucleotides including RNA-specific moieties (e.g., ribose sugars, etc.), and the like. This cleavage treatment can effectively fragment the original amplification primers and non-specific amplification products into small nucleic acid fragments that include relatively few nucleotides each. Such fragments are typically incapable of promoting further amplification at elevated temperatures. Such fragments can also be removed relatively easily from the reaction pool through the various post-amplification cleanup procedures known in the art (e.g., spin columns, NaEtOH precipitation, etc).

In some embodiments, amplification products following cleavage or other selective destruction of the nucleotide linkages of the modified nucleotides are optionally treated to generate amplification products that possess a phosphate at the 5′ termini. In some embodiments, the phosphorylation treatment includes enzymatic manipulation to produce 5′ phosphorylated amplification products. In one embodiment, enzymes such as polymerases can be used to generate 5′ phosphorylated amplification products. For example, T4 polymerase can be used to prepare 5′ phosphorylated amplicon products. Klenow can be used in conjunction with one or more other enzymes to produce amplification products with a 5′ phosphate. In some embodiments, other enzymes known in the art can be used to prepare amplification products with a 5′ phosphate group. For example, incubation of uracil nucleotide containing amplification products with the enzyme UDG, Fpg and T4 polymerase can be used to generate amplification products with a phosphate at the 5′ termini. It will be apparent to one of skill in the art that other techniques, other than those specifically described herein, can be applied to generate phosphorylated amplicons. It is understood that such variations and modifications that are applied to practice the methods, systems, kits, compositions and apparatuses disclosed herein, without resorting to undue experimentation are considered within the scope of the disclosure.

In some embodiments, primers that are incorporated in the intended (specific) amplification products, these primers are similarly cleaved or destroyed, resulting in the formation of “sticky ends” (e.g., 5′ or 3′ overhangs) within the specific amplification products. Such “sticky ends” can be addressed in several ways. For example, if the specific amplification products are to be cloned, the overhang regions can be designed to complement overhangs introduced into the cloning vector, thereby enabling sticky ended ligations that are more rapid and efficient than blunt ended ligations. Alternatively, the overhangs may need to be repaired (as with several next-generation sequencing methods). Such repair can be accomplished either through secondary amplification reactions using only forward and reverse amplification primers (e.g., correspond to A and P1 primers) comprised of only natural bases. In this manner, subsequent rounds of amplification rebuild the double-stranded templates, with nascent copies of the amplicon possessing the complete sequence of the original strands prior to primer destruction. Alternatively, the sticky ends can be removed using some forms of fill-in and ligation processing, wherein the forward and reverse primers are annealed to the templates. A polymerase can then be employed to extend the primers, and then a ligase, optionally a thermostable ligase, can be utilized to connect the resulting nucleic acid strands. This could obviously be also accomplished through various other reaction pathways, such as cyclical extend-ligation, etc. In some embodiments, the ligation step can be performed using one or more DNA ligases.

In some embodiments, provided are methods for single-tube multiplex PCR. In some embodiments, the method for single-tube multiplex PCR can include target-specific or exon-specific primers. In some embodiments, the exon-specific or target-specific primers can include at least one uracil nucleotide. In some embodiments, single-tube multiplex PCR can include selective amplification of at least 10, 50, 100, 150, 200, 250, 300, 350, 398 or more target nucleic acid molecules using target-specific or exon-specific uracil based primers.

In some embodiments, provided are methods for generating a target-specific or exon-specific amplicon library. In some embodiments, the amplicon library generated using target-specific or exon-specific primers can be associated with aberrant expression in human cancers. In some embodiments, the amplicon library generated using target-specific or exon-specific primers can be associated with mutations of human cancers. In some embodiments, the amplicon library can be generated from expressed RNA or cDNA or formalin-fixed, paraffin-embedded (FFPE) tissue. In some embodiments, the amplicons of the amplicon library prepared using the methods disclosed herein can be about 100 to about 300 base pairs in length, about 100 to about 250 base pairs in length, about 120 to about 220 base pairs in length or about 135 to about 205 base pairs in length. In some embodiments, the amplicon library can be prepared using primer pairs that are targeted to cancer specific mutations. In some embodiments, the primer pairs can be used to generate amplicons that once sequenced by any sequencing platform, including semi-conductor sequencing technology can be used to detect genetic mutations such as inversion, deletions, point mutations and variations in copy number.

In some embodiments, the primer pairs used to produce an amplicon library can result in the amplification of target-specific nucleic acid molecules possessing one or more of the following metrics: greater than 97% target coverage at 20× if normalized to 100× average coverage depth; greater than 97% of bases with greater than 0.2× mean; greater than 90% base without strand bias; greater than 95% of all reads on target; greater than 99% of bases with greater than 0.01× mean; and greater than 99.5% per base accuracy.

In some embodiments, the amplicon library can be used to detect and/or identify known mutations or de novo mutations in a sample.

In some embodiments, the amplicon library prepared using target—specific primer pairs can be used in downstream enrichment applications such as emulsion PCR or bridge PCR. In some embodiments, the amplicon library can be used in an enrichment application and a sequencing application. For example, an amplicon library can be sequenced using any suitable DNA sequencing platform. In some embodiments, an amplicon library can be sequenced using an Ion Torrent PGM Sequencer (Life Technologies). In some embodiments, a PGM sequencer can be coupled to server that applies parameters or software to determine the sequence of the amplified target nucleic acid molecules. In some embodiments, the amplicon library can be prepared, enriched and sequenced in less than 24 hours. In some embodiments, the amplicon library can be prepared, enriched and sequenced in approximately 9 hours. In some embodiments, an amplicon library can be a paired library, that is, a library that contains amplicons from a tumor sample and amplicons from a non-diseased sample. Each pair can be aligned, to detect and/or identify mutations present in the target nucleic acid molecules.

In some embodiments, methods for generating an amplicon library can include:

    • amplifying RNA expression or cDNA targets using exon-specific or target-specific primers to generate amplicons; purifying the amplicons from the input RNA or cDNA and primers;

phosphorylating the amplicons; ligating adapters to the phosphorylated amplicons; purifying the ligated amplicons; nick-translating the amplified amplicons; and purifying the nick-translated amplicons to generate the amplicon library. In some embodiments, additional amplicon library manipulations can be conducted following the step of amplification of RNA expression targets to generate the amplicons. In some embodiments, any combination of additional reactions can be conducted in any order, and can include: purifying; phosphorylating; ligating adapters; nick-translating; amplification and/or sequencing. In some embodiments, any of these reactions can be omitted or can be repeated. It will be readily apparent to one of skill in the art that the method can repeat or omit any one or more of the above steps. It will also be apparent to one of skill in the art that the order and combination of steps may be modified to generate the required amplicon library, and is not therefore limited to the exemplary methods provided.

A phosphorylated amplicon can be joined to an adapter to conduct a nick translation reaction, subsequent downstream amplification (e.g., template preparation), or for attachment to particles (e.g., beads), or both. For example, an adapter that is joined to a phosphorylated amplicon can anneal to an oligonucleotide capture primer which is attached to a particle, and a primer extension reaction can be conducted to generate a complementary copy of the amplicon attached to the particle or surface, thereby attaching an amplicon to a surface or particle. Adapters can have one or more amplification primer hybridization sites, sequencing primer hybridization sites, barcode sequences, and combinations thereof. In some embodiments, amplicons prepared by the methods disclosed herein can be joined to one or more Ion Torrent™ compatible adapters to construct an amplicon library. Amplicons generated by such methods can be joined to one or more adapters for library construction to be compatible with a next generation sequencing platform. For example, the amplicons produced by the teachings of the present disclosure can be attached to adapters provided in the Ion Fragment Library Kit (Life Technologies, Catalog No. 4466464).

In some embodiments, amplification of expressed RNA targets or FFPE samples can be conducted using a 2× AmpliSeq Hi Fi Master Mix. In some embodiments, the AmpliSeq Hi Fi Master Mix can include glycerol, dNTPs, and a DNA polymerase, such as Platinum® Taq DNA polymerase High Fidelity. In some embodiments, the 2× AmpliSeq Hi Fi Master Mix can further include at least one of the following: a preservative, magnesium sulfate, tris-sulfate and/or ammonium sulfate.

In some embodiments, amplification of expressed RNA targets or FFPE samples can be conducted using a 5× Ion AmpliSeq Hi Fi Master Mix. In some embodiments, the 5× Ion AmpliSeq Hi Fi Master Mix can include glycerol, dNTPs, and a DNA polymerase such as Platinum® Taq DNA polymerase High Fidelity. In some embodiments, the 5× Ion AmpliSeq Hi Fi Master Mix can further include at least one of the following: a preservative, magnesium chloride, magnesium sulfate, tris-sulfate and/or ammonium sulfate.

In some embodiments, phosphorylation of the amplicons can be conducted using a FuPa reagent. In some embodiments, the FuPa reagent can include a DNA polymerase, a DNA ligase, at least one uracil cleaving or modifying enzyme, and/or a storage buffer. In some embodiments, the FuP reagent can further include at least one of the following: a preservative and/or a detergent.

In some embodiments, phosphorylation of the amplicons can be conducted using a FuPa reagent. In some embodiments, the FuPa reagent can include a DNA polymerase, at least one uracil cleaving or modifying enzyme, an antibody and/or a storage buffer. In some embodiments, the FuPa reagent can further include at least one of the following: a preservative and/or a detergent. In some embodiments, the antibody is provided to inhibit the DNA polymerase and 3′-5′ exonuclease activities at ambient temperature.

In some embodiments, the amplicon library produced by the teachings of the present disclosure are sufficient in yield to be used in a variety of downstream applications including the Ion Xpress™ Template Kit using an Ion Torrent™ PGM system (e.g., PCR-mediated addition of the nucleic acid fragment library onto Ion Sphere™ Particles)(Life Technologies, Part No. 4467389). For example, instructions to prepare a template library from the amplicon library can be found in the Ion Xpress Template Kit User Guide (Thermo Fisher Scientific). Instructions for loading the subsequent template library onto the Ion Torrent™ Chip for nucleic acid sequencing are described in the Ion Sequencing User Guide (Thermo Fisher Scientific). In some embodiments, the amplicon library produced by the teachings of the present disclosure can be used in paired end sequencing (e.g., paired-end sequencing on the Ion Torrent™ PGM system (Thermo Fisher Scientific).

It will be apparent to one of ordinary skill in the art that numerous other techniques, platforms or methods for clonal amplification such as wildfire PCR and bridge amplification can be used in conjunction with the amplified target sequences of the present disclosure. It is also envisaged that one of ordinary skill in art upon further refinement or optimization of the conditions provided herein can proceed directly to nucleic acid sequencing (for example using the Ion Torrent PGM™ or Proton™ sequencers, Life Technologies) without performing a clonal amplification step.

In some embodiments, at least one of the amplified targets sequences to be clonally amplified can be attached to a support or particle. The support can be comprised of any suitable material and have any suitable shape, including, for example, planar, spheroid or particulate. In some embodiments, the support is a scaffolded polymer particle as described in U.S. Published App. No. 20100304982, hereby incorporated by reference in its entirety.

In some embodiments, nucleic acid sequencing of the amplified target sequences produced by the teachings of this disclosure include de novo sequencing or targeted resequencing. In some embodiments, nucleic acid sequencing further includes comparing the nucleic acid sequencing results of the amplified target sequences against a reference nucleic acid sample. In some embodiments, the reference sample can be normal tissue or well documented tumor sample. In some embodiments, nucleic acid sequencing of the amplified target sequences further includes determining the presence or absence of a mutation within the nucleic acid sequence. In some embodiments, the method further includes correlating the presence of a mutation with drug susceptibly, prognosis of treatment and/or organ rejection. In some embodiments, nucleic acid sequencing includes the identification of genetic markers associated with cancer. In some embodiments, nucleic acid sequencing includes the identification of copy number variation in a sample under investigation.

In some embodiments, a kit is provided for amplifying multiple immune response target expression sequences from a population of nucleic acid molecules in a single reaction. In some embodiments, the kit includes a plurality of target-specific primer pairs containing one or more cleavable groups, one or more DNA polymerases, a mixture of dNTPs and at least one cleaving reagent. In one embodiment, the cleavable group can be 8-oxo-deoxyguanosine, deoxyuridine or bromodeoxyuridine. In some embodiments, the at least one cleaving reagent includes RNaseH, uracil DNA glycosylase, Fpg or alkali. In one embodiment, the cleaving reagent can be uracil DNA glycosylase. In some embodiments, the kit is provided to perform multiplex PCR in a single reaction chamber or vessel. In some embodiments, the kit includes at least one DNA polymerase, which can be a thermostable DNA polymerase. In some embodiments, the concentration of the one or more DNA polymerases is present in a 3-fold excess as compared to a single PCR reaction. In some embodiments, the final concentration of each target-specific primer pair is present at about 25 nM to about 50 nM. In one embodiment, the final concentration of each target-specific primer pair can be present at a concentration that is 50% lower than conventional single-plex PCR reactions. In some embodiments, the kit provides amplification of at least 100, 150, 200, 250, 300, 350, 398, or more, target immune response expression sequences from a population of nucleic acid molecules in a single reaction chamber. In particular embodiments, a provided kit of the invention is a test kit.

In some embodiments, the kit further comprises one or more adapters, barcodes, and/or antibodies.

A common concern in study of RNA expression analyses is determination of how gene expression changes between different samples. Current methodology for RNA sequencing typically requires an amplification step followed by sequence analysis of select amplified products (e.g., transcripts) to a fixed number of reads. In particular, with the provided methods of the invention, expression of each gene can be inferred by amplifying an amplicon that represents a transcript (e.g., an amplicon crosses a selected exon/exon junction). Due to the selected amplification, there is a dependency between the number of reads associated with a particular gene transcript and the number of reads produced by all other genes. Such interdependency means an apparent identical fold-change can be induced by two distinct biological processes: an increase in the ration of expression of a particular gene, or a decrease in the expression ration of all other genes measured. To accurately measure which of the two scenarios takes place, number of reads should be correctly normalized within each sample before cross comparison of samples takes place. Normalization has been studies for full transcriptome RNA sequencing analyses. Among such studies, log RPM (read counts per million) has been widely accepted to provide an interpretable measure of gene expression that robustly reflects the difference in expression between samples. The accuracy of this measure, however, depends on the underlying assumption that the majority of the genes measured are not differentially expressed. This assumption tends to hold true for transcriptomic studies, where more than 20,000 genes of the same tissues are typically profiled. However, such an assumption is unlikely to always hold for smaller gene panels, for panels comprising very low expression genes, or for panels that focus on similar biological processes, such as, for example, the methods and compositions provided herein. As a simple illustration, consider a targeted panel of 20 genes made up of 2 functional categories, one with 16 and the other with 4 genes, where each gene of a function is perfectly correlated to the others. In this example, the actual number of RNA molecules in sample 1 and sample 2 is known, and can sequence an exact of proportion of reads. When a standard RPM computation is applied to measure the expression of each gene and calculate the fold change using our estimated expression, there is 2-fold increase in the smaller category, the fold change estimate maintains a fairly accurate estimate of true fold change (FIG. 8, left panel). However, if the number of molecules in the larger category of 16 genes increases 2-fold, the estimated fold change based on RPM, suggests that instead of an increase of 16 genes, we have decrease in 4 genes (FIG. 8, middle panel). Thus, on a targeted panel with a small number of total genes and strong correlation structure of most genes, standard RPM normalization provides inaccurate fold change estimation. However, if we assume that a smaller category is a set of standard endogenous controls, e.g., housekeeping (HK) genes, and are comparable across samples, this set of genes as a reference point to accurately measure the expression the target functional set of genes. Using the latter strategy provides an accurate fold change result (FIG. 8, right panel). Knowing how correlated the genes are to each other provides some guidelines for whether or not standard expression measurements will be biased on small, targeted RNA-seq panels.

Thus, in conjunction with the compositions and methods of the invention, provided compositions include primer pairs and methods for determination of expression of a set of housekeeping genes for use of one or more as endogenous controls and an effective means for normalization of expression levels and determination of expression changes across samples.

The following description of various exemplary embodiments is exemplary and explanatory only and is not to be construed as limiting or restrictive in any way. Other embodiments, features, objects, and advantages of the present teachings will be apparent from the description and accompanying drawings, and from the claims.

Although the present description described in detail certain exemplary embodiments, other embodiments are also possible and within the scope of the present invention. Variations and modifications will be apparent to those skilled in the art from consideration of the specification and figures and practice of the teachings described in the specification and figures, and the claims.

EXEMPLIFICATION

Materials and Methods

Provided immune response assay compositions include reagents directed to 395 immune response markers designed for library preparation technology of expressed target sequences. Generally, RNAs extracted from samples (e.g., tumor samples, e.g., fresh, frozen, FFPE, of various types, e.g., lung tumor, NSCLC) were reverse transcribed; libraries were generated, template prepared, e.g., using Ion Chef™ or Ion OneTouch™ 2 System, then prepared samples were sequenced using next generation sequencing technology, e.g., an Ion S5™, an Ion PGM™ System.

Total RNA was extracted from samples, 10 ng of total RNA was first reverse transcribed to and used as template to amplify targets (e.g., target expression sequences in Table 1) with the immune response assay comprising primer pairs in Table 2. Manual library prep was performed with the Ion AmpliSeg™ Library Kit 2.0 which was then templated with either the Ion OneTouch™ 2 System or Ion Chef™ instrument. Automated library prep and templating was performed with the Ion AmpliSeg™ Kit for Chef DL8 and Ion Chef™ Instrument. Sequencing was performed on either the Ion S5™ System or the Ion PGM™ System. Gene expression was quantified with the Ion Torrent Suite™ software.

HL-60 and human normal lung (AM7968) were purchased from ATCC and Ambion, respectively. De-identified melanoma and NSCLC FFPE samples were procured from Asterand and Research Dx respectively.

TABLE 1 genes and amplicon insert sequences of the immune response assay SEQ Gene/ ID Accession Amplicon Sequence NO: CD63/NM_00178 CCCGACTCCTGCTGCATTAATGTTACTGTGGGCTGTGGGATTAATTTCAACGA 1 0 GAAGGCGATCCATAAGGAGGGCTGTGTGGAGAAGATTGGGGGCTGGCTGAGGA AA CD69/NM_00178 GTTCCTGTCCTGTGTGCTGTAATGAATGTGGTCTTCATCACCATTTTAATCAT 2 1 AGCTCTCATTGCCTTATCAGTGGGCCAATACAATTGTCCAGGCCAATACACAT TC CXCL1/NM_0015 TAAGAAAATCATCGAAAAGATGCTGAACAGTGACAAATCCAACTGACCAGAAG 3 11 GGAGGAGGAAGCTCACTGGTGGCTGTTCCTGAAGGAGGCCCTGCCCTTA KLRD1/NM_0022 CGGCATCTCTGTGCTTCTCAGAAATCCAGCCTGCTTCAGCTTCAAAACACAGA 4 62 TGAACTGGATTTTATGAGCTCCAGTCAACAATTTTACTGGATTGGACTCTC HLA- CTCCAGAAGATTTTGTGATTCAGGCAAAGGCTGACTGTTACTTCACCAACGGG 5 DOB/NM_002120 ACAGAAAAGGTGCAGTTTGTGGTCAGATTCATCTTTAACTTGGAGGAGTATGT AC CXCR5/NM_0017 GGTGACTCACAGCCGGCACAGCCATGAACTACCCGCTAACGCTGGAAATGGAC 6 16 CTCGAGAACCTGGAGGACCTGTTCTGGGAACTGGACAGATTGGACA IL12B/NM_0021 CAAAGGAGGCGAGGTTCTAAGCCATTCGCTCCTGCTGCTTCACAAAAAGGAAG 7 87 ATGGAATTTGGTCCACTGATATTTTAAAGGACCAGAAAGAACCCAAAAATAAG AC PTK7/NM_00282 TTGCCTGACCCAGGCCACACCAAAACCTACAGTTGTCTGGTACAGAAACCAGA 8 1 TGCTCATCTCAGAGGACTCACGGTTCGAGGTCTTCAAGAATGGGACCTTGCGC ATC CEACAM1/NM_00 GTCACTGGCTGCAACAGGACCACAGTCAAGACGATCATAGTCACTGAGCTAAG 9 1712 TCCAGTAGTAGCAAAGCCCCAAATCAAAGCCAGCAAGACCACAGTCACAGGAG AT CXCL9/NM_0024 CCAAGGGACTATCCACCTACAATCCTTGAAAGACCTTAAACAATTTGCCCCAA 10 16 GCCCTTCCTGCGAGAAAATTGAAATCATTGCTACACTGAAGAATGGAG IL13/NM_00218 CAACGTGTCAGGCTGCAGTGCCATCGAGAAGACCCAGAGGATGCTGAGCGGAT 11 8 TCTGCCCGCACAAGGTCTCAGCTGGGCAGTTTTCCAGCTTGCATGTCCGAGAC ACCA NT5E/NM_00252 TGAGGGGTGTGGACGTCGTGGTGGGAGGACACTCCAACACATTTCTTTACACA 12 6 GGCAATCCACCTTCCAAAGAGGTGCCTGCTGGGAAGTACCCATTCATAG VEGFA/NM_0011 CAGATGTGACAAGCCGAGGCGGTGAGCCGGGCAGGAGGAAGGAGCCTCCCTCA 13 71623 GGGTTTCGGGAACCAGATCTCTCACCAGGAAAGACTGATACAGAACGATCGA ABCF1/NM_0010 TGATGTGTTGCTGTGTGAGCAGGAGGTGGTAGCAGATGAGACACCAGCAGTCC 14 25091 AGGCTGTTCTTCGAGCTGACACCAAGCGATTGAAGCTGCTGGAAGAGGAGCGG CGG CD38/NM_00177 TCCAGCGGGACATGTTCACCCTGGAGGACACGCTGCTAGGCTACCTTGCTGAT 15 5 GACCTCACATGGTGTGGTGAATTCAACACTTCCAAAATAAACTATCAATCTTG CCC JAML/NM_00109 CTTTCAGTTGAGCTTGGGGACTGCAGCTGTGGGGAGATTTCAGTGCATTGCCT 16 8526 CCCCTGGGTGCTCTTCATCTTGGATTTGAAAGTTGAGAGCAGCATGTTTTGCC CACT S100A8/NM_002 GGGAATTTCCATGCCGTCTACAGGGATGACCTGAAGAAATTGCTAGAGACCGA 17 964 GTGTCCTCAGTATATCAGGAAAAAGGGTGCAGACGTCTGGTT MYC/NM_002467 GCAGCTGCTTAGACGCTGGATTTTTTTCGGGTAGTGGAAAACCAGCAGCCTCC 18 CGCGACGATGCCCCTCAACGTTAGCTTCACCAACAGGAACTATGACCTCGACT ACG IRF1/NM_00219 GGAGGTGGAGCAGGCCCTGACTCCAGCACTGTCGCCATGTGCTGTCAGCAGCA 19 8 CTCTCCCCGACTGGCACATCCCAGTGGAAGTTGTGCCGGACAGCACCAGTGAT CTG CCL22/NM_0029 GCTGTGGCGCTTCAAGCAACTGAGGCAGGCCCCTACGGCGCCAACATGGAAGA 20 90 CAGCGTCTGCTGCCGTGATTACGTCCGTTACCGTCTGCCCCTGCGCGTGGTGA CXCR2/NM_0015 AGCCCAGCGACCCAGTCAGGATTTAAGTTTACCTCAAAAATGGAAGATTTTAA 21 57 CATGGAGAGTGACAGCTTTGAAGATTTCTGGAAAGGTGAAGATCTTAGTAATT A IFIT1/NM_0015 ACAGCAACCATGAGTACAAATGGTGATGATCATCAGGTCAAGGATAGTCTGGA 22 48 GCAATTGAGATGTCACTTTACATGGGAGTTATCCATTGATGACGATGA IFIT2/NM_0015 GCAGCTGCCTGAACCGAGCCCTGCCGAACAGCTGAGAATTGCACTGCAACCAT 23 47 GAGTGAGAACAATAAGAATTCCTTGGAGAGCAGCCTACGGCAACTAA CD68/NM_00125 TGGGTGAGGCGGTTCAGCCATGAGGCTGGCTGTGCTTTTCTCGGGGGCCCTGC 24 1 TGGGGCTACTGGCAGCCCAGGGGACAGGGAATGACTGTCCTCACAAAAAATCA GC M6PR/NM_00235 TCACTGGGGATTCTGAGCTTTGGCTACTCCAGTTTCCCACGACACGATGTTCC 25 5 CTTTCTACAGCTGCTGGAGGACTGGACTGCTACTACTACTCCTGGCTGTGGCA G SH2D1A/NM_002 AACAGGTTCTTGGAGTGCTGAGACAGCACCTGGGGTACATAAAAGATATTTCC 26 351 GGAAAATAAAAAATCTCATTTCAGCATTTCAGAAGCCAGATCAAGGCAT ISG20/NM_0022 GGCGTGAGGCCAAGCTGGACCACTGCAGGCGTGTCTCCCTGCGGGTGCTGAGT 27 01 GAGCGCCTCCTGCACAAGAGCATCCAGAACAGCCTGCTTGGACACAGCTCGGT GG GBP1/NM_00205 TTTCTCCCTGGACTTGGAAGCAGATGGACAACCCCTCACACCAGATGAGTACC 28 3 TGACATACTCCCTGAAGCTGAAGAAAGGTACCAGTCAAAAAGATGAAA TBP/NM_003194 CCAAGAAGAAAGTGAACATCATGGATCAGAACAACAGCCTGCCACCTTACGCT 29 CAGGGCTTGGCCTCCCCTCAGGGTGCCATGACTCCCGGAATCCCTATCTTTAG TC STAT6/NM_0031 GGGGCCTGGAAGTGCCCGCTGAGAAAGGGAGAAGACAGCAGAGGGGTTGCCGA 30 53 GGCAACCTCCAAGTCCCAGATCATGTCTCTGTGGGGTCTGGTCTCCAAGATGC CC ID3/NM_002167 ATCCTACAGCGCGTCATCGACTACATTCTCGACCTGCAGGTAGTCCTGGCCGA 31 GCCAGCCCCTGGACCCCCTGATGGCCCCCACCTTCCCATCCAGACAGCCGAGC CX3CL1/NM_002 AAGATACCTGTAGCTTTGCTCATCCACTATCAACAGAACCAGGCATCATGCGG 32 996 CAAACGCGCAATCATCTTGGAGACGAGACAGCACAGGCTGTTCTGTGCCGACC CG KLRB1/NM_0022 TTGCCCTGAAACTTAGCTGTGCTGGGATTATTCTCCTTGTCTTGGTTGTTACT 33 58 GGGTTGAGTGTTTCAGTGACATCCTTAATACAGAAATCATCAATAGAAAAATG C TNFSF4/NM_003 ACAATTTACCGAATATAAGAAGGAGAAAGGTTTCATCCTCACTTCCCAAAAGG 34 326 AGGATGAAATCATGAAGGTGCAGAACAACTCAGTCATCA CD52/NM_00180 CCAAGACAGCCACGAAGATCCTACCAAAATGAAGCGCTTCCTCTTCCTCCTAC 35 3 TCACCATCAGCCTCCTGGTTATGGTACAGATACAAACTGGACTCTCAGGACA IL10RA/NM_001 TCCACCACATCCTCCACTGGACACCCATCCCAAATCAGTCTGAAAGTACCTGC 36 558 TATGAAGTGGCGCTCCTGAGGTATGGAATAGAGTCCTGGAACTCCATCTCC HLA- TGATTAAAGCACCAGAGTGTAATGGCCCTCAGAGCAGGGCTGGTCCTGGGGTT 37 DOA/NM_002119 CCACACCCTGATGACCCTCCTGAGCCCGCAGGAGGCAGGGGCCACCAAGGCTG AC IFNB1/NM_0021 GTGGAAATCCTAAGGAACTTTTACTTCATTAACAGACTTACAGGTTACCTCCG 38 76 AAACTGAAGATCTCCTAGCCTGTGCCTCTGGGACTGGACAATTGCTTCAAGCA T CCR5/NM_00110 CGTTCCCCTACAAGAAACTCTCCCCGGGTGGAACAAGATGGATTATCAAGTGT 39 0168 CAAGTCCAATCTATGACATCAATTATTATACATCGGAGCCCTGCCAAAAAATC AA IKZF3/NM_0124 GTGTGGATTATCCTGCATCAGCTTCAATGTCTTAATGGTTCATAAGCGAAGCC 40 81 ATACTGGTGAACGCCCATTCCAGTGTAATCAGTGTGGGGCATCTTTTACTCAG A STAT1/NM_0073 AGTGCTGAGTTGGCAGTTTTCTTCTGTCACCAAAAGAGGTCTCAATGTGGACC 41 15 AGCTGAACATGTTGGGAGAGAAGCTTCTTGGTCCTAACGCCAGCCCCGATGGT CTC CD6/NM_006725 GCAGGTTCCAGATGCCACCCTTGGAGGAAGGACTTGAAGAGTTGCATGCCTCC 42 CACATCCCAACTGCCAACCCTGGACACTGCATTACAGACCCGCCATCCCTGGG CCC BRCA1/NM_0073 CTGAGTGACAAGGAATTGGTTTCAGATGATGAAGAAAGAGGAACGGGCTTGGA 43 00 AGAAAATAATCAAGAAGAGCAAAGCATGGATTCAAACTTAGGTGAAGCAGC CORO1A/NM_007 GGCCCTGATCTGTGAGGCCAGCGGGGGAGGGGCCTTCCTGGTGCTGCCCCTGG 44 074 GCAAGACTGGACGTGTGGACAAGAATGCGCCCACGGTCTGTGGCCACACAGCC TBX21/NM_0133 TTTGGGAAACTAAAGCTCACAAACAACAAGGGGGCGTCCAACAATGTGACCCA 45 51 GATGATTGTGCTCCAGTCCCTCCATAAGTACCAGCCCCGGCTGCATATCGTTG AGGTG KLRK1/NM_0073 CTCACCCAACCTACTAACAATAATTGAAATGCAGAAGGGAGACTGTGCACTCT 46 60 ATGCCTCGAGCTTTAAAGGCTATATAGAAAACTGTTCAACTCCAAATACGTAC CXCR6/NM_0065 GCATCTCTGCTGGTGTTCATCAGAACAGACACCATGGCAGAGCATGATTACCA 47 64 TGAAGACTATGGGTTCAGCAGTTTCAATGACAGCAGCCAGGAGGAGCATCAA PTEN/NM_00031 TATCTAGTACTTACTTTAACAAAAAATGATCTTGACAAAGCAAATAAAGACAA 48 4 AGCCAACCGATACTTTTCTCCAAATTTTAAGGTGAAGCTGTACTTCACAAAA PMEL/NM_00692 CCAGAGTGGACAGAAGCCCAGAGACTTGACTGCTGGAGAGGTGGTCAAGTGTC 49 8 CCTCAAGGTCAGTAATGATGGGCCTACACTGATTGGTGCAAATGCCTCCTTCT DMBT1/NM_0073 AGCACCAACCTGCTCTGTCTGCCAAATCACATGCAAGCCAGTGTGAGCAGGAG 50 29 CTATCTCCAATCCTTGGGCTTTTCTGCCAGTGACCTTGTCATTTCCACCTGGA IFI44L/NM_006 CTTCAAGACAACTTTTTAAACATGAGTAGATCTATGACTTCTCAAAGCCGGGT 51 820 CATGAATGTCCATAAAATGCTAGGCATTCCTATTTCCAATATTTTG LAPTM5/NM_006 CATGGAAGTGCCCACCTATCTCAACTTCAAGTCCATGAACCACATGAATTACC 52 762 TCCCCAGCCAGGAGGATATGCCTCATAACCAGTTCATCAAGATGATGATCATC TTT CD226/NM_0065 GTCACAGTCTCAGACTCGGGGCTTTACCGCTGCTACTTGCAGGCCAGCGCAGG 53 66 AGAAAACGAAACCTTCGTGATGAGATTGACTGTAGCCGAGGGTAAAACCGA TNFSF13B/NM_0 CTCAGCTTTAAAAGGGGAAGTGCCCTAGAAGAAAAAGAGAATAAAATATTGGT 54 06573 CAAAGAAACTGGTTACTTTTTTATATATGGTCAGGTTTTATATACTGATAAG ICOS/NM_01209 TATTACTTCTGCAACCTATCAATTTTTGATCCTCCTCCTTTTAAAGTAACTCT 55 2 TACAGGAGGATATTTGCATATTTATGAATCACAACTTTGTTGCCAGCTGAAG CD160/NM_0070 CTGGGCCTGCTGACAGCGTGCAGGATGCTGTTGGAACCCGGCAGAGGCTGCTG 56 53 TGCCCTGGCCATCCTGCTGGCAATTGTGGACATCCAGTCTGGTGGATGCATTA TRIM29/NM_012 GAATCATAGCACCGTGACAGTGGAGGAGGCCAAGGCCGAGAAGGAGACGGAGC 57 101 TGTCATTGCAAAAGGAGCAGCTGCAGCTCAAGATCATTGAGATTGAGGATGAA GC LST1/NM_00716 CCAGCCCCTGATCATTTCGCCTAAAAGAGCAAGGACTAGAGTTCCTGACCTCC 58 1 AGGCCAGTCCCTGATCCCTGACCTAATGTTATCGCGGAATGATGA ZBTB46/NM_025 GCCTGCTGTCGCTGAAGGCCGACGTGCTGGGGGATGACGGCTCCCTGCTGTTC 59 224 GAGTACCTGCCCAGAGGGGCCCACTCGCTGTCCCTGAATGAGTTCACGGTG VTCN1/NM_0246 GGCAGCGGCAGCTCCACTCAGCCAGTACCCAGATACGCTGGGAACCTTCCCCA 60 26 GCCATGGCTTCCCTGGGGCAGATCCTCTTCTGGAGCATAATTAGCATCATCAT TA KREMEN1/NM_03 AAGATACTTCTGCACGTCACATTCAAATCCCATCGTGTTCCTGCTTCAGGGGA 61 2045 CCTTAGGGATTGTCATCAACCAGGGACTTCGGGGGAAATCTGGAGCATTT PDCD1LG2/NM_0 CACCAGATAGCAGCTTTATTCACAGTGACAGTCCCTAAGGAACTGTACATAAT 62 25239 AGAGCATGGCAGCAATGTGACCCTGGAATGCAACTTTGACACTGGAAGTCATG TG TUBB/NM_17801 TGGCCAGATCTTTAGACCAGACAACTTTGTATTTGGTCAGTCTGGGGCAGGTA 63 4 ACAACTGGGCCAAAGGCCACTACACAGAGGGCGCCGAGCTGGTTGATTCTGTC CTGG CLEC4C/NM_130 TGACCCTGCATGGTGTGCGGTGCCCTCCTGCCTCAGGCCGCGAAGAAGGATCT 64 441 AAGGGCTTGGCTTGTTTGAAAGAACCACACCCCGAAAGTAACATCTTTGGAGA A CD86/NM_17586 TGTGATGGCCTTCCTGCTCTCTGGTGCTGCTCCTCTGAAGATTCAAGCTTATT 65 2 TCAATGAGACTGCAGACCTGCCATGCCAATTTGCAAACTCTCAAA HAVCR2/NM_032 TCCCAGGCATAATGAATGATGAAAAATTTAACCTGAAGTTGGTCATCAAACCA 66 782 GCCAAGGTCACCCCTGCACCGACTCGGCAGAGAGACTTCACTGCAGCCTTTCC AAGGA GZMH/NM_03342 GAGAAGAGTCGGAAGAGGTGTGGCGGCATCCTAGTGAGAAAGGACTTTGTGCT 67 3 GACAGCTGCTCACTGCCAGGGAAGCTCCATAAATGTCACCTTGGGGGCCCACA NFATC1/NM_172 GCGAGAGCCTGAAGAGTTGGACCAGTTGTACCTGGATGACGTAAATGAAATAA 68 387 TACGAAATGACCTCTCCAGCACGAGCACCCACTCCTAGTTGCCACATTGGAGC A CD8B/NM_17221 CTGCATGATCGTCGGGAGCCCCGAGCTGACCTTCGGGAAGGGAACTCAGCTGA 69 3 GTGTGGTTGATTTCCTTCCCACCACTGCCCAGCCCACCAAGAAGTCCACCCTC A BCL2/NM_00063 GGCACCTGCACACCTGGATCCAGGATAACGGAGGCTGGGATGCCTTTGTGGAA 70 3 CTGTACGGCCCCAGCATGCGGCCTCTGTTTGATTTCTCCTGGCTGTCTCTG GADD45GIP1/NM GGCGACCATGCAGGAGTCGCTGCGGGTGAAGCAGCTGGCCGAAGAGCAGAAGC 71 _052850 GTCGGGAGAGGGAGCAGCACATCGCAGAGTGCATGGCCAAGATGCCA CBLB/NM_17066 CGGCGCCGAAAGAACTAAAATTCCAGATGGCAAACTCAATGAATGGCAGAAAC 72 2 CCTGGTGGTCGAGGAGGAAATCCCCGAAAAGGTCGAATTTTGGGTATTATTGA TGCTA ITGA1/NM_1815 GACACAGCAAGAAAGGAGGCATTCACGGAAGCCCGGGGTGCCCGAAGAGGAGT 73 01 TAAAAAAGTCATGGTTATTGTGACAGATGGAGAGTCTCATGACAATC CD8A/NM_17182 GTCCCCGGCCTGTGGTCAAATCGGGAGACAAGCCCAGCCTTTCGGCGAGATAC 74 7 GTCTAACCCTGTGCAACAGCCACTACATTACTTCAAACTGAGATCCTTCCT IL2RA/NM_0004 TGGGGACTGCTCACGTTCATCATGGTGCCTGGCTGCCAGGCAGAGCTCTGTGA 75 17 CGATGACCCGCCAGAGATCCCACACGCCACATTCAAAGCCATGGC EIF2AK2/NM_00 CCACACTTCCGTGATTATCTGCGTGCATTTTGGACAAAGCTTCCAACCAGGAT 76 1135651 ACGGGAAGAAGAAATGGCTGGTGATCTTTCAGCAGGTTTCTTCATGG MADCAM1/NM_13 CAACAGGCTCGTCCAAACCTGCGGGTGACCAGCTGCCCGCGGCTCTGTGGACC 77 0760 AGCAGTGCGGTGCTGGGACTGCTGCTCCTGGCCTTGCCCACC PTPN6/NM_0805 CTATCCCCCAGCCATGAAGAATGCCCATGCCAAGGCCTCCCGCACCTCGTCCA 78 48 AACACAAGGAGGATGTGTATGAGAACCTGCACACTAAGAACAAGAGGGAGGAG AAAG LRG1/NM_05297 GCAGACAGCGACCAAAAAGCCCAGGGGGCATTCAACCCCATGTTTCTAGAACT 79 2 CTGTTCCTGCTGCTGCTGTTGGCAGCCTCAGCCTGGGGGGTCACCCTGAGCCC C ADGRE5/NM_078 TCTTCGACGATCGGAGCTTGGTGCTGACCTATGTGTTTACCATCCTCAACTGC 80 481 CTGCAGGGCGCCTTCCTCTACCTGCTGCACTGCCTGCTCAACAAGAAGGTTCG GGA SH2D1B/NM_053 TTGCTGCTCAAGGAAGGGGTGGATGGCAACTTTCTTTTAAGAGACAGCGAGTC 81 282 GATACCAGGAGTCCTGTGCCTCTGTGTCTCGTTTAAAAATATTGTCTAC ITGB2/NM_0002 TGACCGTGCAGGTTCTTCCCCAGTGTGAGTGCCGGTGCCGGGACCAGAGCAGA 82 11 GACCGCAGCCTCTGCCATGGCAAGGGCTTCTTGGAGTGCGGCATCTGCAGGTG HLA- TCCATATCAGAGCTGTGATCTTGAGAGCCCTCTCCTTGGCTTTCCTGCTGAGT 83 DPA1/NM_03355 CTCCGAGGAGCTGGGGCCATCAAGGCGGACCATGTGTCAACTTATGCCGCGTT 4 TG DGAT2/NM_0325 AGGTCCAAGGTGGAAAAGCAGCTACAGGTCATCTCAGTGCTCCAGTGGGTCCT 84 64 GTCCTTCCTTGTACTGGGAGTGGCCTGCAGTGCCATCCTCATGTACATATTCT GCA IGF1R/NM_0008 CATGCCTTGGTCTCCTTGTCCTTCCTAAAAAACCTTCGCCTCATCCTAGGAGA 85 75 GGAGCAGCTAGAAGGGAATTACTCCTTCTACGTCCTCGACAACCAGAACTTGC AGC TAGAP/NM_0541 TGCTCACCCTGGAGAATGACCAGAGCCTGTCATTTGAAGCCCAGAAGGACCTG 86 14 AACAACAAGGTGAAGACACTGGTGGAATTCCTCATTGATAACTGCTTTGAAAT A LMNA/NM_17070 AAGGAGCTGAAAGCGCGCAATACCAAGAAGGAGGGTGACCTGATAGCTGCTCA 87 7 GGCTCGGCTGAAGGACCTGGAGGCTCTGCTGAACTCCAAGGAGGCCGCACTGA GC NGAM1/NM_1813 CTGAACAAGTGTGGCCTGTTCATGTGCATTGCGGTCAACCTGTGTGGAAAAGC 88 51 CGGGCCCGGGGCCAAGGGCAAGGACATGGAGGAGGGCAAGGCCGCCTTCTCGA AA TIGIT/NM_1737 CAGGGGAGTACTTCTGCATCTATCACACCTACCCTGATGGGACGTACACTGGG 89 99 AGAATCTTCCTGGAGGTCCTAGAAAGCTCAGTGGCTGAGCACGGTGCCAGGTT CC ILI7F/NM_0528 AAGAGCTTCCTGCACAAAGTAAGCCACCAGCGCAACATGACAGTGAAGACCCT 90 72 GCATGGCCCAGCCATGGTCAAGTACTTGCTGCTGTCGATATTGGGGCTTGC HLA-F- CTGCAGCTGTGCTCACACCTGGAGGAAACCTCAATGGTCACATCCTCTTTCCT 91 AS1/NR_026972 GTGTCAAGAAGCATCGTCCGGGGAGGTGAGAAGAAGACAGTCCTCCCTAGAAT TG CD247/NM_1980 GGAAGGCGCTTTTCACCGCGGCCATCCTGCAGGCACAGTTGCCGATTACAGAG 92 53 GCACAGAGCTTTGGCCTGCTGGATCCCAAACTCTGCTACCTGCTGGA CD79B/NM_0010 CCAGGGCTGCGGCACAGAGCTGCGAGTCATGGGATTCAGCACCTTGGCACAGC 93 39933 TGAAGCAGAGGAACACGCTGAAGGATGGTATCATCATGATCCAGACGCTGCTG IDO2/NM_19429 CAAGCTCATGTGGACAAGATGCCCCTGCTGAGCTGCCAGTTCCTGAAGGGTCA 94 4 CCGGGAGCAGCGCCTGGCCCACCTGGTCCTGAGCTTCCTCACCATGGGTTA IL4/NM_000589 TTCCTGAAACGGCTCGACAGGAACCTCTGGGGCCTGGCGGGCTTGAATTCCTG 95 TCCTGTGAAGGAAGCCAACCAGAGTACGTTGGAAAACTTCTTGG TYROBP/NM_198 TCAGGAGCTCCAGGGTCAGAGGTCGGATGTCTACAGCGACCTCAACACACAGA 96 125 GGCCGTATTACAAATGAGCCCGAATCATGACAGTCAGCAACATGATACCTGG BTLA/NM_18178 GGAACTGGGAAATTATTTTGGGTCTTCTTCTTAATCCCATATCTGGACATCTG 97 0 GAACATCCATGGGAAAGAATCATGTGATGTACAGCTTTATATAAAGAGACAAT C AKT1/NM_00101 GTGAAGGAGGGTTGGCTGCACAAACGAGGGGAGTACATCAAGACCTGGCGGCC 98 4431 ACGCTACTTCCTCCTCAAGAATGATGGCACCTTCATTGGCTACAAGGAGCGGC CGC IL2RG/NM_0002 ATCTCTGTTGGCTCCATGGGATTGATTATCAGCCTTCTCTGTGTGTATTTCTG 99 06 GCTGGAACGGACGATGCCCCGAATTCCCACCCTGAAGAACCTAGAGGATCT POLR2A/NM_000 TCTTCCTGCGCTGCATCGAGTCCAACATGCTGACAGATATGACCCTGCAGGGC 100 937 ATCGAGCAGATCAGCAAGGTGTACATGCACTTGCCACAGACAGACAAC ITGAX/NM_0008 GTTCTAAGAACCTGCTTGGGAGCCGTGACCTCCAAAGCTCTGTGACCTTGGAC 101 87 CTGGCCCTCGACCCTGGCCGCCTGAGTCCCCGTGCCACCTTCCAGGAAAC IL1B/NM_00057 TGATAAGCCCACTCTACAGCTGGAGAGTGTAGATCCCAAAAATTACCCAAAGA 102 6 AGAAGATGGAAAAGCGATTTGTCTTCAACAAGATAGAAATCAATAACAAGCTG G CSF2RB/NM_000 GGCAGAGAAACACATAAAGAGCTCAGTGAACATCCAGATGGCCCCTCCATCCC 103 395 TCAACGTGACCAAGGATGGAGACAGCTACAGCCTGCGCTGGGAAACAATGA DDX58/NM_0143 CTGAATGTTTAATTAATCAGGAATGTGAAGAAATTCTACAGATTTGCTCTACT 104 14 AAGGGGATGATGGCAGGTGCAGAGAAATTGGTGGAATGCCTTCTCAGATC KIAA0101/NM_0 AAGGAATTGGAGAATTCTTTAGGTTGTCCCCTAAAGATTCTGAAAAAGAGAAT 105 14736 CAGATTCCTGAAGAGGCAGGAAGCAGTGGCTTAGGAAAAGCAAAGAGAAAAGC A CD274/NM_0141 TGGTGGTGCCGACTACAAGCGAATTACTGTGAAAGTCAATGCCCCATACAACA 106 43 AAATCAACCAAAGAATTTTGGTTGTGGATCCAGTCACCTCTGAACATGAACTG AC LAMP3/NM_0143 AGTGCGTGAGTGAACAGAGCCTCCAGTTGTCAGCCCACCTGCAGGTGAAAACA 107 98 ACCGATGTCCAACTTCAAGCCTTTGATTTTGAAGATGACCACTTTGGAAATGT GG TNFAIP8/NM_01 TTATGCACTCCGAAGCAGAAGAATCCAAGGAAGTGGCCACAGATGTCTTTAAT 108 4350 TCCAAAAACCTGGCCGTTCAGGCACAAAAGAAGATCTTGGGTAAAATGGTGTC C FOXP3/NM_0140 GAAGGGCAGGGCACAATGTCTCCTCCAGAGAGAGATGGTACAGTCTCTGGAGC 109 09 AGCAGCTGGTGCTGGAGAAGGAGAAGCTGAGTGCCATGCAGGCCCACCTGGCT GGGA IL12A/NM_0008 GACCATGAATGCAAAGCTTCTGATGGATCCTAAGAGGCAGATCTTTCTAGATC 110 82 AAAACATGCTGGCAGTTATTGATGAGCTGATGCAGGCCCTGAATTTCAAC SAMHD1/NM_015 ATATTTTGTTCAGTGGTGTGCAGACAGAAATTTCACCAAGCCGCAGGATGGCG 111 474 ATGTTATAGCCCCACTCATAACACCTCAAAAAAAGGAATGGAACGACAGTACT SIT1/NM_014450 TCTGCAGCCATGAGCAGAGGCGACAACTGCACGGATCTACTCGCACTGGGAAT 112 CCCCTCCATAACCCAGGCCTGGGGACTGTGGGTCCTCTTAGGGGCTGTGACGC CD3E/NM_00073 CTGGGGGCTTGCTGCTGCTGGTTTACTACTGGAGCAAGAATAGAAAGGCCAAG 113 3 GCCAAGCCTGTGACACGAGGAGCGGGTGCTGGCGGCAGGCAAAGGGGACA ICOSLG/NM_015 GCCAATCCCTGGGATTCCAGGAGGTTTTGAGCGTTGAGGTTACACTGCATGTG 114 259 GCAGCAAACTTCAGCGTGCCCGTCGTCAGCGCCCCCCACAGCCCCTCCCAGG HGF/NM_000601 GAACATGGAAGACTTACATCGTCATATCTTCTGGGAACCAGATGCAAGTAAGC 115 TGAATGAGAATTACTGCCGAAATCCAGATGATGATGCTCATGGACCCTGG MELK/NM_01479 AAAAATCATGGATAAAAACACACTAGGGAGTGATTTGCCCCGGATCAAAACGG 116 1 AGATTGAGGCCTTGAAGAACCTGAGACATCAGCATATATGTCAACTCT IGSF6/NM_0058 TTGAGTTCCACATGCAGAGCAGATGCGACAGCTAGAAGTGAGTAGGGCCCAGA 117 49 CCCTGGCCCAGGAAGATCCACTAAAGGAGGCCATCCTTCCGCCTTCTTCTGCA GGAGTCAGGATGGAAAGGCAGATGTAAAGTCCCTCATGGCGAAATATAACACG GGGGGCAACCCGACAGAGGATGTCTCAGTCAATAGCCGACCCTTCAGAGTCAC AGGGCCAAACTCATCTTCAGGAATACAAGCAAGAAAGAACTTATTCAACAACC AAGGAAATGCCAGCCCTCCTGCAGGACCCAGCAATGTACCTAAGTTTGGGTCC CCAAAGCCACCTGTGGCAGTCAAACCTTCTTCTGAGGAAAAGCCTGACAAGGA ACCCAAGCCCCCGTTTCTAAAGCCCACTGGAGCAGGCCAAAGATTCGGAACAC CAGCCAGCTTGACCACCAGAGACCCCGAGGCGAAAGTGGGATTTCTGAAACCT GTAGGCCCCAAGCCCATCAACTTGCCCAAAGAAGATTCCAAACCTACATTTCC CTGGCCTCCTGGAAACAAGCCATCTCTTCACAGTGTAAACCAAGACCATGACT TAAAGCCACTAGGCCCGAAATCTGGGCCTACTCCTCCAACCTCAGAAAATGAA CAGAAGCAAGCGTTTCCCAAATTGACTGGGGTTAAAGGGAAATTTATGTCAGC ATCACAAGATCTTGAACCCAAGCCCCTCTTCCCCAAACCCGCCTTTGGCCAGA AGCCGCCCCTAAGTACCGAGAACTCCCATGAAGACGAAAGCCCCATGAAGAAT GTGTCTTCATCAAAAGGGTCCCCAGCTCCCCTGGGAGTCAGGTCCAAAAGCGG CCCTTTAAAACCAGCAAGGGAAGACTCAGAAAATAAAGACCATGCAGGGGAGA TTTCAAGTTTGCCCTTTCCTGGAGTGGTTTTGAAACCTGCTGCGAGCAGGGGA GGCCCAGGTCTCTCCAAAAATGGTGAAGAAAAAAAGGAAGATAGGAAGATAGA TGCTGCTAAGAACACCTTCCAGAGCAAAATAAATCAGGAAGAGTTGGCCTCAG GGACTCCTCCTGCCAGGTTCCCTAAGGCCCCTTCTAAGCTGACAGTGGGGGGG CCATGGGGCCAAAGTCAGGAAAAGGAAAAGGGAGACAAGAATTCAGCCACCCC GAAACAGAAGCCATTGCCTCCCTTGTTTACCTTGGGTCCACCTCCACCAAAAC CCAACAGACCACCAAATGTTGACCTGACGAAATTCCACAAAACCTCTTCTGGA AACAGTACTAGCAAAGGCCAGACGTCTTACTCAACAACTTCCCTGCCACCACC TCCACCATCCCATCCGGCCAGCCAACCACCATTGCCAGCATCTCAC GNLY/NM_00643 TCCTGCCCGTGCCTGGCCCAGGAGGGCCCCCAGGGTGACCTGTTGACCAAAAC 118 3 ACAGGAGCTGGGCCGTGACTACAGGACCTGTCTGACGATAGTCCAAAAACTGA TD02/NM_00565 GCCTTTTCACCATGAGTGGGTGCCCATTTTTAGGAAACAACTTTGGATATACT 119 1 TTTAAAAAACTCCCCGTAGAAGGCAGCGAAGAAGACAAATCACAAACTGGTGT G KRT7/NM_00555 TCATCGACAAGGTGCGGTTCCTGGAGCAGCAGAACAAGGTTCTGGACACCAAG 120 6 TGGACCCTGCTGCAGGAGCAGGGCACCAAGACTGTGAGGCAGAACCTGGAGCC GTTGTTCGAGCAGTACATCAACAACCTCAGGAGGCAGCTGGACAGCATCGTGG GGGAACGGGGCCGCCTGGACTCAGAGCTGAGAAACATGCAGGACCTGGTGGAA GACTTCAAGAACAAGTATGAGGATGAAATCAACAAGCGTACCACTGCTGAGAA TGAGTTTGTGATGCTGAAGAAGGATGTAGATGCTGCCTACATGAACAAGGTGG AGCTGGAGGCCAAGGTTGATGCACTGATGGATGAGATTAACTTCATGAAGATG TTCTTTGATGCGGAGCTGTCCCAGATGCAGACGCATGTCTCTGACACCTCAGT GGTCCTCTCCATGGACAACAACCGCAACCTGGAC HLA- TCTGAGGCGGAGCACCAGAGAGCCTACCTGGAAGACACATGCGTGGAGTGGCT 121 E/NM_005516 CCACAAATACCTGGAGAAGGGGAAGGAGACGCTGCTTCACCTGGAGCCCCCAA AG HLA- CTGCTGGAGAATGTGCTGTGTGGCGTGGCCTTTGGCCTGGGTGTGCTGGGCAT 122 DMA/NM_006120 CATCGTGGGCATTGTTCTCATCATCTACTTCCGGAAGCCTTGCTCAGGTGAC LAMP1/NM_0055 ATGGTGAAAAATGGCAACGGGACCGCGTGCATAATGGCCAACTTCTCTGCTGC 123 61 CTTCTCAGTGAACTACGACACCAAGAGTGGCCCTAAGAACATGACCTTTGACC TGCCA NTN3/NM_00618 CCCTGGCCGTGCCCTGAGTGACCGTCGGGCTTGCAGGGCCTGCGACTGTCACC 124 1 CGGTTGGTGCTGCTGGCAAGACCTGCAACCAGACCACAGGCC CD28/NM_00613 AGAGCAATGGAACCATTATCCATGTGAAAGGGAAACACCTTTGTCCAAGTCCC 125 9 CTATTTCCCGGACCTTCTAAGCCCTTTTGGGTGCTGGTGGTGGTTGGTGG TARP/NM_00100 CAATTGTTCAAAAGATGCAAATGATACACTACTGCTGCAGCTCACAAACACCT 126 3806 CTGCATATTACATGTACCTCCTCCTGCTCCTCAAGAGTGTGGTCTATT EGFR/NM_00522 GGCCCGAGACCCCCAGCGCTACCTTGTCATTCAGGGGGATGAAAGAATGCATT 127 8 TGCCAAGTCCTACAGACTCCAACTTCTACCGTGCCCTGATGGATGAAGAA CCR4/NM_00550 GCCTCACAGACCTTCCTCAGAGCCGCTTTCAGAAAAGCAAGCTGCTTCTGGTT 128 8 GGGCCCAGACCTGCCTTGAGGAGCCTGTAGAGTTAAAAAATGAACCCCACGGA MAGEA3/NM_005 CGGCCTGACGTCGGTGGAGGGAAGCAGGCGCAGGCTCCGTGAGGAGGCAAGGT 129 362 TCTGAGGAGACAGGCCCCGGAGCAGCACTAGCTCCTGCCCACACTCCTACC BATF/NM_00639 CAGCCATGCCTCACAGCTCCGACAGCAGTGACTCCAGCTTCAGCCGCTCTCCT 130 9 CCCCCTGGCAAACAGGACTCATCTGATGATGTGAGAAGAGTTCAGAGGAGGGA GA KLRG1/NM_0058 GGTCTGAGGAACAATTCTGGCTGGAGGTGGGAAGATGGATCACCTCTAAACTT 131 10 CTCAAGGATTTCTTCTAATAGCTTTGTGCAGACATGCGGTGCCATCAACAAAA AT IRS1/NM_00554 TAAGCGCCTATGCCAGCATCAGTTTCCAGAAGCAGCCAGAGGACCGTCAGTAG 132 4 CTCAACTGGACATCACAGCAGAATGAAGACCTAAATGACCTCAGCAA CSF1R/NM_0052 AGGGCAACAGTTATACTTTCATCGACCCCACGCAGCTGCCTTACAACGAGAAG 133 11 TGGGAGTTCCCCCGGAACAACCTGCAGTTTGGTAAGACCCTCGGAGCTGGAGC CTLA4/NM_0052 GGCAACGGAACCCAGATTTATGTAATTGATCCAGAACCGTGCCCAGATTCTGA 134 14 CTTCCTCCTCTGGATCCTTGCAGCAGTTAGTTCGGGGTTGTTTTTTTATAGC TNFSF18/NM_00 GATCATCCTGGAAGCTGTGGCTCTTTTGCTCAATAGTTATGTTGCTATTTCTT 135 5092 TGCTCCTTCAGTTGGCTAATCTTTATTTTTCTCCAATTAGAGACTGC POU2AF1/NM_00 AAAAGGCAACATCCTGTCACAGGCCATGCTCTGGCAAAAACCCACAGCTCCGG 136 6235 AGCAAGCCCCAGCCCCGGCCCGGCCATACCAGGGCGTCCGTGTGAAGGAGCCA G GZMA/NM_00614 GGTCCTACTTAGTCTTGACAGAAAAACCATCTGTGCTGGGGCTTTGATTGCAA 137 4 AAGACTGGGTGTTGACTGCAGCTCACTGTAACTTGAACAAAAGGTCC PIK3CA/NM_006 AAGAGTACCTTGTTCCAATCCCAGGTGGAATGAATGGCTGAATTATGATATAT 138 218 ACATTCCTGATCTTCCTCGTGCTGCTCGACTTTGCCTTTCCATTTGCTCTGTT ITK/NM_005546 CTGAAGACAACAGGCGACCACTTTGGGAACCTGAAGAAACTGTGGTCATTGCC 139 TTATATGACTACCAAACCAATGATCCTCAGGAACTCGCACTGCGGCGCAACGA IFI27/NM_0055 CACATTCTCAGGAACTCTCCTTCTTTGGGTCTGGCTGAAGTTGAGGATCTCTT 140 32 ACTCTCTAGGCCACGGAATTAACCCGAGCAGGCATGGAGGCCTCTGCTCTCAC EOMES/NM_0054 AATGTGTTCGTAGAGGTGGTGCTGGCGGACCCCAACCACTGGCGCTTCCAGGG 141 42 GGGCAAATGGGTGACCTGTGGCAAAGCCGACAATAACATGCAGGGCAACAAA LCN2/NM_00556 GATCCCAGCCCCACCTCTGAGCAAGGTCCCTCTGCAGCAGAACTTCCAGGACA 142 4 ACCAATTCCAGGGGAAGTGGTATGTGGTAGGCCTGGCAGGGAATGCAATTCTC CD80/NM_00519 GCTCTTGGTGCTGGCTGGTCTTTCTCACTTCTGTTCAGGTGTTATCCACGTGA 143 1 CCAAGGAAGTGAAAGAAGTGGCAACGCTGTCCTGTGGTCACAATGTT CD83/NM_00423 CTCGGGGACATACAGGTGCACTCTGCAGGACCCGGATGGGCAGAGAAACCTAA 144 3 GTGGCAAGGTGATCTTGAGAGTGACAGGATGCCCTGCACAGCGTAAA CXCL13/NM_006 TTATCCCTAGACGCTTCATTGATCGAATTCAAATCTTGCCCCGTGGGAATGGT 145 419 TGTCCAAGAAAAGAAATCATAGTCTGGAAGAAGAACAAGTCAATTGTGTGTG MTOR/NM_00495 TTGGGGAAAGGAACGTGAAAGGCATGTTTGAGGTGCTGGAGCCCTTGCATGCT 146 8 ATGATGGAACGGGGCCCCCAGACTCTGAAGGAAACATCCTTTAATCAGG FCER1G/NM_004 CCAGCAGTGGTCTTGCTCTTACTCCTTTTGGTTGAACAAGCAGCGGCCCTGGG 147 106 AGAGCCTCAGCTCTGCTATATCCTGGATGCCATCCTGTTTCTGTATGGAATTG T TFRC/NM_00112 CAATCACACTCAGTTTCCACCATCTCGGTCATCAGGATTGCCTAATATACCTG 148 8148 TCCAGACAATCTCCAGAGCTGCTGCAGAAAAGCTGTTTGGGAATATGGAAGGA G RORC/NM_00506 TGTGCCGGGCCTACAATGCTGACAACCGCACGGTCTTTTTTGAAGGCAAATAC 149 0 GGTGGCATGGAGCTGTTCCGAGCCTTGGGCTGCAGCGAGCTCATCAGCTCCAT MMP9/NM_00499 GGCTTCTGCCCGGACCAAGGATACAGTTTGTTCCTCGTGGCGGCGCATGAGTT 150 4 CGGCCACGCGCTGGGCTTAGATCATTCCTCAGTGCCGGAGGCGCTCATGTACC CTA BST2/NM_00433 GCAACAAGAGCTGACCGAGGCCCAGAAGGGCTTTCAGGATGTGGAGGCCCAGG 151 5 CCGCCACCTGCAACCACACTGTGATGGCCCTAATGGCTTCCCTGGATGCAG PIK3CD/NM_005 GGCTCCTTCGCCATCAAGTCGCTGCGGAAACTGACGGACGATGAGCTGTTCCA 152 026 GTACCTGCTGCAGCTGGTGCAGGTGCTCAAGTACGAGTCCTACCTGGACTGCG AGCTG FCGR2B/NM_004 GGGGATCATTGTGGCTGTGGTCACTGGGATTGCTGTAGCGGCCATTGTTGCTG 153 001 CTGTAGTGGCCTTGATCTACTGCAGGAAAAAGCGGATTTCAGCTCTCCCAGGA TACCC TNFRSF14/NM_0 CCAAGTGCAGCTGGCTGGTGACGAAGGCCGGAGCTGGGACCAGCAGCTCCCAC 154 03820 TGGGTATGGTGGTTTCTCTCAGGGAGCCTCGTCATCGTCATTGTTTGCTCC OAS3/NM_00618 TTCGCCCAGCCAAGTTGAAGAACCTAATCTTGCTGGTGAAGCACTGGTACCAC 155 7 CAGGTGTGCCTACAGGGGTTGTGGAAGGAGACGCTGCCCCCGGTCTATGCCCT GGA GRAP2/NM_0048 TGATTTCACTGCTTCAGGTGAGGATGAACTGAGCTTTCACACTGGAGATGTTT 156 10 TGAAGATTTTAAGTAACCAAGAGGAGTGGTTTAAGGCGGAGCTTGGGAGCC CCNB2/NM_0047 ACTTCTTAAGGCGAGCATCAAAAGCCGGGGAGGTTGATGTTGAACAGCACACT 157 01 TTAGCCAAGTATTTGATGGAGCTGACTCTCATCGACTATGATATGGTGCATTA TCA MLANA/NM_0055 TCCTGGGAGTCTTACTGCTCATCGGCTGTTGGTATTGTAGAAGACGAAATGGA 158 11 TACAGAGCCTTGATGGATAAAAGTCTTCATGTTGGCACTCAATGTGCCTTAAC MAGEA12/NM_00 CGGCCTGACGTCGGTGGAGGGAAGCAGGCGCAGGCTCCGTGAGGAGGCAAGGT 159 5367 TCTGAGGAGACAGGCCCCGGAGCAGCACTAGCTCCTGCCCACACTCCTACC VGAM1/NM_0010 AATGGATTCTGTGCCCACAGTAAGGCAGGCTGTAAAAGAATTGCAAGTCTACA 160 78 TATCACCCAAGAATACAGTTATTTCTGTGAATCCATCCACAAAGCTGCAAG CDKN3/NM_0051 AATCGCAGATGGAGGGACTCCTGACATAGCCAGCTGCTGTGAAATAATGGAAG 161 92 AGCTTACAACCTGCCTTAAAAATTACCGAAAAACCTTAATACACTGC NCR1/NM_00482 GACGGGACTCCAGAAAGACCATGCCCTCTGGGATCACACTGCCCAGAATCTCC 162 9 TTCGGATGGGCCTGGCCTTTCTAGTCCTGGTGGCTCTAGTGTGGTTCCTGGTT G FAS/NM_000043 CAGTTGAGACTCAGAACTTGGAAGGCCTGCATCATGATGGCCAATTCTGCCAT 163 AAGCCCTGTCCTCCAGGTGAAAGGAAAGCTAGGGACTGCACAGTCAATG GZMB/NM_00413 GTGCGAATCTGACTTACGCCATTATTACGACAGTACCATTGAGTTGTGCGTGG 164 1 GGGACCCAGAGATTAAAAAGACTTCCTTTAAGGGGGACTCTGGAGGCCCTCTT G IRF9/NM_00608 CTTATCACAGTGAAGATGGAGCAGGCCTTTGCCCGATACTTGCTGGAGCAGAC 165 4 TCCAGAGCAGCAGGCAGCCATTCTGTCCCTGGTGTAGAGCCTGGGGGACC IFITM2/NM_006 CGCCTACTCCGTGAAGTCTAGGGACAGGAAGATGGTTGGCGACGTGACCGGGG 166 435 CCCAGGCCTATGCCTCCACCGCCAAGTGCCTGAACATCTGGGCCCTGATTCTG TNFSF14/NM_00 GAAGGGAAAGCTGGGGGCTCCCCACTGCACTTGCCACCTGAGTCACATTTTCA 167 3807 GAAGCCTCTGGAAAGTCGTGCACAGCCCAGGAGTGTTGAGCAATTTCGGTTTC CTCT HLA- CTGGAGAACGGGAAGGACAAGCTGGAGCGCGCTGACCCCCCAAAGACACACGT 168 B/NM_005514 GACCCACCACCCCATCTCTGACCATGAGGCCACCCTGAGGTGCTGGGCCCTGG G SDHA/NM_00416 ACATCGGAACTGCGACTCAGCATGCAGAAGTCAATGCAAAATCATGCTGCCGT 169 8 GTTCCGTGTGGGAAGCGTGTTGCAAGAAGGTTGTGGGAAAATCAGC NRP1/NM_00387 TCCTACCGAGAGTGGATACAGGTAGACTTGGGCCTTCTGCGCTTTGTCACGGC 170 3 TGTCGGGACACAGGGCGCCATTTCAAAAGAAACCAAGAAGAAATATTATGTC EBI3/NM_00575 GCTGCGCGCTTCCACCGGGTGGGGCCCATTGAAGCCACGTCCTTCATCCTCAG 171 5 GGCTGTGCGGCCCCGAGCCAGGTACTACGTCCAAGTGGCGGCTCAGGACCTCA GAG EFNA4/NM_0052 GGAGAGGAAGTCTGAGTCAGCCCATCCTGTTGGGAGCCCTGGAGAGAGTGGCA 172 27 CATCAGGGTGGCGAGGGGGGGACACTCCCAGCCCCCTCTGTCTCTTGC PVR/NM_006505 CCTGCAAGGTGGAGCACGAGAGCTTTGAGAAGCCTCAGCTGCTGACTGTGAAC 173 CTCACCGTGTACTACCCCCCAGAGGTATCCATCTCTGGCTATGATAACAACTG G BUB1/NM_00433 TATCTTCAGCTTGTGATAAAGAGTCAAATATGGAACGAAGAGTGATCACGATT 174 6 TCTAAATCAGAATATTCTGTGCACTCATCTTTGGCATCCAAAGTTGATG SKAP2/NM_0039 ATGAATAACACTCTAAGAAAGGATGGAAAGAAAGATTGCTGTTTTGAAATCTC 175 30 TGCTCCTGATAAACGTATATATCAGTTTACAGCAGCTTCTCCCAAAGATG PRF1/NM_00504 AGTTTCCATGTGGTACACACTCCCCCGCTGCACCCTGACTTCAAGAGGGCCCT 176 1 CGGGGACCTGCCCCACCACTTCAACGCCTCCACCCAGCCCGCCTACCTCAGGC CCL20/NM_0045 TCCAAAACAGACTTGGGTGAAATATATTGTGCGTCTCCTCAGTAAAAAAGTCA 177 91 AGAACATGTAAAAACTGTGGCTTTTCTGGAATGGAATTGGACATAGCCCAAGA TNFRSF18/NM_0 GACTGCATGTGTGTCCAGCCTGAATTCCACTGCGGAGACCCTTGCTGCACGAC 178 04195 CTGCCGGCACCACCCTTGTCCCCCAGGCCAGGGGGTACAGTCCCAGGGGAAAT TC CTSS/NM_00407 GAAAACCTATGGCAAACAATACAAGGAAAAGAATGAAGAAGCAGTACGACGTC 179 9 TCATCTGGGAAAAGAATCTAAAGTTTGTGATGCTTCACAACCTGGAGCATT NKG7/NM_00560 CTCCTGGTCCTTCTACCTGGGCTGGGTCTCAGCTATCCTCTTGCTCTGTACAG 180 1 GTGCCCTGAGCCTGGGTGCTCACTGTGGCGGTCCCCGTCCTGGCTATGAAACC T 1SGI5/NM_0051 CAGGAGCTTGTGCCGTGGCCCACAGCCCACAGCCCACAGCCATGGGCTGGGAC 181 01 CTGACGGTGAAGATGCTGGCGGGCAACGAATTCCAGGTGTCCCTGAGCAGCTC C PDCD1/NM_0050 GACTCCCCAGACAGGCCCTGGAACCCCCCCACCTTCTCCCCAGCCCTGCTCGT 182 18 GGTGACCGAAGGGGACAACGCCACCTTCACCTGCAGCTTCTCCAACAC SNAI1/NM_0059 TACAGCGAGCTGCAGGACTCTAATCCAGAGTTTACCTTCCAGCAGCCCTACGA 183 85 CCAGGCCCACCTGCTGGCAGCCATCCCACCTCCGGAGATCCTCAACCCCACCG CC CXCL11/NM_005 GTTGTTCAAGGCTTCCCCATGTTCAAAAGAGGACGCTGTCTTTGCATAGGCCC 184 409 TGGGGTAAAAGCAGTGAAAGTGGCAGATATTGAGAAAGCCTCCATAA CIITA/NM_0002 GGACCTGGCTGGAGAAGAAGAGATTGAGCTCTACTCAGAACCCGACACAGACA 185 46 CCATCAACTGCGACCAGTTCAGCAGGCTGTTGTGTGACATGGAAGGTGAT IFI35/NM_0055 TAGTTTCCAATTTGCGGATCCACTGCCCTCTGCTTGCGGGCTCTGCTCTGATC 186 33 ACCTTTGATGACCCCAAAGTGGCTGAGCAGGTGCTGCAACAAAAGGAGCACAC G TNFSF9/NM_003 GGCCCAAAATGTTCTGCTGATCGATGGGCCCCTGAGCTGGTACAGTGACCCAG 187 811 GCCTGGCAGGCGTGTCCCTGACGGGGGGCCTGAG TNFSF10/NM_00 GCAGATGCAGGACAAGTACTCCAAAAGTGGCATTGCTTGTTTCTTAAAAGAAG 188 3810 ATGACAGTTATTGGGACCCCAATGACGAAGAGAGTATGAACAGCCCCTGCTGG MMP2/NM_00453 TGAGAAGGATGGCAAGTACGGCTTCTGTCCCCATGAAGCCCTGTTCACCATGG 189 0 GCGGCAACGCTGAAGGACAGCCCTGCAAGTTTCCATTCCGCTTCCAGGGCACA TC EGR3/NM_00443 CCAACTGCCTGACAATCTGTACCCCGAGGAGATCCCCAGCGCGCTCAACCTCT 190 0 TCTCCGGCAGCAGCGACTCGGTAGTCCATTACAATCAGATGGCTACAGAGAA MAGEA1/NM_004 CTGAGGGACGGCGTAGAGTTCGGCCGAAGGAACCTGACCCAGGCTCTGTGAGG 191 988 AGGCAAGGTTTTCAGGGGACAGGCCAACCCAGAGGACAGGATTCCCTGGAGGC CACAG GDI63/NM_0042 GTCCTTCAGAGCAAGTGGCCTCTGTAATCTGCTCAGGAAACCAGTCCCAAACA 192 44 CTGTCCTCGTGCAATTCATCGTCTTTGGGCCCAACAAGGCCTACCATTCC IL6/NM_000600 AGTGAGGAACAAGCCAGAGCTGTGCAGATGAGTACAAAAGTCCTGATCCAGTT 193 CCTGCAGAAAAAGGCAAAGAATCTAGATGCAATAACCACCCCTGACCCAACC KLRF1/NM_0165 GAATATCTGGAACCGTGAATGGTATTCTCACTTTGACTTTGATCTCCTTGATC 194 23 CTGTTGGTTTCTCAGGGAGTATTGCTAAAATGCCAAAAAGGAAG B3GAT1/NM_018 CCAGTTGGGCCGGACTCTCCAAACCTGCTTCCGCAATGGGTGGGTTGTGAGTG 195 644 CTGGTAATGAGGAGCCGTGGGTGCAGCCAGCCTTGGAGATGCCGAAGAGACGG GACA C1QA/NM_01599 ACAGGAGGCAGAGGCATCATGGAGGGTCCCCGGGGATGGCTGGTGCTCTGTGT 196 1 GCTGGCCATATCGCTGGCCTCTATGGTGACCGAGGACTTGTGCCGAGCACC OAS1/NM_01681 AATTGTAAGAAGAAGCTTGGGAAGCTGCCACCTCAGTATGCCCTGGAGCTCCT 197 6 GACGGTCTATGCTTGGGAGCGAGGGAGCATGAAAACACATTTCAACACAGCCC AG IKZF2/NM_0162 GCCCAATGTGCTTATGGTACATAAAAGGAGTCACACTGGTGAACGCCCCTTCC 198 60 ACTGTAACCAGTGTGGAGCTTCTTTTACTCAGAAGGGCAACCTTCTGAG TLR9/NM_01744 CCCCTGCCCCCCAGCATGGGTTTCTGCCGCAGCGCCCTGCACCCGCTGTCTCT 199 2 CCTGGTGCAGGCCATCATGCTGGCCATGACCCTGGCCCTGGGTACCTTGCCTG CC KLF2/NM_01627 CGCGGGCTGCGGCAAGACCTACACCAAGAGTTCGCATCTGAAGGCGCATCTGC 200 0 GCACGCACACAGGTGAGAAGCCCTACCACTGCAACTGGGACGGCTGCGGCTGG GUSB/NM_00018 TTGCAGGGTTTCACCAGGATCCACCTCTGATGTTCACTGAAGAGTACCAGAAA 201 1 AGTCTGCTAGAGCAGTACCATCTGGGTCTGGATCAAAAACGCAGAAAATACG NFKBIA/NM_020 GTCAATGCTCAGGAGCCCTGTAATGGCCGGACTGCCCTTCACCTCGCAGTGGA 202 529 CCTGCAAAATCCTGACCTGGTGTCACTCCTGTTGAAGTGTGGGGCTGATGTCA IL23A/NM_0165 GAGATGGCTGTGACCCCCAAGGACTCAGGGACAACAGTCAGTTCTGCTTGCAA 203 84 AGGATCCACCAGGGTCTGATTTTTTATGAGAAGCTGCTAGGATCGGATATTT HERC6/NM_0179 CTGTGTGTGAAATGAGTAAACAATCTTTGCAAGTCCTAAAGAAGTGTTGGGCA 204 12 TTTTTGCAAGAATCTTCTCTGAATCCGCTGATCCAGATGCTTAAAGCAGCC SLAMF8/NM_020 ATGTGCTGCTGGTGGTGGTGCCTGTCTCGCTGCTCCTGATGCTGGTTACTCTC 205 125 TTCTCTGCCTGGCACTGGTGCCCCTGCTCAGGGAAAAAGAAAAAGGATGTCCA TGC ILI5/NM_00058 CACTGCCTTCGAAGTCCGGCGCCCCCCGGGAGGGAACTGGGTGGCCGCACCCT 206 5 CCCGGCTGCGGTGGCTGTCGCCCCCCACCCTGCAGCCAGGACTCGATGGAGAA TLR7/NM_01656 CAACCAGACCTCTACATTCCATTTTGGAAGAAGACTAAAAATGGTGTTTCCAA 207 2 TGTGGACACTGAAGAGACAAATTCTTATCCTTTTTAACATAATCCTAATTTCC A OAS2/NM_01681 AATGCACTGGGTCAGCTGAGTTCTGGCTCCACACCCAGCCCCGAGGTTTATGC 208 7 AGGGCTCATTGATCTGTATAAATCCTCGGACCTCCCGGGAGGAGAGTTTTCTA GCT HLA- CAAAGAAGAACATGTGATCATCCAGGCCGAGTTCTATCTGAATCCTGACCAAT 209 DRA/NM_019111 CAGGCGAGTTTATGTTTGACTTTGATGGTGATGAGATTTTCCATGTGGATATG GC CRTAM/NM_0196 GTAAGCACAAAGGAAGTGAAAGTGATTGTGCTGGCAACTCCTTTCAAGCCAAT 210 04 CCTGGAAGCTTCAGTTATCAGAAAGCAAAATGGAGAAGAACATGTTGTACTC MAGEC2/NM_016 GCAGAGGAGGCCCAGGCAGTGCCAGGAGTCAAGGCCTGTTGGATCTCATCATC 211 249 CATATCCCTGTTGATACGTTTACCTGCTGCTCCTGAAGAAGTCGTCATGCCTC CCG IGAM1/NM_0002 GCCCTGATGGGCAGTCAACAGCTAAAACCTTCCTCACCGTGTACTGGACTCCA 212 01 GAACGGGTGGAACTGGCACCCCTCCCCTCTTGGCAGCCAGTGGGCAAGAACCT CD4/NM_000616 CGCCTGTTTGAGAAGCAGCGGGCAAGAAAGACGCAAGCCCAGAGGCCCTGCCA 213 TTTCTGTGGGCTCAGGTCCCTACTGGCTCAGGCCCCTGCCTCCCTCGGCAAGG CC MAPK14/NM_139 TTGCTCAGTACCACGATCCTGATGATGAACCAGTGGCCGATCCTTATGATCAG 214 012 TCCTTTGAAAGCAGGGACCTCCTTATAGATGAGTGGAAAAGCCTGACCTATGA TG C1QB/NM_00049 CCAGGAGGCGTCTGACACAGTATGATGATGAAGATCCCATGGGGCAGCATCCC 215 1 AGTACTGATGTTGCTCCTGCTCCTGGGCCTAATCG NOTCH3/NM_000 CTGTGCCTGTCTTCCTGGGTTTGAGGGTCAGAATTGTGAAGTGAACGTGGACG 216 435 ACTGTCCAGGACACCGATGTCTCAATGGGGGGACATGCGTGGATGGCGTCAAC ACC NCR3/NM_14713 TCCCCTTCGCCAACTGGGACATCTTCCGACATGGCCTGGATGCTGTTGCTCAT 217 0 CTTGATCATGGTCCATCCAGGATCCTGTGCTCTCTGGGTGTCCCAGCCCCCTG AG STAT3/NM_1392 GGCCGAGGTCCTGAGCTGGCAGTTCTCCTCCACCACCAAGCGAGGACTGAGCA 218 76 TCGAGCAGCTGACTACACTGGCAGAGAAACTCTTGGGACCTGGTGTGAATTA TLR8/NM_13863 CCATTCTGCGCTGCTGCAAGTTACGGAATGAAAAATTAGAACAACAGAAACAT 219 6 GGAAAACATGTTCCTTCAGTCGTCAATGCTGACCTGCATTTTCCTGCT CYBB/NM_00039 TGCAGATCTGCTGCAACTGCTGGAGAGCCAGATGCAGGAAAGGAACAATGCCG 220 7 GCTTCCTCAGCTACAACATCTACCTCACTGGCTGGGATGAGTCTCAGGCCAAT C IKZF4/NM_0224 TAATGGAATCTTTATTTTGTGAAAGTAGCGGGGACTCATCTCTGGAGAAGGAG 221 65 TTCCTCGGGGCCCCAGTGGGGCCCTCGGTGAGCACCCCCAACAGCCAGCACTC IFIH1/NM_0221 TGCCATTGCAGATGCAACCAGAGAAGATCCATTTAAAGAGAAACTTCTAGAAA 222 68 TAATGACAAGGATTCAAACTTATTGTCAAATGAGTCCAATGTCAGATTTTGGA LCK/NM_001042 GGGGATCCCAGGATCTCACAATCTCAGGGACCATGGGCTGTGGCTGCAGCTCA 223 771 CACCCGGAAGATGACTGGATGGAAAACATCGATGTGTGTGAGAACTGCCA BCL2L11/NM_13 CAAATCAACACAAACCCCAAGTCCTCCTTGCCAGGCCTTCAACCACTATCTCA 224 8621 GTGCAATGGCTTCCATGAGGCAGGCTGAACCTGCAGATATGCGCCCAGAGATA TGG ITGAM/NM_0011 GGAGCGCTTGCCCTCTCACTCCGACTTTCTGGCTGAGCTTCGGAAGGCCCCCG 225 45808 TGGTGAACTGCTCCATCGCTGTCTGCCAGAGAATCCAGTGTGACATCCCGTTC ITGB7/NM_0008 GAGAAGGAGCAGCAACAACTCAACTGGAAGCAGGACAGTAATCCTCTCTACAA 226 89 AAGTGCCATCACGACCACCATCAATCCTCGCTTTCAAGAGGCAGACAGTCCCA CTCT JCHAIN/NM_144 AGTCAAGATGAAGAACCATTTGCTTTTCTGGGGAGTCCTGGCGGTTTTTATTA 227 64 6 AGGCTGTTCATGTGAAAGCCCAAGAAGATGAAAGGATTGTTCTTGTTGACA CD209/NM_0211 GAAGTAACCGCTTCACCTGGATGGGACTTTCAGATCTAAATCAGGAAGGCACG 228 55 TGGCAATGGGTGGACGGCTCACCTCTGTTGCCCAGCTTCAAGCAGTATTGGAA CAG SLAMF7/NM_021 ATACGGTTTACTCCACTGTGGAAATACCGAAAAAGATGGAAAATCCCCACTCA 229 181 CTGCTCACGATGCCAGACACACCAAGGCTATTTGCCTATGAGAATGTTATC IL10/NM_00057 GCCTTTAATAAGCTCCAAGAGAAAGGCATCTACAAAGCCATGAGTGAGTTTGA 230 2 CATCTTCATCAACTACATAGAAGCCTACATGACAATGAAGATACGAAACTGAG A IL1A/NM_00057 GAAGCAGTGAAATTTGACATGGGTGCTTATAAGTCATCAAAGGATGATGCTAA 231 5 AATTACCGTGATTCTAAGAATCTCAAAAACTCAATTGTATGTGACTGCCC FCGR3A/NM_000 CCCTCAGTGACCCGGTGCAGCTAGAAGTCCATATCGGCTGGCTGTTGCTCCAG 232 569 GCCCCTCGGTGGGTGTTCAAGGAGGAAGACCCTATTCACCTGAGGTGTCACAG CTGGAAGAACACTGCTCTGCATAAGGTCACATATTTACAGAATGGCAAAGGCA GGAAGTATTTTCATCATAATTCTGACTTCTACATTCCAAAAGCCACACTCAAA GACAGCGGCTCCTACTTCTGCAGGGGGCTTTTTGGGAGTAAAAATGTGTCTTC AGAGACTGTGAACATCACCATCACTCAAGGTTTGGCAGTGTCAACCATCTCA IFNA17/NM_021 CAAAGACTCACTTCTATAACCACCACGAGTTGAATCAAAATTTTCAAATGTTT 233 268 TCAGCAGTGTAAAGAAGCGTCGTGTATACCTGTGCAGGCACTAG EGR2/NM_00039 CCCAATGCCGAACTGGGAGGCCCCTTTGACCAGATGAACGGAGTGGCCGGAGA 234 9 TGGCATGATCAACATTGACATGACTGGAGAGAAGAGGTCGTTGGATCTCCCA TOP2A/NM_0010 CCAACTTTGATGTGCGTGAAATTGTAAATAACATCAGGCGTTTGATGGATGGA 235 67 GAAGAACCTTTGCCAATGCTTCCAAGTTACAAGAACTTCAAGGGTACTAT C10orf54/NM_0 GCGGCCTCTACTGCTGCCTGGTGGTGGAGATCAGGCACCACCACTCGGAGCAC 236 22153 AGGGTCCATGGTGCCATGGAGCTGCAGGTGCAGACAGGCAAAGATGCACCATC CAAC FOXM1/NM_0219 CTTTGAAAGACATCTATACGTGGATTGAGGACCACTTTCCCTACTTTAAGCAC 237 53 ATTGCCAAGCCAGGCTGGAAGAACTCCATCCGCCACAACCTTTCCCTGCACGA CATGT AXL/NM_021913 TCGCCTGAAGCAGCCTGCGGACTGTCTGGATGGACTGTATGCCTTGATGTCGC 238 GGTGCTGGGAGCTAAATCCCCAGGACCGGCCAAGTTTTACAGAGCTGCGGGAA GAT MS4A1/NM_0219 GGAGACTCAGGAGTTTTGAGAGCAAAATGACAACACCCAGAAATTCAGTAAAT 239 50 GGGACTTTCCCGGCAGAGCCAATGAAAGGCCCTATTGCTATGCAATCTGGTC IF16/NM_02287 CAGCAGGCTCCGGGCTGAAGATTGCTTCTCTTCTCTCCTCCAAGGTCTAGTGA 240 3 CGGAGCCCGCGCGCGGCGCCACCATGCGGCAGAAGGCGGTATCGCTTTTCTTG TGC CD3D/NM_00073 TCATTATCGAATGTGCCAGAGCTGTGTGGAGCTGGATCCAGCCACCGTGGCTG 241 2 GCATCATTGTCACTGATGTCATTGCCACTCTGCTCCTTGCTTTGGGAGTC GPR18/NM_0010 ATGCTCATCTCTCACACAGACTTTTGATGGACAGGAGTTTCTAAGTATCATGC 242 98200 CTACCAACAAGCTGTAAAATGATCACCCTGAACAATCAAGATCAACCTG CD3G/NM_00007 GAGGAGGAATTGAACTCAGGACTCAGAGTAGTCCAGGTGTTCTCCTCCTATTC 243 3 AGTTCCCAGAATCAAAGCAATGCATTTTGGAAAGCTCCTAGCAGAGAG ZAP70/NM_0010 CGGCCCACGAGCGGATGCCCTGGTACCACAGCAGCCTGACGCGTGAGGAGGCC 244 79 GAGCGCAAACTTTACTCTGGGGCGCAGACCGACGGCAAGTTCCTGCTGAGGCC GC HMBS/NM_00019 GCTGTGGGCCAGGGGGCCTTGGGCGTGGAAGTGCGAGCCAAGGACCAGGACAT 245 0 CTTGGATCTGGTGGGTGTGCTGCACGATCCCGAGACTCTGCTTCGCTGCATCG CT IL7/NM_000880 GAGCTTGCTCCTGCTCCAGTTGCGGTCATCATGACTACGCCCGCCTCCCGCAG 246 ACCATGTTCCATGTTTCTTTTAGGTATATCTTTGGACTTCCTCCCCTGATCCT T IFIT3/NM_0010 GGAATCAGTAAGCTAAAAACAAAATCAACCGGGACCCCAGCTTTTCAGAACTG 247 31683 CAGGGAAACAGCCATCATGAGTGAGGTCACCAAGAATTCCCTGGAGAAA RB1/NM_000321 AAACAAATATTTTGCAGTATGCTTCCACCAGGCCCCCTACCTTGTCACCAATA 248 CCTCACATTCCTCGAAGCCCTTACAAGTTTCCTAGTTCACCCTTACGG PTGS2/NM_0009 AGTCTTTTAATGAGTACCGCAAACGCTTTATGCTGAAGCCCTATGAATCATTT 249 63 GAAGAACTTACAGGAGAAAAGGAAATGTCTGCAGAGTTGGAAGCACTCTATGG T TGFB1/NM_0006 AAAGTGGAGCAGCACGTGGAGCTGTACCAGAAATACAGCAACAATTCCTGGCG 250 60 ATACCTCAGCAACCGGCTGCTGGCACCCAGCGACTCGCCAGAGTGGTTATCTT NCF1/NM_00026 GTACCGCGACAGACATCACCGGCCCCATCATCCTGCAGACGTACCGCGCCATT 251 5 GCCAACTACGAGAAGACCTCGGGCTCCGAGATGGCTCTGTCCACGGGGGACGT GG TWIST1/NM_000 GGGGGCCTGGTCCATGTCCGCGTCCCACTAGCAGGCGGAGCCCCCCACCCCCT 252 474 CAGCAGGGCCGGAGACCTAGATGTCATTGTTTCCAGAGAAGGAGAAAATGG CA4/NM_000717 CCAGGCCAAACAGTTGCACCTGCACTGGTCCGACTTGCCATATAAGGGCTCGG 253 AGCACAGCCTCGATGGGGAGCACTTTGCCATGGAGATGCACATAGTACATGAG AAAG SELL/NM_00065 ACCCTTGTGCTAAGTCAAGAGGCTCAATGGGCTGCAGAAGAACTAGAGAAGGA 254 5 CCAAGCAAAGCCATGATATTTCCATGGAAATGTCAGAGCACCCAGAGGGACTT LILRB1/NM_001 GGCCGTCATCCTACTGCTCCTCCTCCTCCTCCTCCTCTTCCTCATCCTCCGAC 255 081637 ATCGACGTCAGGGCAAACACTGGACATCGACCCAGAGAAAGGCTG GDI4/NM_00059 GCACTTCCAGAGCCTGTCCGGAGCTCAGAGGTTCGGAAGACTTATCGACCATG 256 1 GAGCGCGCGTCCTGCTTGTTGCTGCTGCTGCTGCCGCTGGTGCACGTCTCTGC ALOX15B/NM_00 CACCCTGCACATCAACACACTCGCCCGGGAGCTGCTTATCGTGCCAGGGCAGG 257 1141 TGGTGGACAGGTCCACAGGCATCGGCATTGAAGGCTTCTCTGAGTTGATACAG PECAM1/NM_000 TCCAGCCATGGCTGCCATTACCTGACCAGCGCCACAGCCGGTCTCTCTGCAGG 258 442 CGCCGGGAGAAGTGACCAGAGCAATTTCTGCTTTTCACAGGGCGGGTTTCTCA ACGG NOS2/NM_00062 GGGACTGGGCAGTTCTAGACAGTCCCGAAGTTCTCAAGGCACAGGTCTCTTCC 259 5 TGGTTTGACTGTCCTTACCCCGGGGAGGCAGTGCAGCCAGCTGCAAGCCCC FASLG/NM_0006 TTTCATGGTTCTGGTTGCCTTGGTAGGATTGGGCCTGGGGATGTTTCAGCTCT 260 39 TCCACCTACAGAAGGAGCTGGCAGAACTCCGAGAGTCTACCAGCCAGATGC CD44/NM_00061 CTTTTACACCTTTTCTACTGTACACCCCATCCCAGACGAAGACAGTCCCTGGA 261 0 TCACCGACAGCACAGACAGAATCCCTGCTACCACTTTGATGAGCACTAGTGCT AC ENTPD1/NM_001 TGGCCAAGAGGAAGGTGCCTATGGCTGGATTACTATCAACTATCTGCTGGGCA 262 098175 AATTCAGTCAGAAAACAAGGTGGTTCAGCATAGTCCCATATGAAACCAATAAT C CMKLR1/NM_001 GGAGACATGGAGGAGGAAGAGGCTGAGGGGAAGATGGAAGAAGGAGGGAAGGA 263 142345 AGGAGAAATCTTTGCTTTTGGGTAATCAGGTGTTTCTAGCTGTGTACAGGGAC T CD53/NM_00104 GGTCTTGAAGGACACTGGGATCCTGTAACACAGCCCCGGATATCTGTGTTACC 264 0033 AGCCTTGTCTCGGCCACCTCAAGGATAATCACTAAATTCTGCCGAAAGGACT TNF/NM_000594 TGTAGCAAACCCTCAAGCTGAGGGGCAGCTCCAGTGGCTGAACCGCCGGGCCA 265 ATGCCCTCCTGGCCAATGGCGTGGAGCTGAGAGATAACCAGCTGGTGGTGCC CXCL8/NM_0005 TTCTGCAGCTCTGTGTGAAGGTGCAGTTTTGCCAAGGAGTGCTAAAGAACTTA 266 84 GATGTCAGTGCATAAAGACATACTCCAAACCTTTCCACCCCAAATTTATCAA CD40LG/NM_000 TGTGTTACAGTGGGCTGAAAAAGGATACTACACCATGAGCAACAACTTGGTAA 267 074 CCCTGGAAAATGGGAAACAGCTGACCGTTAAAAGACAAGGACTCTAT HLA- TCCCAGACGCGGAGGTTGGGGTCATGGCGCCCCGAAGCCTCCTCCTGCTGCTC 268 F/NM_00109847 TCAGGGGCCCTGGCCCTGACCGATACTTGGGCGGGCTCCCACTCCTTGAG 9 GATA3/NM_0010 CTGTGCAACGCCTGCGGGCTCTATCACAAAATGAACGGACAGAACCGGCCCCT 269 02295 CATTAAGCCCAAGCGAAGGCTGTCTGCAGCCAGGAGAGCAGGGACGTCCTGTG CG LYZ/NM_000239 AAGGTGTGAGTTGGCCAGAACTCTGAAAAGATTGGGAATGGATGGCTACAGGG 270 GAATCAGCCTAGCAAACTGGATGTGTTTGGCCAAATGGGAGAGTGGTTA ARG1/NM_00004 AAAGGCTGGTCTGCTTGAGAAACTTAAAGAACAAGAGTGTGATGTGAAGGATT 271 5 ATGGGGACCTGCCCTTTGCTGACATCCCTAATGACAGTCCCTTTCAAATTG IL2RB/NM_0008 CCCAGCTGAGCTCAGAGCATGGAGGAGACGTCCAGAAGTGGCTCTCTTCGCCC 272 78 TTCCCCTCATCGTCCTTCAGCCCTGGCGGCCTGGCACCTGAGATCTCGCCACT AG NECTIN2/NM_00 TGCCGTGGGCATGGGCCGCGCTGAGCAGGTCATCTTTGTCCGAGAGACCCCCA 273 1042724 ACACAGCAGGCGCAGGGGCCACAGGCGGCATCATCGGGGGCATCATCGCCGCC AT MPO/NM_000250 GCCCGGAAGATCGTGGGGGCCATGGTCCAGATCATCACTTACCGGGACTACCT 274 GCCCCTGGTGCTGGGGCCAACGGCCATGAGGAAGTACCTGCCCACGTACCGTT CC CCR2/NM_00112 TGCTGAGAAGCCTGACATACCAGGACTGCCTGAGACAAGCCACAAGCTGAACA 275 3396 GAGAAAGTGGATTGAACAAGGACGCATTTCCCCAGTACATCCACA BRCA2/NM_0000 CTGTACTTCAGGGCCGTACACTGCTCAAATCATTCCTGGTACAGGAAACAAGC 276 59 TTCTGATGTCTTCTCCTAATTGTGAGATATATTATCAAAGTCCTTTATCACT ADORA2A/NM_00 GCCATCCGCATCCCGCTCCGGTACAATGGCTTGGTGACCGGCACGAGGGCTAA 277 0675 GGGCATCATTGCCATCTGCTGGGTGCTGTCGTTTGCCATCGGCCTGACT G6PD/NM_00040 GACGTCCGTGATGAGAAGGTCAAGGTGTTGAAATGCATCTCAGAGGTGCAGGC 278 2 CAACAATGTGGTCCTGGGCCAGTACGTGGGGAACCCCGATGGAGAGGGCGAGG CCAC TAP1/NM_00059 CTGGACCACTAGTATTTCAGGTATGCTGCTGAAAGTGGGAATCCTCTACATTG 279 3 GTGGGCAGCTGGTGACCAGTGGGGCTGTAAGCAGTGGGAACCTTGTCACATT MX1/NM_001178 CGAAGTGGACATCGCAAAAGCTGATCCAGCTGCTGCATCCCACCCTCTATTAC 280 046 TGAATGGAGATGCTACTGTGGCCCAGAAAAATCCAGGCTCGGTGGCTGAGA HLA- GCTGAGCACCCCAGTGGCTGAGGCCAGAGACTTTCCCAAGGATTTCTTGGTCC 281 DQB2/NM_00119 AGTTTAAGGGCATGTGCTACTTCACCAACGGGACAGAGCGCGTGCGCG 8858 CD27/NM_00124 CCGGGGGTCTCCTTCTCTCCTGACCACCACACCCGGCCCCACTGTGAGAGCTG 282 2 TCGGCACTGTAACTCTGGTCTTCTCGTTCGCAACTGCACCATCACTGCCAATG CT CD276/NM_0010 CATCCCTGAGGCTGCAGCGCGTGCGTGTGGCGGACGAGGGCAGCTTCACCTGC 283 24736 TTCGTGAGCATCCGGGATTTCGGCAGCGCTGCCGTCAGCCTGCAGGTGGCCGC TCC STAT4/NM_0031 GGGACTGTGAGGGGCGCTTCTGACTTTGGACTTGAGCACTGCCTGGGACCTGT 284 51 GCTGAGAGAGCGCTAGCATGTCTCAGTGGAATCAAGTCCAACAGTTAGAAATC A PTPN7/NM_0011 TCTGCGCACTGCTGGACACCCCCTTACCCGCTGGGCCCTTCAGCGCCAGCCAC 285 99797 CCAGCCCCAAGCAACTGGAAGAAGAATTCTTGAAGATCCCTTCAAACTTTGTC AGCC PTPRC/NM_0028 GACAACCACCCTCAGCCTTGCACACCACAGCTCTGCTGCCTTACCTGCACGCA 286 38 CCTCCAACACCACCATCACAGCGAACACCTCAGATGCCTACCTTAATGCCTCT G PSMB9/NM_0028 ATCAGCTATAAATATCGAGAGGACTTGTCTGCACATCTCATGGTAGCTGGCTG 287 00 GGACCAACGTGAAGGAGGTCAGGTATATGGAACCCTGGGAGGAATGCTGACTC G CD244/NM_0011 CTCAGGACTGTCAGAATGCCCATCAGGAATTCAGATTTTGGCCGTTTTTGGTG 288 66663 ATCATCGTGATTCTAAGCGCACTGTTCCTTGGCACCCTTGCCTGCTTCTGTG CXCR4/NM_0034 GGGGATCAGTATATACACTTCAGATAACTACACCGAGGAAATGGGCTCAGGGG 289 67 ACTATGACTCCATGAAGGAACCCTGTTTCCGTGAAGAAAATGCTAATTTCAAT AA MAPK1/NM_0027 TTTGTCAGGACAAGGGCTCAGAGGACTGGACGTGCTCAGACATCGGTGTTCTT 290 45 CTTCCCAGTTCTTGACCCCTGGTCCTGTCTCCAGCCCGTCTTGGCTTA TP63/NM_00372 ATCCAGCGTTTCGTAGAAACCCCAGCTCATTTCTCTTGGAAAGAAAGTTATTA 291 2 CCGATCCACCATGTCCCAGAGCACACAGACAAATGAATTCCTCAGTCCAG IRF4/NM_00246 TATGCTTGTGCCCCACCTGAGTCCCAGGCTCCCGGAGTCCCCACAGAGCCAAG 292 0 CATAAGGTCTGCCGAAGCCTTGGCGTTCTCAGACTGCCGGCTGCACATCTGCC TGT CCL3/NM_00298 TCATAGCTGACTACTTTGAGACGAGCAGCCAGTGCTCCAAGCCCGGTGTCATC 293 3 TTCCTAACCAAGCGAAGCCGGCAGGTCTGTGCTGACCCCAGTGAGGAGTGGG CCL18/NM_0029 TATTCTGAAACCAGCCCCCAGTGCCCCAAGCCAGGTGTCATCCTCCTAACCAA 294 88 GAGAGGCCGGCAGATCTGTGCTGACCCCAATAAGAAGTGGGTCCA IL7R/NM_00218 TTAATACATCACACTTGCAAAAGAAGTATGTAAAAGTTTTAATGCACGATGTA 295 5 GCTTACCGCCAGGAAAAGGATGAAAACAAATGGACGCATGTGAATTTATCCAG C HLA- ACAGGATTCCTGAGCTGAAATGCAGATGACCACATTCAAGGAAGAACTTTCTG 296 DRB1/NM_00212 CCCCGGCTTTGCAGGATGAAAAGCTTTCCTGCTTGGCAGTTATTCTTCCACA 4 CEACAM8/NM_00 GTGAAATACAGAACCCAGCGAGTGCAAACTTCAGTGACCCAGTCACCCTGAAT 297 1816 GTCCTCTATGGCCCAGATGCCCCCACCATTTCCCCTTCAGACACCTATT CXCL10/NM_001 AATTTACTGAAAGCAGTTAGCAAGGAAAGGTCTAAAAGATCTCCTTAAAACCA 298 565 GAGGGGAGCAAAATCGATGCAGTGCTTCCAAGGATGGACCACACAGAGGCTG CCL2/NM_00298 GCAAGTGTCCCAAAGAAGCTGTGATCTTCAAGACCATTGTGGCCAAGGAGATC 299 2 TGTGCTGACCCCAAGCAGAAGTGGGTTCAGGATTCCATGGACCACCTGGAC SRGN/NM_00272 TGCCTTGAAGAAAAAGGACCAATGTTCGAACTACTTCCAGGTGAATCCAACAA 300 7 GATCCCCCGTCTGAGGACTGACCTTTTTCCAAAGACGAGAATCCA CD19/NM_00117 TGGGGGGCTTCTACCTGTGCCAGCCGGGGCCCCCCTCTGAGAAGGCCTGGCAG 301 8098 CCTGGCTGGACAGTCAATGTGGAGGGCAGCGGGGAGCTGTTCCGGTGGAATGT TTCGGACCTAGGTGGCCTGGGCTGTGGCCTGAAGAACAGGTCCTCAGAGGGCC CCAGCTCCCCTTCCGGGAAGCTCATGAGCCCCAAGCTGTATGTGTGGGCCAAA GACCGCCCTGAGATCTGGGAGGGAGAGCCTCCGTGTCTCCCACCGAGGGACAG CCTGAACCAGAGCCTCAGCCAGGACCTCACCATGGCCCCTGGCTCCACACTCT GGCTGTCCTGTGGGGTACCCCCTGACTCTGTGTCCAGGGGCCCCCTCTCCTGG ACCCATGTGCACCCCAAGGGGCCTAAGTCATTGCTGAGCCTAGAGCTGAAGGA CGATCGCCCGGCCAGAGATATGTGGGTAATGGAGACGGGTCTGTTGTTGCCCC GGGCCACAGCTCAAGACGCTGGAAAGTATTATTGTCACCGTGGCAACCTGACC ATGTCATTCCACCTGGAGATCACTGCTCGGCCAGTACTATGGCACTGGCTGCT GAGGACTGGTGGCTGGAAGGTCTCAGCTGTGACTTTGGCTTATCTGATCTTCT GCCTGTGTTCCCTTGTGGGCATTCTTCATCTTCAAAGAGCCCTGGTCCTGAGG AGGAAAAGAAAGCGAATGACTGACCCCACCAGGAGATTCTTCAAAGTGACGCC TCCCCCAGGAAGCGGGCCCCAGAACCAGTACGGGAACGTGCTGTCTCTCCCCA CACCCACCTCAGGCCTCGGACGCGCCCAGCGTTGGGCCGCAGGCCTGGGGGGC ACTGCCCCGTCTTATGGAAACCCGAGCAGCGACGTCCAGGCGGATGGAGCCTT GGGGTCCCGGAGCCCGCCGGGAGTGGGCCCAGAAGAAGAGGAAGGGGAGGGCT ATGAGGAACCTGACAGTGAGGAGGACTCCG ITGB1/NM_0022 CTCAATGAAAGACGATTTGGAGAATGTAAAAAGTCTTGGAACAGATCTGATGA 302 11 ATGAAATGAGGAGGATTACTTCGGACTTCAGAATTGGATTTGGCT IFITM1/NM_003 CGTGAAGTCTAGGGACAGGAAGATGGTTGGCGACGTGACCGGGGCCCAGGCCT 303 641 ATGCCTCCACCGCCAAGTGCCTGAACATCTGGGCCCTGATTCTGGGC CCL21/NM_0029 AAGGGCTCCAAAGGCTGCAAGAGGACTGAGCGGTCACAGACCCCTAAAGGGCC 304 89 ATAGCCCAGTGAGCAGCCTGGAGCCCTGGAGACCCCACCAGCCTCACCAGCGC T MRC1/NM_00243 GGAACCACAGACAATCTGTGCTCCAGAGGTTATGAAGCCATGTATACGCTACT 305 8 AGGCAATGCCAATGGAGCAACCTGTGCATTCCCGTTCAAGTTTGAAAACAAG PGF/NM_002632 GCCCATCCTGTGTCTCCCTGCTGCGCTGCACCGGCTGCTGCGGCGATGAGAAT 306 CTGCACTGTGTGCCGGTGGAGACGGCCAATGTCACCATGCAGCTCCT ITGAL/NM_0022 CCAGAACACCTATCTGAGTGGCCTGTGTTACCTCTTCCGCCAGAATCTGCAGG 307 09 GTCCCATGCTGCAGGGGCGCCCTGGTTTTCAGGAATGTATCAAGGGCAAC ID2/NM_002166 TGTGGCTGAATAAGCGGTGTTCATGATTTCTTTTATTCTTTGCACAACAACAA 308 CAACAACAAATTCACGGAATCTTTTAAGTGCTGAACTTATTTTTCAA CD22/NM_00177 CGTGGGCCCGGGAAGGTCGGAAGAAGTGTTCCTGCAAGTGCAGTATGCCCCGG 309 1 AACCTTCCACGGTTCAGATCCTCCACTCACCGGCTGTGGAGGGAAGTCAAGTC G CCL17/NM_0029 TCTGAGGACTGCTCCAGGGATGCCATCGTTTTTGTAACTGTGCAGGGCAGGGC 310 87 CATCTGTTCGGACCCCAACAACAAGAGAGTGAAGAATGCAGTTAAATACCTGC ITGAE/NM_0022 ATATCTTTCAACAAATCTCTATATGAGGGACTGAATGCAGAGAACCACAGAAC 311 08 TAAGATCACTGTCGTCTTCCTGAAAGATGAGAAGTACCATTCTTTGCCTATCA IL3RA/NM_0021 GACCTGGGACCTTAACAGAAATGTGACCGATATCGAGTGTGTTAAAGACGCCG 312 83 ACTATTCTATGCCGGCAGTGAACAATAGCTATTGCCAGTTTGGAGCA CCR7/NM_00183 AGGTATGCCTGTGTCAAGATGAGGTCACGGACGATTACATCGGAGACAACACC 313 8 ACAGTGGACTACACTTTGTTCGAGTCTTTGTGCTCCAAGAAGGACGTGCGG CD1C/NM_00176 TATGGTTTAAGAAGCACTGCTCATATCAGGACATCCTGTGAGACTCTTCCCCC 314 5 TGACTCCCCCATTGTGTTAAGAACCCAGCAACCCAGGAGCCTAGTACAATA MAD2L1/NM_002 TCCCTGGCCATGGCGCTGCAGCTCTCCCGGGAGCAGGGAATCACCCTGCGCGG 315 358 GAGCGCCGAAATCGTGGCCGAGTTCTTCTCATTCGGCATCAACAGCATTTTAT PYGL/NM_00286 GCAGCCTATGGATACGGCATTCGGTATGAATATGGGATTTTCAATCAGAAGAT 316 3 CCGAGATGGATGGCAGGTAGAAGAAGCAGATGATTGGCTCAGATATGGAAACC CTTGG CD40/NM_00125 CACACTGCCACCAGCACAAATACTGCGACCCCAACCTAGGGCTTCGGGTCCAG 317 0 CAGAAGGGCACCTCAGAAACAGACACCATCTGCACCTGTGAAGAAGGCTGGCA C LY9/NM_002348 ATCACCCCAACCTCACATGCACAGCCAGCAACCCTGTCAGCAGGAGTTCCCAC 318 CAGTTTCTTTCTGAGAACATCTGTTCAGGACCTGAGAGAAACACAAAG HLA- GCAGACCCTGCGCGGCTACTACAACCAGAGCGAGGCCAGTTCTCACACCCTCC 319 G/NM_002127 AGTGGATGATTGGCTGCGACCTGGGGTCCGACGGACGCCTCCTCCGCGGGTAT GAACAGTATGCCTACGATGGCAAGGATTACCTCGCCCTGAACGAGGACCTGCG CTCCTGGACCGCAGCGGACACTGCGGCTCAGATCTCCAAGCGCAAGTGTGAGG CGGCCAATGTGGCTGAACAAAGGAGAGCCTACCTGGAGGGCACGTGCGTGGAG TGGCTCCACAGATACCTGGAGAACGGGAAGGAGATGCTGCAGCGCGCGGACCC CCCCAAG TLR3/NM_00326 TTGAACTAGAAGCAATTGTTAACAGCATCAAAAGAAGCAGAAAAATTATTTTT 320 5 GTTATAACACACCATCTATTAAAAGACCCATTATGCAAAAGATTCAAGG CD48/NM_00177 GCAGCAAGAATGGCACGGTCTGCCTCAGTCCACCCTGTACCCTGGCCCGGTCC 321 8 TTTGGAGTAGAATGGATTGCAAGTTGGCTAGTGGTCACGGTGCCCACCAT STAT5A/NM_003 GGAGTACCACCAAGCCACGGGCACCCTCAGTGCCCACTTCAGGAACATGTCAC 322 152 TGAAGAGGATCAAGCGTGCTGACCGGCGGGGTGCAGAGTCCGTGACAGAGG FCRLA/NM_0011 CAAAAGGCAGACAGCGGGCACTACCACTGCAGTGGCATCTTCCAGAGCCCTGG 323 84866 TCCTGGGATCCCAGAAACAGCATCTGTTGTGGCTATCACAGTCCAAGAACTG BCL6/NM_00170 CACTCTGGAGAGAAGCCCTACAAATGCGAAACCTGCGGAGCCAGATTTGTACA 324 6 GGTGGCCCACCTCCGTGCCCATGTGCTTATCCACACTGGTGAGAAGCCCTATC C ZEB1/NM_00117 CCCCAGGTGTAAGCGCAGAAAGCAGGCGAACCCGCGGCGCAATAACGTTACAA 325 4093 ATTATAATACTGTGGTAGAAACAAATTCAGATTCAGATGATGAAGACA CCL5/NM_00298 ACTGCCCTCTGCGCTCCTGCATCTGCCTCCCCATATTCCTCGGACACCACACC 326 5 CTGCTGCTTTGCCTACATTGCCCGCCCACTGCCCCGTGCCCACATCAAGGAGT ATT IDO1/NM_00216 CTGGCCAGCTTCGAGAAAGAGTTGAGAAGTTAAACATGCTCAGCATTGATCAT 327 4 CTCACAGACCACAAGTCACAGCGCCTTGCACGTCTAGTTCTGGGATGC IL18/NM_00156 CAGCTTCGGGAAGAGGAAAGGAACCTCAGACCTTCCAGATCGCTTCCTCTCGC 328 2 AACAAACTATTTGTCGCAGGAATAAAGATGGCTGCTGAACCAGTAGAAGACAA TNFRSF9/NM_00 GTTGTTAAACGGGGCAGAAAGAAACTCCTGTATATATTCAAACAACCATTTAT 329 1561 GAGACCAGTACAAACTACTCAAGAGGAAGATGGCTGTAGCTGCCGATTTCC HIF1A/NM_0015 GGTGGATATGTCTGGGTTGAAACTCAAGCAACTGTCATATATAACACCAAGAA 330 30 TTCTCAACCACAGTGCATTGTATGTGTGAATTACGTTGTGAGTGGTATT HLA- GGTGCTGCTCACATCTGTGGTCCAGGGCAGGGCCACTCCAGAGAATTACCTTT 331 DPB1/NM_00212 TCCAGGGACGGCAGGAATGCTACGCGTTTAATGGGACACAGCGCTTCCTGGA 1 FOXO1/NM_0020 AAGGATAAGGGTGACAGCAACAGCTCGGCGGGCTGGAAGAATTCAATTCGTCA 332 15 TAATCTGTCCCTACACAGCAAGTTCATTCGTGTGCAGAATGAAGGAACTGGA CD33/NM_00177 CTTCTTTCGGATGGAGAGAGGAAGTACCAAATACAGTTACAAATCTCCCCAGC 333 2 TCTCTGTGCATGTGACAGACTTGACCCACAGGCCCAAAATCCTCATCCCTGGC S100A9/NM_002 TCTGCAAAATTTTCTCAAGAAGGAGAATAAGAATGAAAAGGTCATAGAACACA 334 965 TCATGGAGGACCTGGACACAAATGCAGACAAGCAGCTGAGCTTCGAGGAGTTC HLA- GGGGCTCCTGAGCCCATCCTTCGGGACTGGACACCTGGGCTGTCCCCCATGCA 335 DMB/NM_002118 GACCCTGAAGGTTTCTGTGTCTGCAGTGACTCTGGGCCTGGGCCTCATCATCT TC HLA- ACACGGAATGTGAAGGCCCAGTCACAGACTGACCGAGTGGACCTGGGGACCCT 336 A/NM_002116 GCGCGGCTACTACAACCAGAGCGAGGCCGGTTCTCACACCATCCAGA SNAI2/NM_0030 TACCTTGTGTTTGCAAGATCTGCGGCAAGGCGTTTTCCAGACCCTGGTTGCTT 337 68 CAAGGACACATTAGAACTCACACGGGGGAGAAGCCTTTTTCTTGCCCTCACTG TNFRSF17/NM_0 TATTTTGACAGTTTGTTGCATGCTTGCATACCTTGTCAACTTCGATGTTCTTC 338 01192 TAATACTCCTCCTCTAACATGTCAGCGTTATTGTAATGCAAGTGTGACCAAT LRP1/NM_00233 GGCCCAATGGGCTAAGCCTGGACATCCCGGCTGGGCGCCTCTACTGGGTGGAT 339 2 GCCTTCTACGACCGCATCGAGACGATACTGCTCAATGGCACAGACCGGAAGAT MAGEA4/NM_001 CGCAGCTTGAGACTGGCGGAGGGAAGCCCGCCCAGGCTCTATAAGGAGACAAG 340 011548 GTTCTGAGCAGACAGGCCAACCGGAGGACAGGATTCCCTGGAGGCCACAGAGG AGCACCAAGGAGAAGATCTGCCTGTGGGTCCCCATTGCCCAGCTTTTGCCTGC ACTCTTGCC HLA- TTCCTCCCTTCTGCTGATGAGATTTATGACTGCAAGGTGGAGCACTGGGGCCT 341 DQA1/NM_00212 GGACCAGCCTCTTCTGAAACACTGGGAGCCTGAGATTCCAGCCCCTATGTCAG 2 AGCTCACAGAGACTGTGGTCTGTGCCCTGGGGTTGTCTGTGGGCCTCATGGGC ATTGTGGTGGGCACTGTCTTCATCATCCAAGGCCTGCGTTCAGTTGGTGCTTC CAGACACCAAGGGCCATTGTGAATCCCATCCTGGAAGGGAAGGTGCATCGCCA TCTACAGGAGCAGAAGAATGGACTTGCTAAATGACCTAGCACTATTCTCTGGC CCGATTTATCATATCCCTTTTCTCCTC CD1D/NM_00176 GGCCTCCTTGAGTCAGGGAAGTCGGAACTGAAGAAGCAAGTGAAGCCCAAGGC 342 6 CTGGCTGTCCCGTGGCCCCAGTCCTGGCCCTGGCCGTCTGCTGCTGGTGTGCC A RPS6/NM_00101 ATGAGAAGCGTATGGCCACAGAAGTTGCTGCTGACGCTCTGGGTGAAGAATGG 343 0 AAGGGTTATGTGGTCCGAATCAGTGGTGGGAACGACAAACAAGGTTTCCCCA MKI67/NM_0024 CTCTTCTGACCCTGATGAGAAAGCTCAAGATTCCAAGGCCTATTCAAAAATCA 344 17 CTGAAGGAAAAGTTTCAGGAAATCCTCAGGTACATATCAAGAATGTCAAAGA GZMK/NM_00210 GTATGGCGGACATCACGTTTGTGGAGGTGTTCTGATTGATCCACAGTGGGTGC 345 4 TGACAGCAGCCCACTGCCAATATCGGTTTACCAAAGGCCAGTCTCCCACTGTG GT CD79A/NM_0017 GATGCCTGGGGGTCCAGGAGTCCTCCAAGCTCTGCCTGCCACCATCTTCCTCC 346 83 TCTTCCTGCTGTCTGCTGTCTACCTGGGCCCTGGGTGCCAGGCCCTGTGGATG CD37/NM_00177 ACCATCCAAAAGTACGGCACCAACCCCGAGGAGACCGCGGCCGAGGAGAGCTG 347 4 GGACTATGTGCAGTTCCAGCTGCGCTGCTGCGGCTGGCACTACCCGCAGGACT GGT FUT4/NM_00203 GCAGTGGTATAGCATATCCTCACATTTCTAGTGCCCTTGAGACTGTGCTATGG 348 3 AACCAATCTTGAACATACATGCATTGACTTGACAAGTTACTGAGTAAGCAGCA TA AIF1/NM_00162 TGACTTTCTCAGGATGATGCTGGGCAAGAGATCTGCCATCCTAAAAATGATCC 349 3 TGATGTATGAGGAAAAAGCGAGAGAAAAGGAAAAGCCAACAGGCCCCCCAGCC CCR1/NM_00129 ACCAGAGAGAAGCCGGGATGGAAACTCCAAACACCACAGAGGACTATGACACG 350 5 ACCACAGAGTTTGACTATGGGGATGCAACTCCGTGCCAGAAG PRDM1/NM_0011 CAAGGAATCTGCTTTTCAAGTATGCCACCAACAGTGAAGAGGTTATTGGAGTG 351 98 ATGAGTAAAGAATACATACCAAAGGGCACACGTTTTGGACCCCTAATAGGT CD47/NM_00177 CTACAGGGATATTAATATTACTTCACTACTATGTGTTTAGTACAGCGATTGGA 352 7 TTAACCTCCTTCGTCATTGCCATATTGGTTATTCAGGTGATAGCCTATATCC CD74/NM_00102 AAGCCCACTGACGCTCCACCGAAAGTACTGACCAAGTGCCAGGAAGAGGTCAG 353 5159 CCACATCCCTGCTGTCCACCCGGGTTCATTCAGGCCCAAGTGCGACGAGAACG GCA LAG3/NM_00228 GGATGTGAGCCAGGCCCAGGCTGGGACCTACACCTGCCATATCCATCTGCAGG 354 6 AACAGCAGCTCAATGCCACTGTCACATTGGCAATCATCACAGTGACTCCCAAA TC TNFRSF4/NM_00 CCTCAGAAGTGGGAGTGAGCGGAAGCAGCTGTGCACGGCCACACAGGACACAG 355 3327 TCTGCCGCTGCCGGGCGGGCACCCAGCCCCTG CD2/NM_001767 TCAATATATGATACAAAAGGAAAAAATGTGTTGGAAAAAATATTTGATTTGAA 356 GATTCAAGAGAGGGTCTCAAAACCAAAGATCTCCTGGACTTGTATCAAC CCL4/NM_00298 GCTGCCTTCTGCTCTCCAGCGCTCTCAGCACCAATGGGCTCAGACCCTCCCAC 357 4 CGCCTGCTGCTTTTCTTACACTGCGAGGAAGCTTCCTCGCAACTTT BAGE/NM_18248 GGCCTGAGCGGTAGGAGTGGGGCTGGAGCAGTAAGATGGCGGCCAGAGCGGTT 358 2 TTTCTGGCATTGTCTGCCCAGCTGCTCCAAGCCAGGCTGATGAAGGAGGAGTC CCCTGTGGTGA LEXM/NM_00111 CTCCAAGGGGTCAGGTGCAAAGGCCTGCCAGATGATTATGGGAAGCTGGAACC 359 0533 CAGTAGGTGTGGGCCGCTACCTCAACACCTGGCTGATGGAGACAAAGG CCR6/NM_00436 TTTCTACAACCAGCTTGCATTTTTTCTGCCCACAATGAGCGGGGAATCAATGA 360 7 ATTTCAGCGATGTTTTCGACTCCAGTGAAGATTATTTTG CD70/NM_00125 GCATCCAGCGCTTCGCACAGGCTCAGCAGCAGCTGCCGCTCGAGTCACTTGGG 361 2 TGGGACGTAGCTGAGCTGCAGCTGAATCACACAGGACCTCAGCAGGACCCC CDK1/NM_00178 GGTGACTGATGATCTAAGTTTCCCGAGGTTTCTCAGAGCCTCTCTGGTTCTTT 362 6 CACTGGTGACCAGCCAGCCCCTCCTCTTTCTTCCTCCGGTGCTGGCGGAAGAG CCCCC CTAG1B/NM_001 GCTGCAGATGCGGGGCCAGGGGGCCGGAGAGCCGCCTGCTTGAGTTCTACCTC 363 327 GCCATGCCTTTCGCGACACCCATGGAAGCAGAGCTGGCCCGCAGGAGCCTGGC CCAGGATGCCCCACCGCTTCCCGTGCCAGGGGTGCTTCTGAAGGAGTTCACTG TGTCCGGCAACATACTGACTATCCGACTGACTGCTGCAGACCACCGCCAACTG CAGCTCTCCATCAGCTCCTGTCTCCAGCAGCTTTC CTAG2/NM_0209 CTCTCCCCCGACCAGGGGCGGTTCTGAAGGACTTCACCGTGTCCGGCAACCTA 364 94 CTGTTTATGTCAGTTCGGGACCAGGACAGGGAAGGCGCTGGGCGGATGAGGGT GGTGGGTTGGGGGCTGGGATCCGCCTCCCCGGAGGGGCAGAAAGCTAGAGATC TCAGAACACCCAAACACAAGGTCTCAGAACAGAGACCTGGTACACCAGGCCCG CCGCCACCCGAGGGAGCCCAGGGAGATGGGTGCAGAGGTGTCGCCTTTAATGT GATGTTCTCTGCCCCTCACATTTAGCCGACTGACTGCTGCAGACCACCGCCAA CTGCAGCTCTCCATCAGCTCCTGTCTCCAGCAGCTTTC CX3CR1/NM_001 GCTGACTGCAGGGCCTGCTCCCTGCCCCCCACCTCCAGGTTGGGGCCTTCACC 365 171174 ATGGATCAGTTCCCTGAATCAGTGACAGAAAACTTTGAGTACGATGATTTGG CX3CR1/ENST00 GTGGCTGACTGGCAGATCCAGAGGTTCCCTTGGCAGTCCACGCCAGGCCTTCA 366 000541347 CCATGGATCAGTTCCCTGAATCAGTGACAGAAAACTTTGAGTACGATGATTTG G CX3CR1/ENST00 GCGTTTAAGTTGGCAGACTTGGATTTCAGGAAGAGCTCTCTGGCTTCTGGGTG 367 000435290 GAGAATGGCCAGTGGGGCCTTCACCATGGATCAGTTCCCTGAATCAGTGACAG AAAACTTTGAGTACGATGATTTGG CX3CR1/ENST00 CTCGGAACACCACAGCGACTAGAGGCCTTCACCATGGATCAGTTCCCTGAATC 368 000399220 AGTGACAGAAAACTTTGAGTACGATGATTTGG GAGEI, GAGE12I, ACCTATTATCGGCCTAGACCAAGACGCTACGTAGAGCCTCCTGAAATGATTGG 369 GAGE12F/NM_0 GCCTATGCGGCCCGAGCAGTTCAGTGATGAAGTGGAACCAGCAACACCTGAAG 01040663 AAGGGGAACC GAGE12J/NM_00 ACCTATTATCGGCCTAGACCAAGACGCTACGTAGAGCCTCCTGAAATGATTGG 370 1098406 GCCTATGCGGCCCGAGCAGTTCAGTGATGAAGTGGAACCAGCAACACCTGAAG AAGGGGAACC GAGE2C,GAGE2A, ACCTATTATCGGCCTAGACCAAGACGCTACGTAGAGCCTCCTGAAATGATTGG 371 GAGE2E/NM_00 GCCTATGCGGCCCGAGCAGTTCAGTGATGAAGTGGAACCAGCAACACCTGAAG 1472 AAGGGGAACC GAGEI0/NM_001 ACCTATTATCGGCCTAGACCAAGACGCTACGTAGAGCCTCCTGAAATGATTGG 372 098413 GCCTATGCGGCCCGAGCAGTTCAGTGATGAAGTGGAACCAGCAACACCTGAAG AAGGGGAACC GAGE13/NM_001 ACCTATTATCGGCCTAGACCAAGACGCTACGTAGAGCCTCCTGAAATGATTGG 373 098412 GCCTATGCGGCCCGAGCAGTTCAGTGATGAAGTGGAACCAGCAACACCTGAAG AAGGGGAACC IKZF1/NM_0060 GACATGTCCCAAGTTTCAGGGAAGGAAAGCCCCCCTGTAAGCGATACTCCAGA 374 60 TGAGGGCGATGAGCCCATGCCGATCCCCGAGGACCTCTCCACCACCTCGGGAG GACAGC ILI7A/NM_0021 CTCAGATTACTACAACCGATCCACCTCACCTTGGAATCTCCACCGCAATGAGG 375 90 ACCCTGAGAGATATCCCTCTGTGATCTGGGAGGC IL2/NM_000586 CTTAATCAGCAATATCAACGTAATAGTTCTGGAACTAAAGGGATCTGAAACAA 376 CATTCATGTGTGAATATGCTGATGAGACAGCA IL21/NM_02180 CTTCCACAAATGCAGGGAGAAGACAGAAACACAGACTAACATGCCCTTCATGT 377 3 GATTCTTATGAGAAAAAACCACCCAAAGA IL22/NM_02052 GACGTTCGTCTCATTGGGGAGAAACTGTTCCACGGAGTCAGTATGAGTGAGCG 378 5 CTGCTATCTGATGAAGCAGGTGCTGAACTTCAC KIR2DL2/ENST0 CCATGTCGCTCATGGTCGTCAGCATGGCGTGTGTTGGGTTCTTCTTGCTGCAG 379 0000344867 GGGGCCTGGCCACATGAGGGAGTCCACAGAAAACCTTCCCTCCTGGCCCACCC AGGTCCCCTGGTGAAATCAGAAGAGACAGTCATCCTGCAATGTTGGTCAGATG TCAGGTTTGAGCACTTCCTTCTGCACAGAGAGGGGAAGTATAAGGACACTTTG CACCTCATTGGAGAGCACCATGATGGGGTCTCCAAGGCCAACTTCTCCATCGG TCCCATGATGCAAGACCTTGCAGGGACCTACAGATGCTACGGTTCTGTTACTC ACTCCCCCTATCAGTTGTCAGCTCCCAGTGACCCTCTGGACATCGTCATCACA GGTCTATATGAGAAACCTTCTCTCTCAGCCCAGCCGGGCCCCACGGTTTTGGC AGGAGAGAGCGTGACCTTGTCCTGCAGCTCCCGGAGCTCCTATGACATGTACC ATCTATCCAGGGAGGGGGAGGCCCATGAACGTAGGTTCTCTGCAGGGCCCAAG GTCAACGGAACATTCCAGGCCGACTTTCCTCTGGGCCCTGCCACCCACGGAGG AACCTACAGATGCTTCGGCTCTTTCCGTGACTCTCCCTATGAGTGGTCAAACT CGAGTGACCCACTGCTTGTTTCTGTCACAGGAAACCCTTCAAATAGTTGGCCT TCACCCACTGAACCAAGCTCCAAAACCGGTAAC KIR2DL3/NM_01 CTATGAGTGGTCAAACTCGAGTGACCCACTGCTTGTTTCTGTCACAGGAAACC 380 5868 CTTCAAATAGTTGGCCTTCACCCACTGAACCAAGCTCCAAAACCGGTAAC MAGEA10/NM_02 TGGGACACCACAGAGCAGCACTGAAGGAGAAGACCTGCCTGTGGGTCCCCATC 381 1048 GCCCAAGTCCTGCCCACACTCCCACC MIF/NM_002415 TCCGAGCTCACCCAGCAGCTGGCGCAGGCCACCGGCAAGCCCCCCCAGTACAT 382 CGCGGTGCACGTGGTCCCGGACCAGCTCATGGCCTTCGGCGGCTCCAGCGAGC CGTGCGC PTPRCAP/ENST0 TCACTTCTCGCTCGACACAGCCAGAGCTGGAGGTGGGTGCCCGGCACGGAGGG 383 0000326294 GCCTGCGGACCAATGGCTCTGCCCTGCACCTTAGGGCTCGGGATGCTGCTGGC CCTGCCAGGGGCCTTGGGCTCGGGTGG SSX2/NM_003147 CTCGGAGAAAATCTTCTATGTGTATATGAAGAGAAAGTATGAGGCTATGACTA 384 AACTAGGTTTCAAGGCCACCCTCCCACCTTTCATGTGTAATAAACGGGCCGAA GACTTCCAGG TCF7/NM_003202 CTCTCTACGAACATTTCAACAGCCCACATCCCACCCCTGCACCTGCGGACATC 385 AGCCAGAAGCAAGTTCACAGGCCTCTGCAGACCCCTGACCTCTCTGGCTTCTA CTCCCTGACCTCAGGC XAGE1B/NM_001 AGATAATACCTAAAGAGGAACACTGTAAAATGCCAGAAGCAGGTGAAGAGCAA 386 097594 CCACAAGTTTAAATGAAGACAAGCT CEACAM8/NM_00 GTGAAATACAGAACCCAGCGAGTGCAAACTTCAGTGACCCAGTCACCCTGAAT 387 1816 GTCCTCTATGGCCCAGATGCCCCCACCATTTCCCCTTCAGACACCTATT CXCR3/NM_001504 CACCAAAGCAGAGGGGCAGGCAGCACACCACCCAGCAGCCAGAGCACCAGCCC 388 AGCCATGGTCCTTGAGGTGAGTGACCACCAAGTGCTAAATGACGCCGAGGTTG CCGCCCTCCTGGAGAACTTCA FCGR1A/NM_000 CCATTGCTCAGGCATGGGAAAGCATCGCTACACATCAGCAGGAATATCTGTCA 389 566 CTGTGAAAGAGCTATTTCCAGCTCCAGTGCTGAATGCATCTGTGACATCCCC FCGR3B/NM_000 AGCGGCTCCTACTTCTGCAGGGGGCTTGTTGGGAGTAAAAATGTGTCTTCAGA 390 570 GACTGTGAACATCACCATCACTCAAGGTTTGGCAGTGTCAACCATCTCA FYB/NM_001465 CTGGAAACAGTACTAGCAAAGGCCAGACGTCTTACTCAACAACTTCCCTGCCA 391 CCACCTCCACCATCCCATCCGGCCAGCCAACCACCATTGCCAGCATCTCAC HLA- GCAGCACGAGGGGCTGCAAGAGCCCCTCACCCTGAGCTGGGAGCCATCTTCCC 392 C/NM_002117 AGCCCACCATCCCCATCATGGGCATCGTTGCTGGCCTGGCTGTCCTGGTTG HLA- AGGAAGGTGCATCACCATCTACAGGAGAAGAAGAATGGACTTGCTAAATGACC 393 DQA2NM_020056 TAGCACTATTCTCTGGCCTGATTTATCATATCCCTTTTCTCCTC IFNG/NM_00061 GTGTGGAGACCATCAAGGAAGACATGAATGTCAAGTTTTTCAATAGCAACAAA 394 9 AAGAAACGAGATGACTTCGAAAAGCTGACTAATTATTCGGTAACTGA KIR2DL1/NM_01 CGCGGCTGCCTGTCTGCTCCGGCAGCACCATGTCGCTCTTGGTCGTCAGCATG 395 4218 GCGTGTGTTGGGTTCTTCTTGCTGCAGGGGGCCTGGCCACATGAGGGAGTCCA CAGAAAACCTTCCCTCCTGGCCCACCC KRT5/NM_00042 AACTTCATGAAGATGTTCTTTGATGCGGAGCTGTCCCAGATGCAGACGCATGT 396 4 CTCTGACACCTCAGTGGTCCTCTCCATGGACAACAACCGCAACCTGGAC LMNA/NM_17070 AAGGAGCTGAAAGCGCGCAATACCAAGAAGGAGGGTGACCTGATAGCTGCTCA 397 7 GGCTCGGCTGAAGGACCTGGAGGCTCTGCTGAACTCCAAGGAGGCCGCACTGA GC PTPN11/NM_002 CTCCCGAGTGATTGTCATGACAACGAAAGAAGTGGAGAGAGGAAAGAGTAAAT 398 834 GTGTCAAATACTGGCCTGATGAGTATGCTCTAAAAGAATATGGCGTCATGCGT G

TABLE 2 primer pair sequences of the immune response assay SEQ SEQ Gene/ ID ID Accession PRIMER1 NO: PRIMER2 NO: CD63/ TTCCATGUCGAAGAACCGAGUC 399 GCAGCUACCACCAGCACAUT 400 NM_001780 CD69/ CGTCAUGAAGGGTCCTUCCAA 401 ATGGCTGTCUGATGGCATUG 402 NM_001781 A CXCL1 CAATCCTGCAUCCCCCAUAGT 403 TGTCTCTCTTTCCUCTTCTG 404 NM_001511 TTCCUA KLRD1 GUGAACAGAAAACTUGGAACGA 405 CGGTGTGCUCCTCACTGUAA 406 NM_002262 AAGT HLA-DOB CCAUGACUCAAGGCACAGACT 407 TCCCCACAUCACTGUCGAAA 408 NM_002120 C CXCR5 TCTCAACAUAAGACAGTGACCA 409 ACCAGGGAGGUGTCGTTAUA 410 NM_001716 GUCT GT IL12B CTGGCCAGUACACCTGUCA 411 TCTTGGCCUCGCATCTUAGA 412 NM_002187 AAG PTK7 GCAAACCCGGCUACTUGGA 413 CATCATACACCUCCACGCTG 414 NM_002821 UT CEACAM1 TGTGGAATTAUCACCCATCATC 415 ACAAGATCUCCCAAGTCCTC 416 NM_001712 AUCC CAUA CXCL9 TGUTCCTGCAUCAGCACCAA 417 GCTGAATCTGGGUTTAGACA 418 NM_002416 TGTTUGAA IL13 CAGCCCUGGAATCCCUGAT 419 AAACTGGGCCACCUCGATUT 420 NM_002188 NT5E GAAATGGAUAAACTCATCGCUC 421 CCGCCCAUCATCAGAAGUGA 422 NM_002526 AGAAAG VEGFA CAGCTTGAGTUAAACGAACGTA 423 CGGCAGCGUGGTTTCTGUA 424 NM_001171623 CTUG ABCF1 CTGAGCAUCCCUCCCAACAT 425 TTGTTCCAGCUGTCCCUGAA 426 NM_001025091 G CD38 GGCCCAUCAGTUCACACAGG 427 CTGCAGTCCUTTCTCCAGUC 428 NM_001775 T JAML CUAGAGTGCUCGCAGCAGT 429 ACTGGCAGCAGGAUGAGTTU 430 NM_001098526 C S100A8 GTCTACCACAAGUACTCCCTGA 431 ACCATCAGTGTUGATATCCA 432 NM_002964 UAAAG ACTCTTUG MYC GCTTCTCUGAAAGGCTCTCCUT 433 AAAUACGGCUGCACCGAGT 434 NM_002467 IRF1 CCAGGCTACAUGCAGGACUT 435 GGGTGACACCUGGAAGTTGU 436 NM_002198 A CCL22 TGGTTGTCCUCGTCCTCCUT 437 AGTCTGAGGUCCAGTAGAAG 438 NM_002990 TGUT CXCR2 TGGAGGTGUCCTACAGGUGAAA 439 GGCAGGGUAGAGCTGTAACU 440 NM_001557 G IFITI TGCAGAACGGCUGCCTAATUT 441 CAAGACTCTGTTUTCTAAAT 442 NM_001548 CAGGCAUT IFIT2 GCAGAAGAGGAAGAUTTCUGAA 443 TCAAGTTCCAGGUGAAATGG 444 NM_001547 GAGT CAUT CD68 CAGACAGCCUAGCTGGACUT 445 CGTGAAGGAUGGCAGCAAAG 446 NM_001251 UA M6PR CTCCCAUCCCCAGAAGGGUAT 447 TCTGTCUGCCAGGATTCTCU 448 NM_002355 CA SH2D1A ACCGAGUGUCCCAGACAGA 449 TCAACTGGATACUGCAGAGG 450 NM_002351 TATUACA ISG20 GTCCACUGACAGGCTGTUGT 451 ATCGTTGCCCUCGCATCUT 452 NM_002201 GBPI GACTTTGUGTGGACACUGAGAG 453 ATACAGAGTCUGGGCAGGTU 454 NM_002053 A AAAAG TBP GCGCAAGGGUTTCTGGTTUG 455 CCAGTGCCAUAAGGCATCAT 456 NM_003194 UG STAT6 GCTGGACAGAGCUACAGACCUA 457 GCCGCUGCACTTTTTCUGG 458 NM_003153 T ID3 CTCAGCTUAGCCAGGUGGAA 459 GAGATGACAAGUTCCGGAGU 460 NM_002167 GA CX3CL1 ACGTGCAGCAAGAUGACAUCA 461 GTCCTTGACCCAUTGCTCCU 462 NM_002996 T KLRB1 AGGGTUCACCTTGGCAUCAAT 463 TGCTCUGTTGAATGUCCACA 464 NM_002258 CT TNFSF4 CGGTATCCUCGAATTCAAAGTA 465 GAGATGAGAUAAAACCCATC 466 NM_003326 UCAAAGT ACAGTUGA CD52 CCTGAGATCACCUAAAAAGCTG 467 TGGTTTGGCUGGTGTCGTUT 468 NM_001803 CUA IL10RA GTCTGTGTGGUTTGAAGCAGAA 469 GACAGGGTCUGGCTACAGUT 470 NM_001558 TTUT HLA-DOA ACCACACGGACUGAGACTGAUT 471 GTCCGUAGGAGCCCATGUG 472 NM_002119 IFNB1 TGTGCCUGGACCATAGUCAGA 473 AACAGCAUCTGCTGGTUGAA 474 NM_002176 GA CCR5 GGCCAGAAGAGCUGAGACAUC 475 GGCTGCGAUTTGCTUCACA 476 NM_001100168 IKZF3 AGTGGAAAGAUGAACTGCGAUG 477 TGGCGGAGGAGGUTACCUT 478 NM_012481 T STAT1 CGATGGGCUCAGCTTUCAGA 479 ACAAAACCUCGUCCACGGAA 480 NM_007315 T CD6 CAGAUGAGGAGGUCCAGCAAA 481 TGCTCCUCGGGTGATACUGA 482 NM_006725 BRCA1 CUGAAAGCCAGGGAGTUGGT 483 CUTGTTTCACTCUCACACCC 484 NM_007300 AGAT CorOIA TGTGCTGUCAACCCTAAGTTUG 485 CAGGCGAUGTCUAGCACAGG 486 NM_007074 T TBX21 GGAUGCGCCAGGAAGTTUCA 487 CTCTGGCUCTCCGTCGUT 488 NM_013351 KLRK1 GTGGGAAGAUGGCTCCATUCT 489 ACACAGUCCTTTGCAUGCAG 490 NM_007360 AT CXCR6 TCATCTCUGGAACAAACUGGCA 491 CTTGCTGAACUGCAGGAAGU 492 NM_006564 AA C PTEN AGCGUGCAGATAAUGACAAGGA 493 GATTTGACGGCUCCTCTACU 494 NM_000314 A GT PMEL CCUGGAACAGGCAGCTGUAT 495 TTCCAGGGAAGUTCAAGGCA 496 NM_006928 AUAG DMBT1 ACTACTCCAGUCCCTCCAAUGA 497 GCCGACACUCGTAGTAUCCA 498 NM_007329 C T IFI44L AAGTGGATGATUGCAGTGAGGU 499 CAAATCUGAAGCATAGTTUC 500 NM_006820 T CAACCAT LAPTM5 TGACCCTCUGCAGCTCCUA 501 GACAGTGAUGAAGGCGAUGG 502 NM_006762 A CD226 CGUCATCGTCAUCCCCGAT 503 CAGCCACAAAGAGGGUATAT 504 NM_006566 TGGTUA TNFSF13B GATCTTACACATUTGTTCCATG 505 GATGTCCCAUGGCGUAGGT 506 NM_006573 GCUT ICOS CTTGGACCAUTCTCAUGCCAAC 507 GCACAUCCTATGGGUAACCA 508 NM_012092 GAA CD160 AGAGCCAACAUTTGCTTCAAGT 509 GGAAGCUGAGCTGGTGAUGT 510 NM_007053 UC TRIM29 TGCAUGTUCCAGGAGCACAA 511 CTTCTCCTTCUGCCACTTCU 512 NM_012101 CA LST1 ATGAGGAACUTGAGGCAAGUCA 513 CCAGGCCCCCGUAGATACAT 514 NM_007161 AUA ZBTB46 CGGCGCUCATGAGUAAGAACA 515 CGGACACTUGAACTTCTTCC 516 NM_025224 UGAT VTCN1 CUGGTTGTGAGUCACCAAGGAA 517 TGCAAUTGCUCCAGCCAGAA 518 NM_024626 KREMENI ACAGUCACAGCCATTGUAGCA 519 GGAAATTGAAGUGGAAGGCT 520 NM_032045 TGUAAA PDCD1LG2 TGAGCCUGGAATTGCAGCUT 521 TGGCTGTTATUGCTCCAAGG 522 NM_025239 UT TUBB CTCTGTTCGCUCAGGTCCTUT 523 GCCTCCUTCCGUACCACAT 524 NM_178014 CLEC4C TCCCAAGUGCAACCTCTGUC 525 GGTGCAGAAGCUCTTGTAUC 526 NM_130441 ACT CD86 CACTATGGGACUGAGTAACATT 527 CTAGCTCACUCAGGCTTUGG 528 NM_175862 CTCUT T HAVCR2 CTACTGCUGCCGGAUCCAAA 529 TGTCCCCUGGTGGUAAGCA 530 NM_032782 GZMH GGCCTTTGUTCAGTTTCUGCAA 531 GTCCGCTCCUGTTCCTTGAU 532 NM_033423 AT NFATC1 CCCCTATUCCTGTAACGGUCAA 533 GCATACCCCUGCTGAACUGA 534 NM_172387 G CD8B CGGAAGACAGUGGCATCTACUT 535 GTAACCGGCACACUCTCTUC 536 NM_172213 T BCL2 GTGGATGACUGAGTACCUGAAC 537 GGCCAAACUGAGCAGAGTCU 538 NM_000633 C T GADD45GIP1 CGAAUGGUACCCGAGCCT 539 GCTGCCAGUTCACAATCATC 540 NM_052850 UG CBLB GCGTGTCUCTGGACAGCUA 541 GGGTCCAACUGCATCCUGAA 542 NM_170662 ITGA1 CCAGACTAUGACAGCTCTTGGA 543 CTTGGATGACCUTCTTCAGU 544 NM_181501 AUA CGAT CD8A GAACCGAAGACGUGTTUGCAAA 545 GGAAGGACUTGCTCCCUCAA 546 NM_171827 T A IL2RA GGAAGATGGATUCATACCTGCT 547 AGTTCAACATGGUTCCTTCC 548 NM_000417 GAUG TTGUAG EIF2AK2 TGGCGGUCTTCAGAAUCAACAT 549 GCTTCTGACGGUATGTATTA 550 NM_001135651 AGTUCCT MADCAM1 CACGCAGGGAGAAGUGAUCC 551 GCAGCGTTUCCAGAGGTGAU 552 NM_130760 A PTPN6 GTCGGAGUACGGGAACAUCAC 553 GCTGACCGCUGCTTCTUCA 554 NM_080548 LRG1 AGAGCTACCAUGTCCTCTUGGA 555 CGGAACACCUGGCAGTCTUT 556 NM_052972 ADGRE5 CTGGGTCTUTGGCCTGTUCA 557 GGCCCACUTCCGGTATUCT 558 NM_078481 SH2D1B ACCAAGCAAGACUGUGAGACC 559 GTGTTTCTCTCUGAAGATTC 560 NM_053282 GGTAUGT ITGB2 GCTGGGCUTCACGGACAUAG 561 TTTTCCCAATGUAGCCAGTG 562 NM_000211 UCA HLA-DPA1 GCGCCCUGAAGACAGAAUGT 563 CCTGTTGGTCUATGCGTCTG 564 NM_033554 UA DGAT2 CTTCTCTGUCACCTGGCUCAAT 565 GCGAUGAGCCAGCAAUCAG 566 NM_032564 IGFIR CTACGTGAAGAUCCGCCATUCT 567 TGGTCCCAGUCCCACAGUT 568 NM_000875 TAGAP CCAUCTGCATUGGACCCAACA 569 GCACUGGAATGTTCUCCCCA 570 NM_054114 AA LMNA TGAACTCTCCCUATGTGTGGTC 571 CCGACAUCAGAGACACACUG 572 NM_170707 UT G NCAM1 TGTGGACAUCACCTGCTACTUC 573 ATGGGCTCCUTGGACTCAUC 574 NM_181351 TIGIT GTCGCUGACCGTGAACGAUA 575 ATGGCUCCAAGCAATGGAAU 576 NM_173799 CT IL17F GCAUACACAGGAAGATACATUC 577 TGCCGCCUCACUCAGAAAG 578 NM_052872 ACAGA HLA-F-ASI ACUGAAGCGACAAGGTGUGT 579 CATCTGGTTCUCCCTGGTTC 580 NR_026972 UT CD247 CUGAGGGAAAGGACAAGAUGAA 581 TGACACCATAGAUGAAGAGG 582 NM_198053 GT ATUCCA CD79B CAACAACACCUCGGAGGTCUA 583 GGCACGAUGATGAAGAGGAU 584 NM_001039933 GAT IDO2 CAATTGATTGAUGCTCACCAGC 585 CCUCTCCTTCCUGCCAGACA 586 NM_194294 UT IL4 GCACAAGCAGCUGAUCCGA 587 CTCTCTCATGAUCGTCTTTA 588 NM_000589 GCCTUT TYROBP ACTGAGACCGAGUCGCCTUA 589 GGCTUCAGGAATGGCUGGAT 590 NM_198125 BTLA GACATTGCCUGCCATGCUT 591 TCTCCTGCUAAGATGGAGTG 592 NM_181780 TUCA AKT1 CCATGAGCGACGUGGCTAUT 593 CTCACGTUGGTCCACAUCCT 594 NM_001014431 IL2RG TGCATUGGAAGCCGTGGUT 595 AAAAGTTCCCGUGGTATTCA 596 NM_000206 GUAACA POLR2A TGGTGGACAAGAUGGATGATGA 597 CCTCCGTGAUGATGATCTTC 598 NM_000937 UG TTCUT ITGAX CCAACATCUGCCTTTACATUGA 599 GGCTCAGACUCCGGTTCTUT 600 NM_000887 CAAAC ILIB CCTGTCCUGCGTGTUGAAAGA 601 GGGAACUGGGCAGACTCAAA 602 NM_000576 UT CSF2RB CGUCTCTGTUCAGCCAAGGAG 603 TGGTCTATGTGUTCGTATCG 604 NM_000395 CATUT DDX58 CCCAACCGAUATCATTTCTGAT 605 TTGGGCCAGUTTTCCTTGUC 606 NM_014314 CUGT T KTAA0101 CCCAACUCCCAAGUGGCAAA 607 TGUGATCAGGTUGCAAAGGA 608 NM_014736 CA CD274 GTACCGCUGCATGATCAGCUA 609 GTAGCCCUCAGCCUGACAT 610 NM_014143 LAMP3 GCAGUCGGGCATTCCTUCA 611 GTGTAGUCAGACGAGCACUC 612 NM_014398 AT TNFAIP8 CTGCAGCUGGTTATCCUGACA 613 AAGGTGGUGGCGATGGATUT 614 NM_014350 FOXP3 GCGGACCAUCTTCTGGAUGA 615 AGCCTTGGUCAGTGCCATUT 616 NM_014009 IL12A GAAGATGUACCAGGTGGAGTUC 617 GATTTTTGUGGCACAGTCUC 618 NM_000882 AA ACT SAMHD1 AGAGUTTGTAUGCCGCAAGACA 619 GGCGAGTUGGATTTTGGACU 620 NM_015474 GA SITI GTGTGCTUGTGGACTCUCACA 621 GCCAGCGAGAUGAGAAAUAG 622 NM_014450 GA CD3E AATTGTCATAGUGGACATCTGC 623 GTGGTGGCCUCTCCTTGTUT 624 NM_000733 AUCA ICOSLG AGTTTCACUGCCTGGTGTUGA 625 GTACACGUGAAGGTGAGCUC 626 NM_015259 AT HGF GGACTAACAUGTTCAATGUGGG 627 GAGTGGATUTCCCGTGUAGC 628 NM_000601 ACAA A MELK CATATCCTTACUGGAGAGATGG 629 GGCTGTCUCTAGCACAUGGT 630 NM_014791 TAGCUAT IGSF6 AGAGTGACTUCAAATGACAGUG 631 TCAGGAAGCUCCGCAGTUC 632 NM_005849 CAA GNLY CACCUGCGTGAUGAGGAGAAA 633 GTGGGCTTAUCCACCATCTU 634 NM_006433 CT TD02 CTCCGUGCTTCUCAGACAGT 635 GACCTCCTTUGCTGGCTCTA 636 NM_005651 UT KRT7 CCTCAACAACAAGUTTGCCUCC 637 AGCAATCUGGGCCTCAAAGA 638 NM_005556 T UG HLA-E CCGAGCAAAAGUCAAATGAUGC 639 GGTGGTGAGUCACGTGUGT 640 NM_005516 C HLA-DMA GAACGCACUGCCCUCAGAT 641 CATCAAACTCUGGTCTGGAA 642 NM_006120 GAAUCA LAMPI GTGCGUCAGCAGCAATGTUT 643 GCACCACUGTGGCATCUGA 644 NM_005561 NTN3 CGGGAGGGCUTCTAUCGAGA 645 CCAUCCTUGCAGGGACACT 646 NM_006181 CD28 CCTCCTCCUTACCTAGACAAUG 647 CAAGCUAUAGCAAGCCAGGA 648 NM_006139 AGA CT TARP ATGTCAUCACAATGGAUCCCAA 649 ACAGCAGGUGATGAUGGCAA 650 NM_001003806 AGA EGFR GTGAGTTGAUCATCGAATTCUC 651 CCACCACGUCGTCCATGUC 652 NM_005228 CAAAAT CCR4 CCTGAGCAAGCCUGGCAUT 653 ATCGAGGGUGGTGTCTGCTA 654 NM_005508 UA MAGEA3 GAGATTCUCGCCCUGAGCAA 655 AGATCTTCUCCTTCAGTGCU 656 NM_005362 CCT BATF GTGUGAGAGCCCGGAAGATUT 657 TCTGGGCGGCAAUACGATTU 658 NM_006399 T KLRG1 AGTGAGGCCUTTTGCTGGAUT 659 ACAGCUTGAGGCTUGAAGAC 660 NM_005810 C IRSI CGCCGCUCAAGTGAGGAUT 661 GGGUACCCATGAGTUAGAAG 662 NM_005544 AGGAT CSF1R GCTGGAAGAUCATCGAGAGCTA 663 CCUCCACCACCTUCCCAAAG 664 NM_005211 UG CTLA4 CGCCATACUACCTGGGCAUA 665 TCAAAGAAACAGCUGUGAGG 666 NM_005214 AGAAA TNFSF18 CATTCAAGAACUCAAGGAGCUC 667 ACTTAGCCAUACAGGGCTCC 668 NM_005092 AGA TUA POU2AF1 CCAAACUGTCGGCTUCAAAGAG 669 TCCTCCUCAGCAGTTCCTUC 670 NM_006235 A GZMA AGTAACTCCTCAUTCAAGACCC 671 TGAGCCCCAAGAAUGACCUG 672 NM_006144 UACAT PIK3CA CCCTTATGUGACAATGTGAACA 673 AGCACCCUTTCGGCCTUT 674 NM_006218 CUCA ITK CAAAGAAGCCUCTTCCTCCTAC 675 TGUCCAGCAGGCAGTACUCT 676 NM_005546 UC IFI27 CTTAAGACGGUGAGGTCAGCUT 677 ACACTGGUCACTGCTGAUGA 678 NM_005532 G EOMES CTCAATCCCACUGCCCACUAC 679 AGGAGACTCUGGGTGAACAU 680 NM_005442 AGAT LCN2 CAGGACUCCACCUCAGACCT 681 TTTTGCGGGUCTTTGTCTTC 682 NM_005564 UCT CD80 CCAAGTGTCCAUACCTCAATTT 683 TTGUGCCAGCTCTUCAACAG 684 NM_005191 CTTUCA A CD83 CCGAAACACUACCAGCUGCAA 685 TCCGCTCTGTATUTCTTAAA 686 NM_004233 AGTCTCTUC CXCL13 TGTGUCCAAGAGAGCTCAGUCT 687 CCATTCAGCUTGAGGGUCCA 688 NM_006419 MTOR GGAAGAGGCAUCTCGTTTGUAC 689 GCCTCCATUAAATCTCGACC 690 NM_004958 T AUAGG FCERIG CGATCUCCAGCCCAAGATGAUT 691 CGACAGUAGAGGAGGGUGAG 692 NM_004106 G TFRC CACACCTGGAUTCCCTTCCUT 693 GUTTTCCAGUCAGAGGGACA 694 NM_001128148 GT RORC GGAAGTGGUGCTGGTUAGGA 695 CTTAGGGAGUGGGAGAAGUC 696 NM_005060 AAAG MMP9 CTTUGACAGCGACAAGAAGUGG 697 CCCTCAGUGAAGCGGUACA 698 NM_004994 BST2 CGCAATGUCACCCATCUCCT 699 TTTCTTTTGTCCUTGGGCCT 700 NM_004335 UCT PIK3CD CTTCCCCGAUTGCCACGUA 701 CGGUCCAGCAGGAATTUGGT 702 NM_005026 FCGR2B CTCCCAGCUCTUCACCGAT 703 CTCCCATTUCCCTGCACUCA 704 NM_004001 TNFRSF14 GGAGGAAUGUCAGCACCAGA 705 TCACACATATGAUTAGGCCA 706 NM_003820 ACUGT OAS3 TGCGGAGGAACUTTGUGAACA 707 AGGCGAAGAUGGUCAGCAAT 708 NM_006187 GRAP2 CATGGAAGCUGTTGCCAAGUT 709 AAATTCTUGGGCACATATCC 710 NM_004810 TUCCT CCNB2 GGTCGACCCUTGCCACUAC 711 AGCTGCUGCTACCTUAGAAG 712 NM_004701 GA MLANA GATCGGCAUCCTGACAGUGA 713 CCCTTCTTGUGGGCATCTTC 714 NM_005511 UT MAGEA12 GAGATTCUCGCCCUGAGCAA 715 TGGUCAGGGCAGCAGGUA 716 NM_005367 VCAM1 TGCCGAGCUAAATTACACATTG 717 CATGGUCACAGAGCCACCUT 718 NM_001078 AUGA CDKN3 GTGGCCCUGTAGGACCTUC 719 CGATGUCGCACGGTACCUG 720 NM_005192 NCRI GGCACCUACCTTTUAACCACAG 721 CTTCCTGCUGAGCCAGTCUT 722 NM_004829 A FAS GGATTGGAATUGAGGAAGACTG 723 ACGCAGUCTGGTTCAUCCC 724 NM_000043 TTACUA GZMB ATGACAGUGCAGGAAGAUCGAA 725 CTGGGCCACCUTGTUACACA 726 NM_004131 A IRF9 CTCCAGCCAUACUCCACAGAAT 727 AACAAAGAGGUGAGGTGGAA 728 NM_006084 GAUG IFITM2 TGCCTGGGCUTCATAGCAUT 729 GCAGAATGGUCATGAAGAUG 730 NM_006435 CC TNFSF14 CCAAGCGAAUGAAGCAUCCAA 731 GCCTGGGUCCTTCAACCUC 732 NM_003807 HLA-B GGAGTGGCUCCGCAGAUAC 733 GTGATCUCCGCAGGGUAGAA 734 NM_005514 A SDHA TTGAGATTTGCUGATGGAAGCA 735 GTGCTTTAGGUCTCCATAGA 736 NM_004168 UAAGA GCUT NRP1 GUGGACUCCCGGAGAGGAT 737 GAGCTAACGUCGATCTTGTA 738 NM_003873 AGTCUT EBI3 CCGTUACAAGCGUCAGGGA 739 AGTCACTCAGUTCCCCGUAG 740 NM_005755 T EFNA4 CAGGTGUCTGTCTGCUGCAA 741 GACGAAGAAUCAGAAGCAGC 742 NM_005227 AGTAAUA PVR GGUGGACGGCAAGAATGUGA 743 GCCTCATTCUGGCCAAGGUA 744 NM_006505 BUBI GGTTCAGAGCUTTCTGGAGUGA 745 CTTGCAATACAUAACAACCT 746 NM_004336 GCUCAA SKAP2 GCAATAGAUGGCTACAGTGUCA 747 AGCTGCTGUACCCATTCTUC 748 NM_003930 GA AG PRF1 GGTGGAGUGCCGCTTCUAC 749 GGGTGCCGUAGTTGGAGAUA 750 NM_005041 A CCL20 AAAAGTUGTCTGTGUGCGCAAA 751 CCCCAGCAAGGUTCTTTCUG 752 NM_004591 T TNFRSF18 GGAGTGCUGTTCCGAGUGG 753 TCGAUACACUGGAAGCCAAA 754 NM_004195 ACT CTSS CACCACUGGCATCTCUGGAA 755 AGATCGTATGAGUGCATTCC 756 NM_004079 CATUG NKG7 CCCCCAGAUCCAGACCTTCUT 757 GCTCTUGCCTTCTGCUCACA 758 NM_005601 ISG15 AGCGAACTCAUCTTTGCCAGUA 759 CUTCAGCTCUGACACCGACA 760 NM_005101 T PDCD1 GCGGCCAGGAUGGTTCTUA 761 GTTUAGCACGAAGCTCUCCG 762 NM_005018 AT SNAI1 ACCCCAAUCGGAAGCCUAAC 763 AGAUGAGCATUGGCAGCGA 764 NM_005985 CXCL11 TTGGCTGTGAUATTGTGTGCUA 765 TTTGTCACAGTUGTTACTTG 766 NM_005409 CA GGUACA CIITA GCCTCUACCACTTCTAUGACCA 767 CATAAGCCUCCCTGGTCTCT 768 NM_000246 GAT UC IFI35 CCCGGAAGUGCCTAAGTCUT 769 CGGCACUCCTCCATGTUGAT 770 NM_005533 TNFSF9 CATGTTUGCGCAGCUGGT 771 GCTCCTTCGUGTCCTCTTTG 772 NM_003811 UAG TNFSF10 CGTGTACTTUACCAACGAGCUG 773 CGGAGTUGCCACTTGACTUG 774 NM_003810 AA MMP2 GTGCUCCACCACCTACAACUT 775 TCAGTGGUGCAGCTGTCAUA 776 NM_004530 G EGR3 GTGACCAUGAGCAGTTTGCUAA 777 GGTCAGACCGAUGTCCATUA 778 NM_004430 A CA MAGEA1 AGAGAGAAGCGAGGUTTCCAUT 779 TCTCCTTGGUGCTCCTCUGT 780 NM_004988 CD163 CTGTAACTGCTCUAGGTGCTTC 781 AGGCCACAGCACUTTCTUCT 782 NM_004244 ATUAT IL6 AGTACCUCCAGAACAGATTUGA 783 TCAGCAGGCUGGCATTUGT 784 NM_000600 GAGT KLRF1 CGTTGCACTGGUATAAAATCTT 785 CCTCATACUGAGTGGCATTU 786 NM_016523 ACUGG GAACAA B3GAT1 AGCGACCCCUTCUCAGACT 787 CGATGAGGACGAUCGCUAGG 788 NM_018644 A CIQA GAGCATCCAGUTGGAGTUGACA 789 CCTCCCCUTTCTTCCCGUCT 790 NM_015991 OASI CCTAGUCAAGCACTGGUACCAA 791 TCCAAGACCGUCCGAAAUCC 792 NM_016816 IKZF2 TGTGGCAUGGTTTGCATUGG 793 CTTCTCTCCAGAGUGTAACT 794 NM_016260 TTATGUGT TLR9 GCCAGACCCUCUGGAGAAG 795 TGGAGCUCACAGGGUAGGAA 796 NM_017442 KLF2 CCACTCACACCUGCAGCUA 797 GTCUGAGCGCGCAAACUT 798 NM_016270 GUSB GCGAGUAUGGAGCAGAAACGA 799 AATTCCAAAUGAGCTCUCCA 800 NM_000181 ACCA NFKBIA GGTGTCCUTGGGTGCUGAT 801 AATAGCCCTGGUAGGTAACT 802 NM_020529 CUGT IL23A AATGATGTTCCCCAUATCCAGT 803 AGAGAAGGCUCCCCTGUGA 804 NM_016584 GUG HERO6 CTGGTGGUTCCATTUGCAAAGG 805 CTGATGAAGCAGCUGAGAGA 806 NM_017912 UGAT SLAMF8 CAGGGAAGGCCUCCUACAAAG 807 TCTCUGGACCCACTCTGUCA 808 NM_020125 IL15 GAATCCCAGCUGACUCGCT 809 CCACATGGCCAUATATTGGA 810 NM_000585 AUGGA TLR7 ACCTCTCATGCUCTGCTCTCUT 811 ACCATCUAGCCCCAAGGAGU 812 NM_016562 T OAS2 CTTTGATGTGCUTCCTGCCTUT 813 CGCUGCAGGACTGUGAAAC 814 NM_016817 HLA-DRA CGCTCAGGAAUCATGGGCUAT 815 GCCAGACCGUCTCCTTCTUT 816 NM_019111 CRTAM AAGTGCTTACAUTACAGCGACU 817 GCUTCTCATGGUGGAGCACA 818 NM_019604 CT T MAGEC2 ACACAGCCUAAAGUCAGCACA 819 CGGAAUGGAACGCCUGGAA 820 NM_016249 ICAMI GCCAACCAAUGTGCTATUCAAA 821 ACCTGGCAGCGUAGGGUA 822 NM_000201 CT CD4 AGCTCCAAGUCCTCACACAGAU 823 GGACUCCCCGGTTCATUGT 824 NM_000616 A MAPK14 GCCCTUGCACATGCCUACT 825 GTGGCACAAAGCUGATGACU 826 NM_139012 T C1QB CCUCACAGGACACCAGCTUC 827 AGCTGGGCCUGGGAGAUAT 828 NM_000491 NOTCH3 GTGGCGACCUCACTUACGA 829 AGGAGGGCACUGGCAGTTAU 830 NM_000435 A NCR3 CCCAGACCUCACTGCUCAGA 831 AGGATCCUTCCAGGGUACGA 832 NM_147130 AT STAT3 CCAATUGGAACCTGGGAUCAAG 833 CCATGTGAUCTGACACCCUG 834 NM_139276 T AA TLR8 CCACCTCCUGCATAGAGGGUA 835 CGCATAACUCACAGGAACCA 836 NM_138636 GATAUT CYBB ACACACATGCCUTTGAGTGGUT 837 CAUCATGGUGCACAGCAAAG 838 NM_000397 T IKZF4 GGCCCAAGACUCCAACCATTTU 839 TGAGUGAGCGGCUAGGAGAA 840 NM_022465 A IFIH1 ACAGGAGCCAUGCAAGAAGUT 841 GCCCATTGUTCATAGGGTUG 842 NM_022168 AGT LCK AGCTTTTCUGTGGCTGGUGAAT 843 CCATCCAGUGGGACTATGGG 844 NM_001042771 AUAA BCL2L11 CCAGCACCCAUGAGTTGUGA 845 CCGCAACUCTUGGGCGAT 846 NM_138621 ITGAM CGAGUACGUGCCACACCAA 847 CATTGAATTCUTCCTGGAUG 848 NM_001145808 CCAAA ITGB7 GCCGGGAAUACAGTCGCTUT 849 AGTGTCCCUCCCTCCTUCAG 850 NM_000889 JCHAIN TGCTCCAGTTUTTCAGAAGAAG 851 TCCGGGCACACUTACATTUG 852 NM_144646 UGA T CD209 CCTACAGCUGCAGTCTUCCA 853 AACGTTGTUGGGCTCUCCT 854 NM_021155 SLAMF7 AACAATCCUAAAGGAAGAUCCA 855 GGGAGTGCACUGCTGTCUA 856 NM_021181 GCAA IL10 CCGUGGAGCAGGUGAAGAAT 857 TCTATAGAGUCGCCACCCTG 858 NM_000572 AUG ILIA GGCTGCUGCATTACATAATCUG 859 AGCACTGGTUGGTCTTCATC 860 NM_000575 GAT UT FCGR3A GUGCCAGACAAACCTCUCCA 861 TTATGCAGAGCAGUGTTCTU 862 NM_000569 CCA IFNA17 TCACACTTTCAUGAGTTCCTCC 863 GACATCAGCAUGGTCATCTG 864 NM_021268 ATUT TAAAGUA EGR2 CCACGTCGGUGACCATCTUT 865 GGAGCAAAGCUGCTGGGAUA 866 NM_000399 TOP2A TGGGTGGUCCTGCAAAAUCC 867 ACATATTGATTUGGAGCCAG 868 NM_001067 TTCTUCA C10orf54 CAACCTGACCCUGCTGGAUA 869 GAGGAUGGGUACACCACACA 870 NM_022153 FOXM1 CACUGAGAGGAAGCGCAUGA 871 CAGACGUCUCCCGGACAA 872 NM_021953 AXL GACUATCUGCGCCAGGGAAA 873 GGCCTUCAGTGTGTTCUCCA 874 NM_021913 A MS4A1 GCCCTTGAGAUTTGAGGCCUT 875 ACATCCTCCUGAAGAGTGGT 876 NM_021950 TTUG IFI6 CGCTGCTGUGCCCATCUAT 877 GCAAGUGAAGAGCAGCAGGU 878 NM_022873 A CD3D GGACAAAGAAUCTACCGUGCAA 879 TCUCATGUCCAGCAAAGCAG 880 NM_000732 GT AA GPR18 CGACAGAAGUGGAAGTGCUGAA 881 CTGGATGUGAGCTGTUAAAA 882 NM_001098200 AA GGGA CD3G GCCACCTUCAAGGAAACCAGUT 883 CTTGAGTCUAGATTTAGGGC 884 NM_000073 UGAAAGT ZAP70 GTGGAGAAGCUCATTGCUACGA 885 GTATGTGCCCUGCTCCTUCC 886 NM_001079 HMBS GCACCCUGAGGAATGCATGUAT 887 GCCUCAGGAAGGCCCTTUC 888 NM_000190 IL7 GAGTGACTAUGGGCGGUGA 889 AGAUGATGCTACUGGCAACA 890 NM_000880 GAAC IFIT3 GGAAACCTCTUCAGCATTTGCU 891 GGCAUTTCAGCTGUGGAAGG 892 NM_001031683 T AT RBI CGGTCTTCAUGCAGAGACUGA 893 GTGAAATAUAGATGTTCCCU 894 NM_000321 CCAGGAAT PTGS2 CCAGAGCAGGCAGAUGAAAUAC 895 AGCTCCACAGCAUCGATGUC 896 NM_000963 C TGFB1 CGTCTGCUGAGGCTCAAGTUA 897 GCACAACUCCGGTGACAUCA 898 NM_000660 A NCF1 TGAUGCCCAAAGAUGGCAAGA 899 CGCTCTTCUCTACGACCUCC 900 NM_000265 A TWIST1 TTCTCGGUCTGGAGGAUGGA 901 CAGCTCCAGAGUCTCTAGAC 902 NM_000474 UGT CA4 AGGACTGCCUGCCCCAUA 903 ACATTCCTCGAUGTCCCCTU 904 NM_000717 CT SELL CCTTUGGGCAAGGACCUGAG 905 CCCCACAACUTGAAGATGTU 906 NM_000655 CCAT LILRB1 GGTTGTGAUCGGCATCTUGGT 907 CCCCUGCAGGATGTUGGAAA 908 NM_001081637 T CD14 CCGCTGTGUAGGAAAGAAGCUA 909 ACAAGGTTCUGGCGTGGUC 910 NM_000591 AA ALOX15B GAUCCCGCACACCCGAUA 911 AATAGTTCAGCUGCTTCATG 912 NM_001141 TUCCT PECAM1 GCCCCATUGTTCCCGGUT 913 GGCACUGCCCACAAGUCA 914 NM_000442 NOS2 TCTCGGCCACCUTTGAUGAG 915 ATTTGAGCTCAGAUGTTCTT 916 NM_000625 CACUGT FASLG GCCTGTGTCUCCTTGTGATGUT 917 GCTTCTCCAAAGAUGATGCT 918 NM_000639 GUGT CD44 GCAGCACTUCAGGAGGTUACAT 919 TGGTTGCUGTCTCAGTUGCT 920 NM_000610 ENTPD1 CCAGGGUGCCAGGATCATUAC 921 TCCAAAGCUCCAAAGGTTUC 922 NM_001098175 CT CMKLRI CUGTGATTCUGCCCACGGAA 923 CCAATGTGAGUCCTCAGCCA 924 NM_001142345 AUC CD53 GACAGACUGAAGAAACAUCCAA 925 TTTCCAGGCACCGUTCCUC 926 NM_001040033 GGT TNF GACAAGCCUGTAGCCCAUGT 927 ATGAGGUACAGGCCCTCUGA 928 NM_000594 T CXCL8 CTCTUGGCAGCCTTCCUGAT 929 TCCACTCTCAAUCACTCTCA 930 NM_000584 GTUCT CD40LG GAGGCCAGCAGUAAAACAACAU 931 ACAGAAGGTGACUTGGGCAT 932 NM_000074 C AGATAUA HLA-F CCCCGCGGGACUCATATTUT 933 GACACAGCGGUGCTGAAAUA 934 NM_001098479 C GATA3 GAGAUGGCACGGGACACUAC 935 GGTTGTGGUGGTCTGACAGU 936 NM_001002295 T LYZ GTCCAGGGCAAGGUCTTUGA 937 CATTGTAGTTTGUAGCTCGT 938 NM_000239 GTGTUG ARG1 GGAAGAAGGCCCUACAGTATUG 939 CCCACAGACCUTGGATTCTU 940 NM_000045 AG CA IL2RB CCAGACCCCUCGAAGTTCTTUT 941 CTTGTCCCUCTCCAGCACUT 942 NM_000878 NECTIN2 TTCGTCUGCACAGUCACCAA 943 CAGCCACAGCAGUAGCAAUG 944 NM_001042724 MPO GGGAGAGGCUCUACCAGGAA 945 GTGGGTCCACUGAGTCATTG 946 NM_000250 UA CCR2 TTCTACCTCUAGATCTGTTTGG 947 AACGAGATGUGGACAGCAUG 948 NM_001123396 TUCAGT T BRCA2 GTGGUCCACCCCAACUAAAGA 949 ACAGACTTCCUTTTGGCCAU 950 NM_000059 ACAA ADORA2A TCGCCATUGACCGCTACAUT 951 AGTTGTUCCAACCTAGCAUG 952 NM_000675 GG G6PD CGCCUCCACCAACUCAGAT 953 GGTCGUCCAGGTACCCTTUG 954 NM_000402 TAPI TGTGGCCTAUGCAGTCAACUC 955 GTGAACUGCATCTGGUAGAG 956 NM_000593 AACA MX1 TTGCAAAGAAGGAAGAUGGTTG 957 ATACTGGCUGCACAGGTUGT 958 NM_001178046 TTUC HLA-DQB2 GTGACCGUGATGCTGGUGAT 959 CGGTUATAGATGTATCUGGC 960 NM_001198858 CACAC CD27 CTGCTCAGTGUGATCCTTGCAU 961 CATUGCGACAGGCACACUC 962 NM_001242 A CD276 CTGCUGGCACAAGGCAAUG 963 TCATGCUGGGCTTCGAGUAG 964 NM_001024736 STAT4 CGTCAGCGGCUTTCTCCUA 965 TTGAUCCACCTGCUCCAAAA 966 NM_003151 ACT PTPN7 CGGGAGGUCACCCTACACUT 967 GGGATGUCCAGGTCTUCGG 968 NM_001199797 PTPRC CTACAGACCCAGUTTCCCCAUT 969 AAGGGCUCAGAGTGGTTGTU 970 NM_002838 T PSMB9 CTGCUGCAAATGTGGUGAGAAA 971 CACCAAUGGCAAAAGGCUGT 972 NM_002800 T CD244 GAAAGCCACACCCUGAATCUCA 973 TCCTTCCUCTTTCTCCUCCA 974 NM_001166663 CA CXCR4 GAACCAGCGGUTACCAUGGA 975 GTAGATGGUGGGCAGGAAGA 976 NM_003467 UT MAPK1 GATTCCAGCCAGGAUACAGATC 977 GGCTCAAAGGAGUCAAAGUG 978 NM_002745 TUAAA GA TP63 CAGTACTGCCCUGACCCTUAC 979 AAAATCCCAGAUATGCUGGA 980 NM_003722 AAACCT IRF4 GCCAGGUGACAGGAACCTUT 981 TCACGAGGAUTTCCCGGUAG 982 NM_002460 T CCL3 CCCGGCAGAUTCCACAGAAUT 983 CAGGTCGCUGACATATTTCU 984 NM_002983 GGA CCL18 GCAGATUCCACAAAAGTTCATA 985 AGCTTCAGGUCGCTGATGTA 986 NM_002988 GTUGAC TTUC IL7R AGCCAAUGACTTTGTGGUGACA 987 TGCAGGAGUGTCAGCTTUGT 988 NM_002185 T HLA-DRB1 GACACUCTGGACTUCAGCCA 989 GGTCCUGAGAAAGCCCTCUC 990 NM_002124 T CEACAM8 CCUGCCACGCCAATAACUCA 991 AGGTCAGGTUCACAGAGTCC 992 NM_001816 UT CXCL10 TCCAGAAUCGAAGGCCAUCAAG 993 TGUAGGGAAGTGAUGGGAGA 994 NM_001565 GG CCL2 CGAGCUATAGAAGAAUCACCAG 995 TCTTCGGAGUTTGGGTTTGC 996 NM_002982 CA UT SRGN GCAAUCCAGACAGTAATTCUGC 997 AAAGTGGGAAGAUACGATTC 998 NM_002727 AAAC AAGUCC CD19 CACUGCCCCGTCTTAUGGAAA 999 AGGTTGGAGUCGTTCTCAUA 1000 NM_001178098 GAACT ITGB1 CCTCTACTACCTUATGGACCTG 1001 GGCATCACAGUCTTTTCCAC 1002 NM_002211 TCTUA AAAUG IFITM1 GGCTTCATAGCAUTCGCCTACU 1003 GGATGAAUCCAATGGTCAUG 1004 NM_003641 C AGGAT CCL21 CUCCAAGACUGGCAAGAAAGGA 1005 GCATCTTGGGUTCAGGCTUC 1006 NM_002989 A MRC1 GTTTATGGAGCAGGUGGAAGAT 1007 TCGTGCAAUCTGCGUACCA 1008 NM_002438 CUAT PGF CGAGGUGGAGCACATGTUCA 1009 GGUCCCCAGAACGGATCTUT 1010 NM_002632 ITGAL GGCTGTCUCGAACGTGUGA 1011 AACCATCAAACAGAAAUACC 1012 NM_002209 AGGTCUAC ID2 ATGTCAAAUGACAGCAAAGCAC 1013 CAACTTGTCCUCCTTGTGAA 1014 NM_002166 UG AUGG CD22 CTGCTGUCAGGTCTCCAAUGA 1015 GCCAGUGACAUGCAAAGAAA 1016 NM_001771 CT CCL17 CTGAAGACGUGGUACCAGACA 1017 GCTTCAAGACCUCTCAAGGC 1018 NM_002987 TUT ITGAE CTGAACTGCAGAUCCTTGGUGA 1019 CACCAACGCUGCCTTTAAUG 1020 NM_002208 A A IL3RA GAAAGCAAAGGCUCAGCAGUT 1021 TGTAGTTGGUCACTTCACAU 1022 NM_002183 AAGGAAAT CCR7 GTGGCTCUCCTTGTCATTTUCC 1023 GGGAGGAACCAGGCUTTAAA 1024 NM_001838 GUT CD1C CCTTGGTGATUCTAATAGTCCT 1025 CGACGGGAUGGCATCACUA 1026 NM_001765 TGUGT MAD2L1 GCGCGUGCTTTTGTTTGUG 1027 CAGATGGATAUATGCCACGC 1028 NM_002358 UGAT PYGL CATGGCAACCCUGGGACUT 1029 AATTCUGGGCGGGACTTCUC 1030 NM_002863 CD40 CCUAGACACCUGGAACAGAGAG 1031 TCACAGGCCUCACTCGUACA 1032 NM_001250 A LY9 AATGTCTCAUGGAGAAGCAGUG 1033 CAUCAGGAACAACCCAAUCC 1034 NM_002348 AAA AAAG HLA-G GCACAGACUGACAGAAUGAACC 1035 GCCATCGUAGGCATACTGTT 1036 NM_002127 T CAUAC TLR3 GGACTTUGAGGCGGGTGTUT 1037 TCAATAGCTTGTUGAACTGC 1038 NM_003265 ATGATGUA CD48 TGCCAAGUCAGCAATTCTGUGA 1039 TCUCAGGTAAGUAACAGGCC 1040 NM_001778 AAGA STAT5A CTGAACAACUGCTGCGUGAT 1041 GACTCAAACAGGACUGTGAA 1042 NM_003152 CTUCT FCRLA GGAATTCTCCAUCACCGTGGUA 1043 TCTGAGAAUTGGCGCUGGAA 1044 NM_001184866 A BCL6 ACCTGAAAACCCACACUCGAAU 1045 GGGUGCCACAGATTUCACAG 1046 NM_001706 T ZEB1 GAGAGGAUCATGGCGGAUGG 1047 ACACTTTCTTCTUCCACAAT 1048 NM_001174093 ATGCAGUT CCL5 CTCGCTGUCATCCTCATUGCT 1049 GCACTUGCCACTGGTGUAGA 1050 NM_002985 IDO1 CTAAACATCUGCCTGATCTCAU 1051 CCCACACAUATGCCATGGUG 1052 NM_002164 AGAGT AT IL18 CCACCTGCUGCAGTCUACA 1053 TTCATUGCCACAAAGTTGAU 1054 NM_001562 GCAA TNFRSF9 CCTCACGCUCCGTTTCUCT 1055 AGTTCACATCCUCCTTCTTC 1056 NM_001561 TTCTUCT HIF1A AGTACAGGAUGCTUGCCAAAAG 1057 GGAGAAAAUCAAGTCGTGCU 1059 NM_001530 A GAAT HLA-DPB1 GCTCUGACGGCGTTACUGAT 1059 TCCTCCCGGUTGTAGATGTA 1060 NM_002121 TCUC FOXO1 TCAAGAGCGUGCCCTACTUC 1061 GATTGAGCAUCCACCAAGAA 1062 NM_002015 CTTUT CD33 CCAGGAGGAGGGAUAATGGTTC 1063 AGUGGCCGGGTTCUAGAGT 1064 NM_001772 AUA S100A9 TCAAAGAGCUGGUGCGAAAAGA 1065 GCCTCGCCAUCAGCAUGAT 1066 NM_002965 HLA-DMB CCUGTGTGGUAGAGCACACT 1067 CCAGCUGAUCACACCAAGAG 1068 NM_002118 A HLA-A CGGAGUATUGGGACCAGGAG 1069 CCACGUCGCAGCCATACATU 1070 NM_002116 A SNAI2 AUGCATATUCGGACCCACACAT 1071 CCTGTCTGCAAAUGCTCTGT 1072 NM_003068 UG TNFRSF17 GGCAGTGCUCCCAAAAUGAA 1073 TCGCATTCGUTCCTTTCACU 1074 NM_001192 GA LRP1 TCACCUCCAAGACAGTGCTUT 1075 TCAGCUCAGGACCTTCAUAC 1076 NM_002332 ACA MAGEA4 CGAGCTUCTGCGTCUGACT 1077 AGATCTTCUCCTTGGTGCUC 1078 NM_001011548 CT HLA-DQA1 CCTTCTTCAAGAUCAGTTACCU 1079 ACAGTCTCUGTGAGCTCUGA 1080 NM_002122 CACC CAT CDID CCUGCCCCCAATTTGUCAGT 1081 GCTTTGGGUAGAATCCUGAG 1082 NM_001766 ACA RPS6 GATGAACGCAAACUTCGTACTT 1083 GGTCAAGACACCCUGCTUCA 1084 NM_001010 UCT MKI67 CGTCGTGTCUCAAGATCTAGCU 1085 TGAGTCATCUGCGGTACTGU 1086 NM_002417 T CT GZMK GCCATTTAUGGCCTCCAUCCA 1087 GAGAGAGAGUGTGCGCCUAA 1088 NM_002104 A CD79A ACUAACCAACCCACUGGGAGAA 1089 TCAATGATGCUGGGACCTTG 1090 NM_001783 UG CD37 GCGGGACGUCGUAGAGAAA 1091 CCTCTCAGGAUGAGGACTUG 1092 NM_001774 GA FUT4 CAGGTUCCCCTCACAGUCAAT 1093 CATGTAGUGGCACCTGCUGA 1094 NM_002033 A AIF1 GGGAGACGUTCAGCUACCC 1095 GGGCAACUCAGAGATAGCTT 1096 NM_001623 TCUT CCR1 CACGGACAAAGUCCCTUGGA 1097 CCAAAGGCCCUCTCGTUCAC 1098 NM_001295 PRDM1 CAGGCGGAGGCAUCCTUAC 1099 AGGAACTGTGUCATTGGTGT 1100 NM_001198 AGATTUC CD47 ACTGGCCTTGGUTTAATTGTGA 1101 CUCAGUCCAACCACAGCGA 1102 NM_001777 CUT CD74 CAGGCACUCCTUGGAGCAA 1103 GCACTGGAGUGGCAGAUAGT 1104 NM_001025159 LAG3 GCGACTTUACCCTTCGACUAGA 1105 GGGAUCCAGGUGACCCAAAG 1106 NM_002286 TNFRSF4 GCCCUGCACGTGGTGUAA 1107 CAACTCCAGGCUTGTAGCTG 1108 NM_003327 UC CD2 GACCGATGAUCAGGATATCTAC 1109 ACCTCACAGGUCAGGGTUGT 1110 NM_001767 AAGGUA CCL4 GTCCTGTCTCUCCTCATGCTAG 1111 CTGCTGGTCUCATAGTAATC 1112 NM_002984 UA UACCAC BAGE CAGGCUCCAACCUCCAGC 1113 AGCUGGAGTGTUAGGAGGGC 1114 NM_182482 LEXM AGCCCCCAUTCCTGTUGAC 1115 GATATCGCUGCCGCCUGT 1116 NM_001110533 CCR6 GGGCTGAACCAUACACTCCUT 1117 AATCAACTGAGTAAUATGAA 1118 NM_004367 GTATTGACUGACA CD70 CCCCCUGCCAGTAUAGCCT 1119 GTGATCUGCCTCGTGGUGT 1120 NM_001252 CDK1 GAAGTGUGGCCAGAAGUGGA 1121 TCGTTTGGCTGGAUCATAGA 1122 NM_001786 TTAACAUT CTAG1B CATCCUGGGCCAGGCUC 1123 GGCTUCAGGGCTGAAUGGAT 1124 NM_001327 CTAG2 TGCGTGAUCCACAUCAACAGG 1125 GUCCCGGGAUGCCGCAC 1126 NM_020994 CX3CR1 ATAACAGGCCUCAGCCAAAUCA 1127 GGUAAAGTCUGAGCAGGACA 1128 NM_001171174 T GG CX3CR1 AATATAACAGGCCUCAGCCAAA 1129 AGCTGACTGUGCTGTGCTUT 1130 ENST0000 UCAT 0541347 CX3CR1 GTCCCCAAUATAACAGGCCUCA 1131 ACAGAGCACCCGCUGUC 1132 ENST0000 G 0435290 CX3CR1 CCCCAATAUAACAGGCCUCAGC 1133 AGGAUGAGAGAACCCCUGGA 1134 ENST0000 G 0399220 GAGE1, GAGE12I, TATGAGTUGGCGAGGAAGAUCG 1135 GGATCCUGACGTTGAGTUGC 1136 GAGE12F T NM_001040663 GAGE12J TATGAGTUGGCGAGGAAGAUCG 1137 GGATCCUGACGTTGAGTUGC 1138 NM_001098406 T GAGE2C, GAGE2A, TATGAGTUGGCGAGGAAGAUCG 1139 GGATCCUGACGTTGAGTUGC 1140 GAGE2E T NM_001472 GAGE10 TATGAGTUGGCGAGGAAGAUCG 1141 GGATCCUGACGTTGAGTUGC 1142 NM_001098413 T GAGE13 TATGAGTUGGCGAGGAAGAUCG 1143 GGATCCUGACGTTGAGTUGC 1144 NM_001098412 T IKZF1 CATGGATGCUGATGAGGGUCAA 1145 GACTCTGTCACUCTTGGAGC 1146 NM_006060 TUT IL17A GAATACCAATACCAAUCCCAAA 1147 CCAAGUGGCGGCACTUT 1148 NM_002190 AGGUC IL2 AATCCATCTGTUCAGAAATTCT 1149 AAAAACUTTCACTUAAGACC 1150 NM_000586 ACAAUGGT CAGGGA IL21 TTTGGAGAAGUGATTTGAATCT 1151 TCAAUTAAAAAGCUGAAGAG 1152 NM_021803 TTCUAGGAAT GAAACCAC IL22 AGGGAACAGCACUTCTUCAAGG 1153 GCTAGCUTGGCTGAUAACAA 1154 NM_020525 CACA KIR2DL2 CCUGTCUGCACAGACAGCA 1155 CAGGCGACCUGGGUGGG 1156 ENST00000 344867 KIR2DL3 GGCTCTTUCCGTGACTCUCC 1157 GAACAUGCAGGTGTCUGGG 1158 NM_015868 MAGEA10 ATGATGACTCUGATCAGGGUAG 1159 GAGAGCAAGAGGUCAAGAGC 1160 NM_021048 GA UG MIF TGCCGGACGGGUTCCUC 1161 TGUGCAGGCUGCAGAGC 1162 NM_002415 PTPRCAP CGCTGTCCUCCGCGCUG 1163 CUGCAGACGAGCACUGAGC 1164 ENST00000 326294 SSX2 CGGTTAGGGUCATTATCCAAAT 1165 AGGAAGAGUGGGAAAAGAUG 1166 NM_003147 CATUCC AAAGC TCF7 ACGGTGUCCCCCAACUCT 1167 GGGAGCUGCCCCAUGCT 1168 NM_003202 XAGEIB GTTCCGGCGUCAAGGUGA 1169 ATCTAATATAAAACCAGCUT 1170 NM_001097594 GCGTTGTTUC CEACAM8 AGGAATGACGUAGGACCCTAUG 1171 GTTGAGATTUACCCCTGCAU 1172 NM_001816 AAT GGT CXCR3 CGTTTTCUCCATAGTCAUAGGA 1173 CUGCAGGTTUCCAACCACAA 1174 NM_001504 AGAGC G FCGR1A CCAACATAAGUCACAATGGCAC 1175 CCAGAUTCCCCUCCAGGAGT 1176 NM_000566 CUA FCGR3B CATUCCAAAAGCCACACUCAAA 1177 GGTACCCAGGUGGAGAGAAU 1178 NM_000570 GAT GA FYB CTGACGAAATUCCACAAAACCT 1179 GGGACTGGUGGTTGTGAUGG 1180 NM_001465 CUT HLA-C GAGCAGAGAUACACGTGCCAUA 1181 AGCUCCAAGGACAGCUAGGA 1182 NM_002117 T HLA-DQA2 GGCTCTTAUGAATCCCATCCUG 1183 AGAGAAGAGGUGAGAGAAGA 1184 NM_020056 AAA AACATTUG IFNG TTTAAAGAUGACCAGAGCAUCC 1185 TGCTTTGCGUTGGACATUCA 1186 NM_000619 AAAAGA AG KIR2DL1 GCTGAGCUGAGCUCGGT 1187 CUGATTUCACCAGGCGACCT 1188 NM_014218 KRT5 GGTTGATGCACUGATGGATGAG 1189 CCTCAGCGAUGATGCTAUCC 1190 NM_000424 AUT AG LMNA AGCAAAGTGCGUGAGGAGTUT 1191 CGCUTCTCACUGAGAGCAGT 1192 NM_170707 PTPN11 GCGGAUGGTGTUCCAAGAAAA 1193 GCGCTTTCTUTGACGTTCCU 1194 NM_002834 AA

RNA and cDNA Preparation

Total RNA was extracted from samples with the RecoverAll™ Total Nucleic Acid Isolation Kit (Ambion, Inc.), according to the manufacturer instructions, and quantified, e.g., using the Qubit™ RNA HS Assay Kit (Thermo Fisher) for quantifying RNA. A total of 10 ng of total RNA was first reverse transcribed to cDNA with SuperScript® VILO™ cDNA Synthesis Kit (Thermo Fisher, Cat. No. 11754050) according to manufacturer instructions. Prepared template was used to amplify targets with the immune response assay comprising primer pairs in Table 2.

PCR Amplify RNA Targets

A multiplex polymerase chain reaction was performed to amplify 398 individual amplicons across a RNA expression sample. A list of genes and the resulting amplicon sequences generated are provided in Table 1. Primer pairs are presented in Table 2. To a single well of a 96-well PCR plate was added 4 microliters of immune response primer pool containing 398 primer pairs at a concentration of 15 μM in TE, 10 microliters prepared cDNA and 4 microliters of an amplification reaction mixture (5× AmpliSeq HiFi Master Mix) that can include glycerol, dNTPs, and Platinum® Taq High Fidelity DNA Polymerase (Invitrogen, Catalog No. 11304) to a final volume of 20 microliters with DNase/RNase Free Water (Life Technologies, CA, Part No. 600004).

The PCR plate was sealed and loaded into a thermal cycler (GeneAmp® PCR system Dual 96-well thermal cycler (Life Technologies, CA, Part No. N8050200 and 4314445)) and run on the following temperate profile to generate the preamplified amplicon library.

An initial holding stage was performed at 99° C. for 2 minutes, followed by 19 cycles of denaturing at 99° C. for 15 seconds and an annealing and extending stage at 60° C. for 4 minutes. After cycling, the preamplified amplicon library was held at 10° C. until proceeding.

Partially Digest Amplicons

The sample was briefly centrifuged to collect contents before proceeding. To the preamplified amplicon library (˜20 microliters), 2 microliters of FuPa reagent was added. The reaction mixture was sealed, mixed thoroughly to ensure uniformity and incubated at 50° C. for 10 minutes, 55° C. for 10 minutes, 60° C. for 20 minutes, then held at 10° C. for up to 1 hour. The sample was briefly centrifuged to collect contents before proceeding.

Ligate Adapters to Amplicons and Purify

After incubation, the reaction mixture proceeded directly to a ligation step. Here, the reaction mixture now containing the phosphorylated amplicon library was combined with 2 microliters of A/P1 Adapters (sold as a component of the Ion Fragment Library Kit, Thermo Fisher), microliters of Switch Solution (sold as a component of the Fragment Library Kit, Thermo Fisher) and 2 microliters of DNA ligase, added last (sold as a component of the Ion Fragment Library Kit, Thermo Fisher), then incubated at the following: 22° C. for 30 minutes, 72° C. for 10 minutes, then held at 10° C. for up to 1 hour. The sample was briefly centrifuged to collect contents before proceeding.

After the incubation step, 45 microliters (1.5× sample volume) of room temperature AgenCourt® AMPure® Reagent (Beckman Coulter, Calif.) was added to ligated DNA. The mixture was pipetted thoroughly to mix the bead suspension with the ligated DNA. The mixture was pulse-spin and incubated at room temperature for 5 minutes. Samples underwent another pulse-spin and were placed on a magnetic rack such as a DynaMag™-2 spin magnet (Life Technologies, CA, Part No. 123-21D) for two minutes. After the solution had cleared, the supernatant was discarded. Without removing the tube from the magnetic rack, 150 microliters of freshly prepared 70% ethanol was introduced into the sample, and incubated while gently rotating the tube on the magnetic rack. After the solution cleared, the supernatant was discarded without disturbing the pellet. A second ethanol wash was performed and the supernatant discarded. Any remaining ethanol was removed by pulse-spinning the tube and carefully removing residual ethanol while not disturbing the pellet. The pellet was air-dried for about 5 minutes at room temperature.

The pellet was resuspended in 20 microliters of DNase/RNase Free Water (Life Technologies, CA, Part No. 600004) and vortexed to ensure the sample was mixed thoroughly. The sample was pulse-spin and placed on the magnetic rack for two minutes. After the solution cleared, the supernatant containing the ligated DNA was transferred to a new Eppendorf LoBind™ tube for long-term storage. After quantification, determine the dilution factor that results in a concentration of ˜100 μM, which is suitable for template preparation using an Ion template kit.

Quantify the Library

Ligated library was quantified, using a Qubit™ 2.0 or 3.0 Fluorometer and/or by qPCR using the Ion Library TaqMan® Quantitation Kit (Cat. No. 4468802), according to manufacturer instructions.

Amplify and Purify the Library

The ligated preamplified library (˜20 microliters) was combined with 50 microliters of Platinum® PCR SuperMix High Fidelity (Thermo Fisher, sold as a component of the Ion Fragment Library Kit) and 2 microliters of Library Amplification Primer Mix (sold as a component of the Ion Fragment Library Kit). The solution was applied to a single well of a 96-well PCR plate and sealed. The plate was loaded into a thermal cycler (GeneAmp® PCR system 9700 Dual 96-well thermal cycler (Life Technologies, CA, Part No. N8050200 and 4314445)) and run on the following temperate profile to generate the final amplicon library: hold at 98° C. for 2 minutes, followed by 5 cycles of denaturing at 98° C. for 15 seconds and an annealing and extending stage at 64° C. for 1 minute. After cycling, the final amplicon library was held at 4° C. until proceeding to the final purification step outlined below.

A two-round purification of the final library was carried out. 25 μL (0.5× sample volume) of Agencourt™ AMPure™ XP Reagent was added to each plate well containing ˜50 μL of sample. The bead suspension was pipetted up and down to thoroughly mix the bead suspension with the final amplicon library. The sample was then pulse-spun and incubated for 5 minutes at room temperature. The plate containing the final amplicon library was placed on a magnetic rack such as a DynaMag™-side magnet (Thermo Fisher) for 5 minutes to capture the beads. Once the solution cleared, the supernatant was carefully transferred without disturbing the bead pellet. A second round of purification was carried out, adding 60 microliters (1.2× sample volume) of Agencourt™ AMPure™ XP Reagent was added to each plate well containing sample. The bead suspension was pipetted up and down to thoroughly mix the bead suspension and incubated for 5 minutes at room temperature. The plate containing the final amplicon library was placed on a magnetic rack for 3 minutes to capture the beads. Without removing the plate from the magnetic rack, 150 microliters of freshly prepared 70% ethanol was introduced into the beads containing sample. The sample was incubated for 30 seconds while gently rotating the tube on the magnetic rack. After the solution cleared, the supernatant was discarded without disturbing the pellet. A second ethanol wash was performed and the supernatant discarded. Any remaining ethanol was removed by pulse-spinning the tube and carefully removing residual ethanol while not disturbing the pellet. The pellet was air-dried for about 5 minutes at room temperature.

Once the tube was dry, the tube was removed from the magnetic rack and 50 microliters of Low TE was added (Thermo Fisher), pipetted and vortexed to ensure the sample was mixed thoroughly. The sample was pulse-spin and placed on the magnetic rack for two minutes. After the solution cleared, the supernatant containing the final amplicon library was analyzed using Qubit™ Fluorometer and Qubit™ dsDNA HS Assay Kit according to manufacturer instructions to quantify the library and calculate the dilution factor for template preparation and sequencing. Library was diluted to ˜50 pM for use in template preparation or stored in 1.5-mL Eppendorf LoBind™ tube for long-term storage.

Template Preparation and Sequencing

An aliquot of the final library was used in template preparation with either Ion OneTouch™ 2 System or Ion Chef™ instrument according to the manufacturer's instructions.

Sequencing was performed on either the Ion S5™ System or the Ion PGM™ System. According to manufacturer instructions, and gene expression was quantified with the Ion Torrent Suite™ software subjected to analysis and sequencing according to manufacturer instructions.

Automated Processes

Alternatively, automated library prep and templating can be performed using cDNA prepared as described above in conjunction with the Ion AmpliSeg™ Kit for Chef DL8 and Ion Chef™ Instrument according to manufacturer instructions and described in the Ion AmpliSeq Library Preparation on the Ion Chef System (Thermo Fisher Scientific publication number MAN0013432), hereby incorporated by reference.

Multiple Library Preparation

When sequencing multiple libraries, a different barcode will be ligated to each library. DNA and RNA libraries from the same sample also require different barcodes. IonCode Adapters are provided at the appropriate concentration and include forward and reverse adapters in a single well. For each barcode selected, prepare a mix of Ion P1 adapter and IonXpress Barcode X (Thermo Fisher) at a final dilution of 1:4 for each adapter. Substitute 2 uL of this barcode adapter mix for the Ion AmpliSeq Adapters in ligation reactions above; and scale volumes as necessary. Multiple barcoded libraries can be sequenced on a single chip by combining equal volumes of each library before template preparation. The manufacturer recommends up to eight libraries for the following chips: Ion 318 Chip v2 (up to 4), Ion 520 Chip (up to 4), Ion 530 Chip (up to 8). A combined library is prepared by diluting all individual libraries to 50 pM concentration; combining 10 uL of each library in a single tube (preferably Eppendorf LoBind tube). After combining the last library, mix thoroughly by pipetting up and down 5 times, then centrifuge briefly to collect in the bottom of the tube. Proceed to templating and sequencing as desired.

Data Analysis

Gene expression level quantification is performed using the Torrent Suite™ RNA plugin that produces gene transcript quantification from sequence read data. The plugin utilizes target region .bed file and an associated reference library .fasta file, and optionally accepts a secont .bed file that specifies a subset of target genes allowing sample clustering. Resulting report includes gene expression counts (number of aligned reads to a given target sequence), data analysis summary, and QC plots. Normalized gene level count data can be determined using the Affymetrix Transcriptome Analysis Console (TAC) 3.1 software (Thermo Fisher Scientific). CHP files generated from Torrent Suite™ plugin can be used directly with Transcriptome Analysis Console (TAC3 software for differential gene expression analysis. Affymetrix® Transcriptome Analysis Console (TAC) Software is freely available, http://www.affymetrix.com/

Normalization of Expression

To observe how expression profiles among the selected target immune response genes vary within a large cohort of samples, we downloaded and processed more than 9,600 patient samples across 18 cancer studies from the ICGC database [Zhang et al., 2011]. To ensure high-quality RNA profiles, we excluded all samples with a library size less than 20 million reads mapped to the transcriptome and ended up with 9,148 full transcriptome datasets. For this study, we used the log 2 CPM full transcriptome (FT) expression of each gene as the ‘true’ expression value and the total number of reads mapping to genes as the ‘true’ normalizing constant for each sample in the cohort.

Results

Total RNA extracted from, samples e.g., NSCLC FFPE samples, were reverse transcribed. Libraries were generated, templated on Ion Chef™ or Ion OneTouch™ 2 System and sequenced on the Ion 55™ or Ion PGM™ System. With an input requirement as low as 10 ng of total RNA, we were able to measure robust expression, including for low expressing transcripts (e.g., IFN, IL2, IL21, IL10, IL23) from samples, e.g., NSCLC FFPE samples, CRC samples. Results show high sensitivity and specificity, with broad dynamic range and great reproducibility. Technical replicates were highly reproducible (see FIG. 1), consistent among various sample types (See FIG. 4), and the assay results were highly specific, with negligible background when tested with gDNA and water control samples in parallel. Dynamic range of the assay was determined, for example, the expression profiles of HL-60 and human lung total RNA were measured individually and mixed at 4 different ratios to evaluate dynamic range (see FIG. 2). Fold changes between pure and mixed samples were correlated, and rank order of fold change correlations between pure and mixed samples were compared. Expected fold change was calculated from pure samples and observed fold change was derived from fitting a line to the mixed samples (see TABLE 3). Ability to realiably detect low expressing markers of the panel was confirmed in replicate experiments carried out in automated and manual workflows, demonstrating consistent results even for very low expressing genes (e.g., IL2, IL21, IL2B, IL13, IL23A, IL10) and higher expressing interleukins (e.g., IL1A, IL12A, IL15, IL6, IL7, IL2RB). Data not shown. Sequencing data from the provided immune response assay also showed high concordance with results from parallel samples using quantitative real-time PCR evaluation (see FIG. 3). A larger concordance study examining results over fifty genes using reference RNA generated similar concordance. Data not shown.

TABLE 3 Sensitivity and specificity for 2-fold and 4-fold differential expression Observed Expected q_value fold change fold change Sensitivity Specificity 0.05 2 2 77.5% 98.5% 0.05 4 4 89.4% 99.1%

Analysis report files include, for example, a sample summary report (table listing each sample, total reads, aligned reads, percent aligned); absolute read count data (absolute reads for genes of each sample), RPM data (normalized by total read count for each sample), mean housekeeping scaled log 2 RPM data (for each sample), CHP files normalized by RPM, CHP files normalized by mean housekeeping genes, and expression data for genomic DNA and water samples. See FIG. 5 and FIG. 6. The analysis output generated data demonstrating distribution of genes showing frequency of genes having similar and differential read counts. See FIG. 5A. Curves are plotted on the same axis scale; and count data is fitted to a Gaussian kernel using the default R ‘density’ function. Expression of housekeeping genes across samples demonstrated similar levels of expression independent of sample type. See FIG. 5B. A heatmap correlation for r-values comparing log 2 RPM reads pair correlation samples, demonstrates correlation across entire expression assay among samples. Sample reads demonstrate correlations across each of the targets for samples. See FIG. 5C. and FIG. 6 (upper panels show Pearson correlation r-values for the regression line; diagonal panels show frequency density plot for the individual log (RPM+1) values for each sample; and lower panels show log 2 RPM+1 values plotted for each barcode pair of the entire panel, with linear least squares regression line overlaid and line slope is reported). Expression levels as measured by read count can also be depicted as a heatmap, depicting the most variation in representation across barcodes as measured by coefficient variation of normalized read counts for genes that have at least one sample with at least 100 RPM reads. For the plot, samples are eliminated if they have less than 105 reads total. An exemplary heatmap analysis demonstrating high and low expression genes across the entire panel is shown in FIG. 7.

Thus, we verified a linear and unbiased estimate of fold change in our assays across mixing concentrations of a cell-line titration experiment. FIG. 2. By achieving a high correlation (r>0.99) of technical replicates, along with robust expression estimation even at low input amounts (10 ng RNA), our assay offers a valuable solution for biomarker research in cancer immunotherapy.

Using endogenous controls accurately estimates fold change. When most genes do not change between the samples, the fold change estimate has negligible error. When most genes do change, the fold change estimate is wildly inaccurate. See FIG. 8. Thus, when using endogenous controls, we accurately measure fold change. In silico analysis of gene expression across 9148 samples across cancer types was assessed. See Table 4. Standard CPM normalization was applied to measure the expression of each gene and calculate the resulting fold change. Full transcriptome profiling provides a true estimate of expression presumably because, over the total, only a small number of the 20,000+ genes are expected to be differentially expressed. We assessed both full transcriptome expression data as well as the immune response assay expression data. Although the relative number of genes within the immune response assay subcategory vary, no single category uses a majority of the genes on the entire assay, and multiple gene categories have to change to dominate the proportion of genes of the assay. Although the immune response assay is not as large as the full transcriptome, the relative diversity of uncorrelated gene categories requires differential expression among samples to bias expression estimates. Thus, housekeeping gene normalization provides an effective optimization approach for effective estimates of targeted expression fold change estimates in cases where uncorrelated gene categories are differentially expressed among samples.

To first understand how assaying only genes from a common function can affect estimate of expression, an assay was made comprising only the 5 genes associated with the Leukocyte Migration category (ITGAL, ITGAM, ITGB7, SELL, VCAM1). Assaying only those genes does not provide an accurate expression estimate. Expanding the assay by 2 Leukocyte Inhibition and 19 TCR co-expression genes (CCR7, CD247, CD3D, CD3E, CD3G, CD6, CD8A, CD8B, CRTAM, GPR18, GRAP2, IKZF3, IL2RB, IL7R, ITK, LAMP3, LCK, PTPRCAP, TIGIT) does not increase the correlation. However, assaying 27 genes from the unrelated Tumor Marker category (AKT1, BRCA1, BRCA2, CDKN2A, EFNA4, EGFR, EGR3, IRS1, KRT5, KRT7, MAPK1, MMP2, MMP9, MYC, NOTCH3, PGF, PTGS2, PTK7, RB1, RPS6, SNAIL SNAI2, TCF7, TP63, TRIM29, TWIST1, ZEB1) or 11 housekeeping (HK) genes (ABCF1, G6PD, GUSB, HMBS, LMNA, LRP1, POLR2A, SDHA, TBP, TFRC, TUBB) improves the correlation with true expression values. See FIG. 9A. HK genes are uncorrelated with each other, however, unlike tumor markers (that are also uncorrelated with each other), HK genes provide low-variance and are able to be measured with high precision. For example, when we compared performance and robustness of various metrics for accurate fold change estimates, we found the geometric mean of housekeeping genes provides a more accurate fold change estimate than a standard CPM calculation (with or without housekeeping genes. See FIG. 9B. Housekeeping genes alone can solely correct expression estimates in small panel assays. Using uncorrelated gene categories also has similar effect to normalization. Furthermore, using the inverse of the sum of only the HK genes as the normalizing factor provides an even stronger correlation than using the sum of all genes on the panel.

TABLE 4 Distribution of samples across cancer types Study ID Cancer Type Sample Size BLCA-US Bladder Urothelial Cancer 318 BRCA-US Breast Cancer 1195 CESC-US Cervical Squamous Cell Carcinoma 264 COAD-US Colon Adenocarcinoma 516 HNSC-US Head and Neck Squamous Cell Carcinoma 524 KIRC-US Kidney Renal Clear Cell Carcinoma 598 KIRP-US Kidney Renal Papillary Cell Carcinoma 254 LIHC-US Liver Hepatocellular carcinoma 345 LUAD-US Lung Adenocarcinoma 543 LUSC-US Lung Squamous Cell Carcinoma 473 OV-US Ovarian Serous Cystadenocarcinoma 530 PAAD-US Pancreatic Cancer 145 PRAD-US Prostate Adenocarcinoma 870 READ-US Rectum Adenocarcinoma 163 SKCM-US Skin Cutaneous melanoma 868 STAD-US Gastric Adenocarcinoma 900 THCA-US Head and Neck Thyroid Carcinoma 566 UCEC-US Uterine Corpus Endometrial Carcinoma 541

Thus, we also found that normalization using endogenous controls (housekeeping genes) provides accurate fold change estimates for small panel assays with highly-correlated genes. Uncorrelated gene categories can have the same effect as house-keeping genes on normalization, however, close cases where uncorrelated gene categories are differentially expressed between samples will bias expression estimates when not using housekeeping genes. The method of housekeeping gene normalization provided herein can improve correlation with true values of expression assays. In particular, housekeeping gene normalization may improve assessment of expression in sub-panels of the provided immune response assay, and may be useful in connection with the relatively large immune response assay provided herein.

From the previous analysis, it is clear that normalization using housekeeping genes in conjunction with compositions and methods of the invention are necessary for accurately estimating fold change expression. It is also clear that using only the housekeeping genes to calculated a normalizing constant is a preferred way to estimate gene expression for accurate fold change estimation. An important factor to using housekeeping genes for calculating normalizing constant is to determine which is metric of the gene counts provides the most accurate fold change estimates. We thus compared 6 metrics across 18 cancer studies; for each study we calculate the log 10 of the total counts and determine the correlation to the following metrics: log 10 of the sum of housekeeping counts (analogous to RPM normalization), log 10 of the geometric mean of the housekeeping counts, the median of the log 10 of housekeeping counts, the 75th quantile of the log 10 of housekeeping counts, and the trimmed mean (15% and 25% of the log 10 of HK counts). Interestingly, the sum of housekeeping counts does not provide an optimal correlation value, instead the geometric mean provides the highest average correlation and number of cancers where the R-squared is greater than 0.5 (data not shown). Of the more robust measures, the 15% trimmed mean provides the highest average R-squared. To further estimate which metric is most robust, we simulate a dropout of a random housekeeping genes for every sample by setting a random housekeeping gene of each sample to have 0 reads. We then reproduce the same analysis to compare the various metrics using dropout samples. Interestingly, the geometric mean still provides the highest correlation with the full transcriptome normalizing constant even in the dropout samples, while the 15% trimmed mean also shows high performances (data not shown). This suggests that the geometric mean is an optimal metric due to its high correlation with the true normalizing constant values and its robustness to dropouts. Additionally, we carried out analysis of normalizing constant, correlations and drop out correlation among various cancer types of interest and confirmed even across various sample types the housekeeping set of genes provided in the instant compositions and methods performed consistently across populations of interest.

Housekeeping (HK) constant:

    • Let x be a vector of counts for every gene on the immune response assay and let HKgenes be the set of housekeeping genes; add a pseudocount 1 to every entry in the vector x, creating a new vector y.
    • Find the geometric mean of the counts of housekeeping genes, calculated as 10{circumflex over ( )}mean (log 10(y_HKgenes)), where y_HKgenes is a subset of the y vector to only include the counts associated with the housekeeping genes. This geometric mean value will be denoted asHKnorm.
    • Multiply each entry of y by the (10{circumflex over ( )}6/HKnorm), creating a new vector z.
    • Finally take the log 2 value of each entry in z and add, creating the final housekeeping gene normalized expression estimate.

Once established, an HKnorm constant was calculated by taking the geometric mean of our 11 housekeeping genes and comparing our normalizing strategy to a standard RNA-seq RPM measurement using the 398 genes of the provided immune response assay. Since this assay is made up of a relatively large number of genes that are on average uncorrelated to each other, one could make a valid argument that housekeeping genes are unnecessary to the assay and do not provide additional information. To address this concern, we compared the HKnorm constant against the standard RPM constant and the RPM constant when housekeeping genes are removed from the assay. We first evaluated how these normalizing strategies compare to the full transcriptome constant across all samples, where we found that removing housekeeping genes led to decrease in the correlation of the RPM constant compared to a standard RPM calculation, however, even the standard RPM calculation under-performs when compared to HKnorm. (data not shown). We then examined which normalizing strategy would be optimal for each cancer study that we evaluated, and found that HKnorm largely outperforms the RPM strategies across all studies except for LIHC-US, where no normalization strategy does particularly well and all the strategies produce equivalent results. When looking at the performance each strategy across target genes on the immune response assay of the invention, we see that HKnorm outperforms the RPM strategies in all genes, as expected. Interestingly, genes with smaller variance have a smaller correlation to their FT values than those with higher variance. See FIG. 9B. Furthermore, genes with low expression and those with high average expression also have smaller correlation values than those closer to the mean of average expression values (data not shown).

REFERENCES

  • Schalper K A, et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst. 2015 Feb. 3; 107(3).
  • Padmanee S, et al. Immune Checkpoint Targeting in Cancer Therapy: Toward Combination Strategies with Curative Potential. Cell 2015 April 161(2):205-214.
  • Love M, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biology, 2014.
  • Law C, Chen Y, Shi W, Smyth G. voom: precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biology, 2014.
  • Hansen K, Irizarry R. Removing technical variability in RNA-seq data using conditional quantile normalization. Biostatistics, 2011.
  • Risso D, Schwartz K, Sherlock G and Dudoit S. GC-Content Normalization for RNA-Seq Data. BMC Bioinformatics, 2011.
  • Zhang, J., Baran, J., Cros, A., Guberman, J. M., Haider, S., Hsu, J. Kasprzyk, A. (2011). International Cancer Genome Consortium Data Portal—a one-stop shop for cancer genomics data. Database: The Journal of Biological Databases and Curation, 2011.

Claims

1-20. (canceled)

21. A method for determining immune response activity in a biological sample, comprising:

multiplex amplification of a plurality of target sequences from the biological sample, wherein the amplification comprises contacting at least a portion of the sample with a plurality of sets of primer pair reagents and a polymerase under amplification conditions, wherein the plurality of target sequences are selected from the group consisting of the following functions: checkpoint pathways, T cell related signaling pathways, markers of tumor infiltrating lymphocytes (TILs), and tumor markers, and housekeeping genes, and wherein the plurality of primer pair reagents are selected from the group consisting of SEQ ID NOs: 399-1194, thereby producing amplified immune response target sequences; and
detecting a level of expression of each of the plurality of immune response target sequences, wherein a change in the level of expression of one or of the immune response target sequences as compared with a control sample determines a change in immune response activity in the sample.

22. The method of claim 21, wherein the immune response target sequences are from target genes selected from the group consisting of immune checkpoint pathways and targets; T and B cell signaling genes, markers of lymphocyte subsets, interferon signaling genes, cytokine signaling genes; tumor markers, tumor antigens, proliferation markers; and housekeeping genes.

23. The method of claim 22, wherein the target genes are selected from the genes of CD63, CD69, CXCL1, KLRD1, HLA-DOB, CXCR5, IL12B, PTK7, CEACAM1, CXCL9, IL13, NT5E, VEGFA, ABCF1, D38, JAML, S100A8, MYC, IRF1, CCL22, CXCR2, IFIT1, IFIT2, CD68, M6PR, SH2D1A, ISG20, GBP1, TBP, STAT6, ID3, CX3CL1, KLRB1, TNFSF4, CD52, IL10RA, HLA-DOA, IFNB1, CCR5, IKZF3, STAT1, CD6, BRCA1, CORO1A, TBX21, KLRK1, CXCR6, PTEN, PMEL, DMBT1, IFI44L, LAPTM5, CD226, TNFSF13B, ICOS, CD160, TRIM29, LST1, ZBTB46, VTCN1, KREMEN1, PDCD1LG2, TUBB, CLEC4C, CD86, HAVCR2, GZMH, NFATC1, CD8B, BCL2, GADD45GIP1, CBLB, ITGA1, CD8A, IL2RA, EIF2AK2, MADCAM1, PTPN6, LRG1, ADGRE5, SH2D1B, ITGB2, HLA-DPA1, DGAT2, IGF1R TAGAP, LMNA, NCAM1, TIGIT, IL17F, HLA-F-AS1, CD247, CD79B, IDO2, IL4, TYROBP, BTLA, AKT1, IL2RG, POLR2A, ITGAX, IL1B, CSF2RB, DDX58, KIAA0101, CD274, LAMPS, TNFAIP8, FOXP3, IL12A, SAMHD1, SIT1, CD3E, ICOSLG, HGF, MELK, IGSF6, GNLY, TDO2, KRT7, HLA-E, HLADMA, LAMP1, NTN3, CD28, TARP, EGFR, CCR4, MAGEA3, BATF, KLRG1, IRS1, CSF1R, CTLA4, TNFSF18, POU2AF1, GZMA, PIK3CA, ITK, IFI27, EOMES, LCN2, CD80, CD83, CXCL13, MTOR, FCER1G, TFRC, RORC, MMP9, BST2, PIK3CD, FCGR2B, TNFRSF14, OAS3, GRAP2, CCNB2, MLANA, MAGEA12, VCAM1, CDKN3, NCR1, FAS, GZMB, IRF9, IFITM2, TNFSF14, HLA-B, SDHA, NRP1, EBB, EFNA4, PVR BUB1, SKAP2, PRF1, CCL20, TNFRSF18, CTSS, NKG7, ISG15, PDCD1, SNAI1, CXCL11, CIITA, IFI35, TNFSF9, TNFSF10, MMP2, EGR3, MAGEA1, CD163, IL6, KLRF1, B3GAT1, C1QA, OAS1, IKZF2, TLR9, KLF2, GUSB, NFKBIA, IL23A, HERC6, SLAMF8, IL15, TLR7, OAS2, HLA-DRA, CRTAM, MAGEC2, ICAM1, CD4, MAPK14, C1QB, NOTCH3, NCR3, STAT3, TLR8, CYBB, IKZF4, IFIH1, LCK, BCL2L11, ITGAM, ITGB7, JCHAIN, CD209, SLAMF7, IL10, IL1A, FCGR3A, IFNA17, EGR2, TOP2A, C10orf54, FOXM1, AXL, MS4A1, IFI6, CD3D, GPR18, CD3G, ZAP70, HMBS, IL7, IFIT3, RBI, PTGS2, TGFB1, NCF1, TWIST1, CA4, SELL, LILRB1, CD14, ALOX15B, PECAM1, NOS2, FASLG, CD44, ENTPD1, CMKLR1, CD53, TNF, CXCL8, CD40LG, HLA-F, GATA3, LYZ, ARG1, IL2RB, NECTIN2, MPO, CCR2, BRCA2, ADORA2A, G6PD, TAP1, MXI, HLA-DQB2, CD27, CD276, STAT4, PTPN7, PTPRC, PSMB9, CD244, CXCR4, MAPK1, TP63, IRF4, CCL3, CCL18, IL7R, HLA-DRB1, CEACAM8, CXCL10, CCL2, SRGN, CD19, ITGB1, IFITM1, CCL21, MRC1, PGF, ITGAL, ID2, CD22, CCL17, ITGAE, IL3RA, CCR7, CD1C, MAD2L1, PYGL, CD40, LY9, HLA-G, TLR3, CD48, STAT5A, FCRLA, BCL6, ZEB1, CCL5, IDOL IL18, TNFRSF9, HIF1A, HLA-DPB1, FOXO1, CD33, S100A9, HLA-DMB, HLAA, SNAI2, TNFRSF17, LRP1, MAGEA4, HLA-DQA1, CD1D, RPS6, MKI67, GZMK, CD79A, CD37, FUT4, AIF1, CCR1, PRDM1, CD47, CD74, LAGS, TNFRSF4, CD2, CCL4, BAGE, LEXM, CCR6, CD70, CDK1, CTAG1B, CTAG2, CX3CR1, GAGE1, GAGE121, GAGE12F, GAGE12J, GAGE2C, GAGE2A, GAGE2E, GAGE10, GAGE13, IKZF1, IL17A, IL2, IL21, IL22, KIR2DL2, KIR2DL3, MAGEA10, MIF, PTPRCAP, SSX2, TCF7, XAGE1B, CXCR3, FCGR1A, FCGR3B, FYB, HLA-C, HLA-DQA2, IFNG, KIR2DL1, KRT5, and PTPN11.

24. The method of claim 23, wherein the target genes comprise the genes of CD63, CD69, CXCL1, KLRD1, HLA-DOB, CXCR5, IL12B, PTK7, CEACAM1, CXCL9, IL13, NT5E, VEGFA, ABCF1, D38, JAML, S100A8, MYC, IRF1, CCL22, CXCR2, IFIT1, IFIT2, CD68, M6PR, SH2D1A, ISG20, GBP1, TBP, STAT6, ID3, CX3CL1, KLRB1, TNFSF4, CD52, IL10RA, HLA-DOA, IFNB1, CCR5, IKZF3, STAT1, CD6, BRCA1, CORO1A, TBX21, KLRK1, CXCR6, PTEN, PMEL, DMBT1, IFI44L, LAPTM5, CD226, TNFSF13B, ICOS, CD160, TRIM29, LST1, ZBTB46, VTCN1, KREMEN1, PDCD1LG2, TUBB, CLEC4C, CD86, HAVCR2, GZMH, NFATC1, CD8B, BCL2, GADD45GIP1, CBLB, ITGA1, CD8A, IL2RA, EIF2AK2, MADCAM1, PTPN6, LRG1, ADGRE5, SH2D1B, ITGB2, HLA-DPA1, DGAT2, IGF1R TAGAP, LMNA, NCAM1, TIGIT, IL17F, HLA-F-AS1, CD247, CD79B, IDO2, IL4, TYROBP, BTLA, AKT1, IL2RG, POLR2A, ITGAX, IL1B, CSF2RB, DDX58, KIAA0101, CD274, LAMPS, TNFAIP8, FOXP3, IL12A, SAMHD1, SIT1, CD3E, ICOSLG, HGF, MELK, IGSF6, GNLY, TDO2, KRT7, HLA-E, HLADMA, LAMP1, NTN3, CD28, TARP, EGFR, CCR4, MAGEA3, BATF, KLRG1, IRS1, CSF1R, CTLA4, TNFSF18, POU2AF1, GZMA, PIK3CA, ITK, IFI27, EOMES, LCN2, CD80, CD83, CXCL13, MTOR, FCER1G, TFRC, RORC, MMP9, BST2, PIK3CD, FCGR2B, TNFRSF14, OAS3, GRAP2, CCNB2, MLANA, MAGEA12, VCAM1, CDKN3, NCR1, FAS, GZMB, IRF9, IFITM2, TNFSF14, HLA-B, SDHA, NRP1, EBB, EFNA4, PVR BUB1, SKAP2, PRF1, CCL20, TNFRSF18, CTSS, NKG7, ISG15, PDCD1, SNAI1, CXCL11, CIITA, IFI35, TNFSF9, TNFSF10, MMP2, EGR3, MAGEA1, CD163, IL6, KLRF1, B3GAT1, C1QA, OAS1, IKZF2, TLR9, KLF2, GUSB, NFKBIA, IL23A, HERC6, SLAMF8, IL15, TLR7, OAS2, HLA-DRA, CRTAM, MAGEC2, ICAM1, CD4, MAPK14, C1QB, NOTCH3, NCR3, STAT3, TLR8, CYBB, IKZF4, IFIH1, LCK, BCL2L11, ITGAM, ITGB7, JCHAIN, CD209, SLAMF7, IL10, IL1A, FCGR3A, IFNA17, EGR2, TOP2A, C10orf54, FOXM1, AXL, MS4A1, IFI6, CD3D, GPR18, CD3G, ZAP70, HMBS, IL7, IFIT3, RBI, PTGS2, TGFB1, NCF1, TWIST1, CA4, SELL, LILRB1, CD14, ALOX15B, PECAM1, NOS2, FASLG, CD44, ENTPD1, CMKLR1, CD53, TNF, CXCL8, CD40LG, HLA-F, GATA3, LYZ, ARG1, IL2RB, NECTIN2, MPO, CCR2, BRCA2, ADORA2A, G6PD, TAP1, MXI, HLA-DQB2, CD27, CD276, STAT4, PTPN7, PTPRC, PSMB9, CD244, CXCR4, MAPK1, TP63, IRF4, CCL3, CCL18, IL7R, HLA-DRB1, CEACAM8, CXCL10, CCL2, SRGN, CD19, ITGB1, IFITM1, CCL21, MRC1, PGF, ITGAL, ID2, CD22, CCL17, ITGAE, IL3RA, CCR7, CD1C, MAD2L1, PYGL, CD40, LY9, HLA-G, TLR3, CD48, STAT5A, FCRLA, BCL6, ZEB1, CCL5, IDOL IL18, TNFRSF9, HIF1A, HLA-DPB1, FOXO1, CD33, S100A9, HLA-DMB, HLAA, SNAI2, TNFRSF17, LRP1, MAGEA4, HLA-DQA1, CD1D, RPS6, MKI67, GZMK, CD79A, CD37, FUT4, AIF1, CCR1, PRDM1, CD47, CD74, LAGS, TNFRSF4, CD2, CCL4, BAGE, LEXM, CCR6, CD70, CDK1, CTAG1B, CTAG2, CX3CR1, GAGE1, GAGE121, GAGE12F, GAGE12J, GAGE2C, GAGE2A, GAGE2E, GAGE10, GAGE13, IKZF1, IL17A, IL2, IL21, IL22, KIR2DL2, KIR2DL3, MAGEA10, MIF, PTPRCAP, SSX2, TCF7, XAGE1B, CXCR3, FCGR1A, FCGR3B, FYB, HLA-C, HLA-DQA2, IFNG, KIR2DL1, KRT5, and PTPN11.

25. The method of claim 21, wherein the plurality of immune response target sequences comprise a plurality of amplicon sequences selected from SEQ ID NOs: 1-398.

26. The method of claim 25, wherein the plurality of target sequences comprises each of the amplicon sequences of SEQ ID NOs: 1-398.

27. The method of claim 26, wherein the plurality of target sequences consists of each of the amplicon sequences of SEQ ID NOs: 1-398.

28. The method of claim 21, wherein the plurality of primer pair reagents comprises each of the primer pairs of SEQ ID NOs: 399-1194.

29. The method of claim 28, wherein the plurality of primer pair reagents consists of each of the primer pairs of SEQ ID NOs: 399-1194.

30. The method of claim 21, wherein the one or more housekeeping genes are selected from ABCF1, G6PD, GUSB, HMBS, LMNA, LRP1, POLR2A, SDHA, TBP, TFRC, and TUBB.

31. The method of claim 21, wherein the biological sample comprises a tumor, tissue surrounding a tumor, or both.

32. The method of claim 21, wherein the biological sample and the control sample are from the same individual.

Patent History
Publication number: 20230088159
Type: Application
Filed: Jul 29, 2022
Publication Date: Mar 23, 2023
Applicant: Life Technologies Corporation (Carlsbad, CA)
Inventors: Ann Mongan (San Francisco, CA), Alex Atkins (San Francisco, CA)
Application Number: 17/816,214
Classifications
International Classification: C12Q 1/6886 (20060101);