US20240021314A1 - Cancer Score for Assessment and Response Prediction from Biological Fluids - Google Patents

Cancer Score for Assessment and Response Prediction from Biological Fluids Download PDF

Info

Publication number
US20240021314A1
US20240021314A1 US18/474,797 US202318474797A US2024021314A1 US 20240021314 A1 US20240021314 A1 US 20240021314A1 US 202318474797 A US202318474797 A US 202318474797A US 2024021314 A1 US2024021314 A1 US 2024021314A1
Authority
US
United States
Prior art keywords
dna
cancer
repair
gene
rna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/474,797
Inventor
Shahrooz Rabizadeh
Patrick Soon-Shiong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nantomics LLC
Original Assignee
Nantomics LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nantomics LLC filed Critical Nantomics LLC
Priority to US18/474,797 priority Critical patent/US20240021314A1/en
Publication of US20240021314A1 publication Critical patent/US20240021314A1/en
Pending legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/20ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for computer-aided diagnosis, e.g. based on medical expert systems
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/10Ploidy or copy number detection
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H10/00ICT specially adapted for the handling or processing of patient-related medical or healthcare data
    • G16H10/40ICT specially adapted for the handling or processing of patient-related medical or healthcare data for data related to laboratory analysis, e.g. patient specimen analysis
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H10/00ICT specially adapted for the handling or processing of patient-related medical or healthcare data
    • G16H10/60ICT specially adapted for the handling or processing of patient-related medical or healthcare data for patient-specific data, e.g. for electronic patient records
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/10ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to drugs or medications, e.g. for ensuring correct administration to patients
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H20/00ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance
    • G16H20/40ICT specially adapted for therapies or health-improving plans, e.g. for handling prescriptions, for steering therapy or for monitoring patient compliance relating to mechanical, radiation or invasive therapies, e.g. surgery, laser therapy, dialysis or acupuncture
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/30ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for calculating health indices; for individual health risk assessment
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H50/00ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics
    • G16H50/70ICT specially adapted for medical diagnosis, medical simulation or medical data mining; ICT specially adapted for detecting, monitoring or modelling epidemics or pandemics for mining of medical data, e.g. analysing previous cases of other patients
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16HHEALTHCARE INFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR THE HANDLING OR PROCESSING OF MEDICAL OR HEALTHCARE DATA
    • G16H70/00ICT specially adapted for the handling or processing of medical references
    • G16H70/60ICT specially adapted for the handling or processing of medical references relating to pathologies
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B25/00ICT specially adapted for hybridisation; ICT specially adapted for gene or protein expression
    • G16B25/10Gene or protein expression profiling; Expression-ratio estimation or normalisation
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A90/00Technologies having an indirect contribution to adaptation to climate change
    • Y02A90/10Information and communication technologies [ICT] supporting adaptation to climate change, e.g. for weather forecasting or climate simulation

Definitions

  • the field of the invention is profiling of omics data as they relate to cancer, especially as it relates to the generation of indicators for cancer prognosis, prediction of treatment outcomes, and/or effectiveness of cancer treatments.
  • Cancer is a multifactorial disease where many diverse genetic and environmental factors interplay and contribute to the development and outcome of the disease.
  • genetic and environmental factors often affect the patient's prognosis in various degrees such that individual patients may show different responses to the same therapeutic and/or prophylactic treatment.
  • Such complexity and diversity render traditional prediction of prognosis, identification of optimal treatments, and prediction of likelihood of success of the treatments based on a single or few factors (e.g., serum level of inflammation-related proteins, etc.), often unreliable.
  • many traditional methods of examining such factors are invasive as they require tumor biopsy samples for histology of tumor cells and tissues.
  • the inventive subject matter is directed to methods of using various omics data of cell free nucleic acids to calculate a composite cancer score that can be used to determine the status, prognosis of a cancer as well as likelihood of treatment outcome and/or effectiveness of current treatments.
  • one aspect of the subject matter includes a method of analyzing omics data.
  • blood is obtained from a patient having or suspected to have a cancer.
  • omics data for a plurality of cancer-related genes are obtained.
  • the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data.
  • a composite score is calculated which can then be associated with at least one of a health status, an omics error status, a cancer prognosis, a therapeutic recommendation, and an effectiveness of a treatment.
  • the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status.
  • the DNA sequence data is obtained from circulating free DNA.
  • the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA.
  • the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease.
  • the neoepitope is tumor-specific and patient-specific.
  • the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In such embodiments, it is preferred that the presence of the mutation in the cancer-specific gene weighs more than the presence of the mutation in the cancer-related genes other than the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio between or among a plurality of splice variants of the cancer gene.
  • the method further comprises a step of comparing the score with a threshold value to thereby determine the therapeutic recommendation.
  • the therapeutic recommendation is a prophylactic treatment if the score is below the threshold value.
  • the method further comprises a step of comparing the omics error status with a threshold value to thereby determine a risk score.
  • the inventors contemplate a method of determining prognosis of a cancer of a patient.
  • blood is obtained from a patient having or suspected to have a cancer.
  • omics data for a plurality of cancer genes are obtained.
  • the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data.
  • a cancer prognosis score is calculated, and the prognosis of the cancer is provided based on the cancer prognosis score.
  • the prognosis comprises a progress of metastasis.
  • the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status.
  • the DNA sequence data is obtained from circulating free DNA.
  • the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA.
  • the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease.
  • the neoepitope is tumor-specific and patient-specific.
  • the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio among or between a plurality of splice variants of the cancer gene.
  • the omics data is a plurality of sets of omics data obtained at a different time points during a time period
  • the prognosis is provided based on a plurality of scores from the plurality of sets of omics data.
  • it is preferred that the prognosis is represented by a change of a plurality of scores during the time period, wherein the change is over a predetermined threshold value.
  • Still another aspect of inventive subject matter is directed towards a method of predicting an outcome of a treatment for a cancer patient.
  • blood is obtained from a patient having a cancer.
  • omics data for a plurality of cancer genes are obtained.
  • the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data.
  • a cancer gene score is calculated, and a predicted outcome of the treatment is provided based on the cancer prognosis score.
  • the predicted outcome is determined by comparing the cancer gene score with a predetermined threshold value.
  • the treatment is a drug, and at least one of the plurality of cancer gene is a predicted target of the drug.
  • the treatment is an immune therapy, and at least one of the plurality of cancer gene is a receptor of an immune cell or a ligand of the receptor.
  • the treatment is a surgery or a radiation therapy, and at least one of the plurality of cancer gene is a neoepitope that is tumor-specific and patient-specific.
  • the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status.
  • the DNA sequence data is obtained from circulating free DNA.
  • the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA.
  • the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease.
  • the neoepitope is tumor-specific and patient-specific.
  • the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio between a plurality of splice variants of the cancer gene.
  • the inventors contemplate a method of evaluating an effectiveness of a treatment for a cancer patient.
  • blood is obtained from a patient having a cancer.
  • omics data for a plurality of cancer genes are obtained before and after the treatment.
  • the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data.
  • the effectiveness of the treatment is provided based on the comparison of the at least two cancer gene scores.
  • the effectiveness of the treatment can be determined by a difference between the cancer gene score before and after the treatment. In such embodiments, it is preferred that the treatment is determined effective when the difference is higher than a predetermined threshold value.
  • the treatment is a drug, and at least one of the plurality of cancer gene is a predicted target of the drug.
  • the treatment is an immune therapy, and at least one of the plurality of cancer gene is a receptor of an immune cell or a ligand of the receptor.
  • the treatment is a surgery or a radiation therapy, and at least one of the plurality of cancer gene is a neoepitope that is tumor-specific and patient-specific.
  • the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status.
  • the DNA sequence data is obtained from circulating free DNA.
  • the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA.
  • the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease.
  • the neoepitope is tumor-specific and patient-specific.
  • the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio between a plurality of splice variants of the cancer gene.
  • the inventors discovered that the status and/or prognosis of a cancer can be more reliably determined in a less invasive and quick manner using a compound score that is generated based on multiple factors associated with the cancer.
  • the inventors also discovered that the compound score can be used to reliably predict a likelihood of outcome of a cancer treatment, and further, effectiveness of a particular cancer treatment.
  • a compound score can be generated from the patient's omics data obtained from nucleic acids in the patient's blood.
  • the omics data include omics data of various cancer-related genes, which can be differentially weighed based on the type and timing of the sampling.
  • the compound score can be a reliable indicator to determine cancer status and/or prognosis of a cancer, a likelihood of outcome of a cancer treatment. Further, the compound scores generated based on omics data obtained before and after a cancer treatment can be compared to determine the effectiveness of a cancer treatment.
  • tumor refers to, and is interchangeably used with one or more cancer cells, cancer tissues, malignant tumor cells, or malignant tumor tissue, that can be placed or found in one or more anatomical locations in a human body.
  • a patient includes both individuals that are diagnosed with a condition (e.g., cancer) as well as individuals undergoing examination and/or testing for the purpose of detecting or identifying a condition.
  • a patient having a tumor refers to both individuals that are diagnosed with a cancer as well as individuals that are suspected to have a cancer.
  • the term “provide” or “providing” refers to and includes any acts of manufacturing, generating, placing, enabling to use, transferring, or making ready to use.
  • the patient's bodily fluid includes, but is not limited to, blood, serum, plasma, mucus, cerebrospinal fluid, ascites fluid, saliva, and urine of the patient.
  • various other bodily fluids are also deemed appropriate so long as cell free DNA/RNA is present in such fluids.
  • the patient's bodily fluid may be fresh or preserved/frozen.
  • Appropriate fluids include saliva, ascites fluid, spinal fluid, urine, etc., which may be fresh or preserved/frozen.
  • the cell free RNA may include any types of DNA/RNA that are circulating in the bodily fluid of a person without being enclosed in a cell body or a nucleus.
  • the source of the cell free DNA/RNA is the tumor cells.
  • the source of the cell free DNA/RNA is an immune cell (e.g., NK cells, T cells, macrophages, etc.).
  • the cell free DNA/RNA can be circulating tumor DNA/RNA (ctDNA/RNA) and/or circulating free DNA/RNA (cf DNA/RNA, circulating nucleic acids that do not derive from a tumor).
  • the cell free DNA/RNA may be enclosed in a vesicular structure (e.g., via exosomal release of cytoplasmic substances) so that it can be protected from nuclease (e.g., RNAase) activity in some type of bodily fluid.
  • nuclease e.g., RNAase
  • the cell free DNA/RNA is a naked DNA/RNA without being enclosed in any membranous structure, but may be in a stable form by itself or be stabilized via interaction with one or more non-nucleotide molecules (e.g., any RNA binding proteins, etc.).
  • non-nucleotide molecules e.g., any RNA binding proteins, etc.
  • the cell free DNA/RNA can be any type of DNA/RNA which can be released from either cancer cells or immune cell.
  • the cell free DNA may include any whole or fragmented genomic DNA, or mitochondrial DNA
  • the cell free RNA may include mRNA, tRNA, microRNA, small interfering RNA, long non-coding RNA (lncRNA).
  • the cell free DNA is a fragmented DNA typically with a length of at least 50 base pair (bp), 100 base pair (bp), 200 bp, 500 bp, or 1 kbp.
  • the cell free RNA is a full length or a fragment of mRNA (e.g., at least 70% of full-length, at least 50% of full length, at least 30% of full length, etc.). While cell free DNA/RNA may include any type of DNA/RNA encoding any cellular, extracellular proteins or non-protein elements, it is preferred that at least some of cell free DNA/RNA encodes one or more cancer-related proteins, or inflammation-related proteins.
  • the cell free DNA/mRNA may be full-length or fragments of (or derived from the) cancer related genes including, but not limited to ABL1, ABL2, ACTB, ACVR1B, AKT1, AKT2, AKT3, ALK, AMER11, APC, AR, ARAF, ARFRP1, ARID1A, ARID1B, ASXL1, ATF1, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, AXL, BAP1, BARD1, BCL2, BCL2L1, BCL2L2, BCL6, BCOR, BCORL1, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRD4, BRIP1, BTG1, BTK, EMSY, CARD11, CBFB, CBL, CCND1, CCND2, CCND3, CCNE1, CD274, CD79A, CD79B, CDC73, CDH1, CDK12, CDK4, CDK6, CDK8, CDKN1A, CDKN1B, CDK
  • genes may be wild type or mutated versions, including missense or nonsense mutations, insertions, deletions, fusions, and/or translocations, all of which may or may not cause formation of full-length mRNA when transcribed.
  • some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of inflammation-related proteins, including, but not limited to, HMGB1, HMGB2, HMGB3, MUC1, VWF, MMP, CRP, PBEF1, TNF- ⁇ , TGF- ⁇ , PDGFA, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, Eotaxin, FGF, G-CSF, GM-CSF, IFN-7, IP-10, MCP-1, PDGF, and hTERT, and in yet another example, the cell free mRNA encoded a full length or a fragment of HMGB1.
  • some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of DNA repair-related proteins or RNA repair-related proteins.
  • Table 1 provides an exemplary collection of predominant RNA repair genes and their associated repair pathways contemplated herein, but it should be recognized that numerous other genes associated with DNA repair and repair pathways are also expressly contemplated herein, and Tables 2 and 3 illustrate further exemplary genes for analysis and their associated function in DNA repair.
  • double-strand break repair /// mitotic recombination /// meiotic recombination /// DNA repair /// DNA recombination /// response to DNA damage stimulus XRCC4 X-ray repair complementing DNA repair /// double-strand break repair /// DNA defective repair in Chinese hamster recombination /// DNA recombination /// response cells 4 to DNA damage stimulus XRCC4 X-ray repair complementing DNA repair /// double-strand break repair /// DNA defective repair in Chinese hamster recombination /// DNA recombination /// response cells 4 to DNA damage stimulus RAD17 RAD17 homolog ( S.
  • some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of a gene not associated with a disease (e.g., housekeeping genes), including, but not limited to, those related to transcription factors (e.g., ATF1, ATF2, ATF4, ATF6, ATF7, ATFIP, BTF3, E2F4, ERH, HMGB1, ILF2, IER2, JUND, TCEB2, etc.), repressors (e.g., PUF60), RNA splicing (e.g., BAT1, HNRPD, HNRPK, PABPN1, SRSF3, etc.), translation factors (EIF1, EIF1AD, EIF1B, EIF2A, EIF2AK1, EIF2AK3, EIF2AK4, EIF2B2, EIF2B3, EIF2B4, EIF2S2, EIF3A, etc.), tRNA synthetases (e.g., AARS, CARS, DARS
  • transcription factors
  • ATP2C1, ATP5F1, etc. lysosome
  • proteasome e.g., PSMA1, UBA1, etc.
  • cytoskeletal proteins e.g., ANXA6, ARPC2, etc.
  • organelle synthesis e.g., BLOC1S1, AP2A1, etc.
  • some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of a neoepitope specific to the tumor.
  • neoepitope it should be appreciated that neoepitopes can be characterized as random mutations in tumor cells that create unique and tumor specific antigens. Therefore, high-throughput genome sequencing should allow for rapid and specific identification of patient specific neoepitopes where the analysis also considers matched normal tissue of the same patient.
  • neoepitopes may be identified from a patient tumor in a first step by whole genome analysis of a tumor biopsy (or lymph biopsy or biopsy of a metastatic site) and matched normal tissue (i.e., non-diseased tissue from the same patient) via synchronous comparison of the so obtained omics information.
  • the data are patient matched tumor data (e.g., tumor versus same patient normal), and that the data format is in SAM, BAM, GAR, or VCF format.
  • non-matched or matched versus other reference e.g., prior same patient normal or prior same patient tumor, or Homo statisticus ) are also deemed suitable for use herein.
  • the omics data may be ‘fresh’ omics data or omics data that were obtained from a prior procedure (or even different patient). However, and especially where genomics ctDNA is analyzed, the neoepitope-coding sequence need not necessarily be expressed.
  • the nucleic acid encoding a neoepitope may encode a neoepitope that is also a suitable target for immune therapy. Therefore, neoepitopes can then be further filtered for a match to the patient's HLA type to thereby increase likelihood of antigen presentation of the neoepitope. Most preferably, and as further discussed below, such matching can be done in silico.
  • the patient-specific epitopes are unique to the patient, but may also in at least some cases include tumor type-specific neoepitopes (e.g., Her-2, PSA, brachyury) or cancer-associated neoepitopes (e.g., CEA, MUC-1, CYPB1).
  • tumor type-specific neoepitopes e.g., Her-2, PSA, brachyury
  • cancer-associated neoepitopes e.g., CEA, MUC-1, CYPB1
  • cell free DNA/mRNA may present in modified forms or different isoforms.
  • the cell free DNA may be present in methylated or hydroxyl methylated, and the methylation level of some genes (e.g., GSTP1, p16, APC, etc.) may be a hallmark of specific types of cancer (e.g., colorectal cancer, etc.).
  • the cell free mRNA may be present in a plurality of isoforms (e.g., splicing variants, etc.) that may be associated with different cell types and/or location.
  • different isoforms of mRNA may be a hallmark of specific tissues (e.g., brain, intestine, adipose tissue, muscle, etc.), or may be a hallmark of cancer (e.g., different isoform is present in the cancer cell compared to corresponding normal cell, or the ratio of different isoforms is different in the cancer cell compared to corresponding normal cell, etc.).
  • tissue e.g., brain, intestine, adipose tissue, muscle, etc.
  • cancer e.g., different isoform is present in the cancer cell compared to corresponding normal cell, or the ratio of different isoforms is different in the cancer cell compared to corresponding normal cell, etc.
  • mRNA encoding HMGB1 are present in 18 different alternative splicing variants and 2 unspliced forms.
  • isoforms are expected to express in different tissues/locations of the patient's body (e.g., isoform A is specific to prostate, isoform B is specific to brain, isoform C is specific to spleen, etc.).
  • identifying the isoforms of cell free mRNA in the patient's bodily fluid can provide information on the origin (e.g., cell type, tissue type, etc.) of the cell free mRNA.
  • regulatory noncoding RNA e.g., microRNA, small interfering RNA, long non-coding RNA (lncRNA)
  • lncRNA long non-coding RNA
  • varied expression of regulatory noncoding RNA in a cancer patient's bodily fluid may due to genetic modification of the cancer cell (e.g., deletion, translocation of parts of a chromosome, etc.), and/or inflammations at the cancer tissue by immune system (e.g., regulation of miR-29 family by activation of interferon signaling and/or virus infection, etc.).
  • the cell free RNA can be a regulatory noncoding RNA that modulates expression (e.g., downregulates, silences, etc.) of mRNA encoding a cancer-related protein or an inflammation-related protein (e.g., HMGB1, HMGB2, HMGB3, MUC1, VWF, MMP, CRP, PBEF1, TNF- ⁇ , TGF- ⁇ , PDGFA, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, Eotaxin, FGF, G-CSF, GM-CSF, IFN-7, IP-10, MCP-1, PDGF, hTERT, etc.).
  • a regulatory noncoding RNA that modulates expression (e.g., downregulates, silences, etc.) of mRNA encoding a cancer-related protein or an inflammation-related protein (e.g.
  • some cell free regulatory noncoding RNA may be present in a plurality of isoforms or members (e.g., members of miR-29 family, etc.) that may be associated with different cell types and/or location.
  • different isoforms or members of regulatory noncoding RNA may be a hallmark of specific tissues (e.g., brain, intestine, adipose tissue, muscle, etc.), or may be a hallmark of cancer (e.g., different isoform is present in the cancer cell compared to corresponding normal cell, or the ratio of different isoforms is different in the cancer cell compared to corresponding normal cell, etc.).
  • identifying the isoforms of cell free regulatory noncoding RNA in the patient's bodily fluid can provide information on the origin (e.g., cell type, tissue type, etc.) of the cell free regulatory noncoding RNA.
  • cell free DNA/RNA is isolated from a bodily fluid (e.g., whole blood) that is processed under a suitable conditions, including a condition that stabilizes cell free RNA.
  • a bodily fluid e.g., whole blood
  • both cell free DNA and RNA are isolated simultaneously from the same badge of the patient's bodily fluid.
  • the bodily fluid sample can be divided into two or more smaller samples from which DNA or RNA can be isolated separately.
  • the bodily fluid of the patient can be obtained at any desired time point(s) depending on the purpose of the omics analysis.
  • the bodily fluid of the patient can be obtained before and/or after the patient is confirmed to have a tumor and/or periodically thereafter (e.g., every week, every month, etc.) in order to associate the cell free DNA/RNA data with the prognosis of the cancer.
  • the bodily fluid of the patient can be obtained from a patient before and after the cancer treatment (e.g., chemotherapy, radiotherapy, drug treatment, cancer immunotherapy, etc.). While it may vary depending on the type of treatments and/or the type of cancer, the bodily fluid of the patient can be obtained at least 24 hours, at least 3 days, at least 7 days after the cancer treatment.
  • the bodily fluid from the patient before the cancer treatment can be obtained less than 1 hour, less than 6 hours before, less than 24 hours before, less than a week before the beginning of the cancer treatment.
  • a plurality of samples of the bodily fluid of the patient can be obtained during a period before and/or after the cancer treatment (e.g., once a day after 24 hours for 7 days, etc.).
  • the bodily fluid of a healthy individual can be obtained to compare the sequence/modification of cell free DNA, and/or quantity/subtype expression of cell free RNA.
  • a healthy individual refers an individual without a tumor.
  • the healthy individual can be chosen among group of people shares characteristics with the patient (e.g., age, gender, ethnicity, diet, living environment, family history, etc.).
  • any suitable methods for isolating cell free DNA/RNA are contemplated.
  • specimens were accepted as 10 ml of whole blood drawn into a test tube.
  • Cell free DNA can be isolated from other from mono-nucleosomal and di-nucleosomal complexes using magnetic beads that can separate out cell free DNA at a size between 100-300 bps.
  • specimens were accepted as 10 ml of whole blood drawn into cell-free RNA BCT® tubes or cell-free DNA BCT® tubes containing RNA stabilizers, respectively.
  • cell free RNA is stable in whole blood in the cell-free RNA BCT tubes for seven days while cell free RNA is stable in whole blood in the cell-free DNA BCT Tubes for fourteen days, allowing time for shipping of patient samples from world-wide locations without the degradation of cell free RNA.
  • the cell free RNA is isolated using RNA stabilization agents that will not or substantially not (e.g., equal or less than 1%, or equal or less than 0.1%, or equal or less than 0.01%, or equal or less than 0.001%) lyse blood cells.
  • the RNA stabilization reagents will not lead to a substantial increase (e.g., increase in total RNA no more than 10%, or no more than 5%, or no more than 2%, or no more than 1%) in RNA quantities in serum or plasma after the reagents are combined with blood.
  • these reagents will also preserve physical integrity of the cells in the blood to reduce or even eliminate release of cellular RNA found in blood cell. Such preservation may be in form of collected blood that may or may not have been separated.
  • contemplated reagents will stabilize cell free RNA in a collected tissue other than blood for at 2 days, more preferably at least 5 days, and most preferably at least 7 days.
  • numerous other collection modalities are also deemed appropriate, and that the cell free RNA can be at least partially purified or adsorbed to a solid phase to so increase stability prior to further processing.
  • fractionation of plasma and extraction of cell free DNA/RNA can be done in numerous manners.
  • whole blood in 10 mL tubes is centrifuged to fractionate plasma at 1600 rcf for 20 minutes.
  • the so obtained plasma is then separated and centrifuged at 16,000 rcf for 10 minutes to remove cell debris.
  • various alternative centrifugal protocols are also deemed suitable so long as the centrifugation will not lead to substantial cell lysis (e.g., lysis of no more than 1%, or no more than 0.1%, or no more than 0.01%, or no more than 0.001% of all cells).
  • Cell free RNA is extracted from 2 mL of plasma using Qiagen reagents.
  • the extraction protocol was designed to remove potential contaminating blood cells, other impurities, and maintain stability of the nucleic acids during the extraction. All nucleic acids were kept in bar-coded matrix storage tubes, with DNA stored at ⁇ 4° C. and RNA stored at ⁇ 80° C. or reverse-transcribed to cDNA that is then stored at ⁇ 4° C. Notably, so isolated cell free RNA can be frozen prior to further processing.
  • DNA sequence data will not only include the presence or absence of a gene that is associated with cancer or inflammation, but also take into account mutation data where the gene is mutated, the copy number (e.g., to identify duplication, loss of allele or heterozygosity), and epigenetic status (e.g., methylation, histone phosphorylation, nucleosome positioning, etc.).
  • mutation data e.g., to identify duplication, loss of allele or heterozygosity
  • epigenetic status e.g., methylation, histone phosphorylation, nucleosome positioning, etc.
  • contemplated RNA sequence data include mRNA sequence data, splice variant data, polyadenylation information, etc.
  • the RNA sequence data also include a metric for the transcription strength (e.g., number of transcripts of a damage repair gene per million total transcripts, number of transcripts of a damage repair gene per total number of transcripts for all damage repair genes, number of transcripts of a damage repair gene per number of transcripts for actin or other household gene RNA, etc.), and for the transcript stability (e.g., a length of poly A tail, etc.).
  • a metric for the transcription strength e.g., number of transcripts of a damage repair gene per million total transcripts, number of transcripts of a damage repair gene per total number of transcripts for all damage repair genes, number of transcripts of a damage repair gene per number of transcripts for actin or other household gene RNA, etc.
  • transcript stability e.g., a length of poly A tail, etc.
  • transcription strength of the cell free RNA can be examined by quantifying the cell free RNA.
  • Quantification of cell free RNA can be performed in numerous manners, however, expression of analytes is preferably measured by quantitative real-time RT-PCR of cell free RNA using primers specific for each gene. For example, amplification can be performed using an assay in a 10 ⁇ L reaction mix containing 2 ⁇ L cell free RNA, primers, and probe. mRNA of ⁇ -actin can be used as an internal control for the input level of cell free RNA. A standard curve of samples with known concentrations of each analyte was included in each PCR plate as well as positive and negative controls for each gene.
  • Test samples were identified by scanning the 2D barcode on the matrix tubes containing the nucleic acids.
  • Delta Ct was calculated from the Ct value derived from quantitative PCR (qPCR) amplification for each analyte subtracted by the Ct value of actin for each individual patient's blood sample.
  • Relative expression of patient specimens is calculated using a standard curve of delta Cts of serial dilutions of Universal Human Reference RNA set at a gene expression value of 10 (when the delta CTs were plotted against the log concentration of each analyte).
  • real time quantitative PCR may be replaced by RNAseq to so cover at least part of a patient transcriptome.
  • analysis can be performed static or over a time course with repeated sampling to obtain a dynamic picture without the need for biopsy of the tumor or a metastasis.
  • genomic data of cell free DNA/RNA preferably comprise a genomic data set that includes genomic sequence information.
  • the genomic sequence information comprises DNA sequence information of cell free DNA of the patient and optionally cell free DNA of a healthy individual.
  • the sequence data sets may include unprocessed or processed data sets, and exemplary data sets include those having BAM format, SAM format, FASTQ format, or FASTA format.
  • BAM format or as BAMBAM diff objects (see e.g., US2012/0059670A1 and US2012/0066001A1).
  • the data sets are reflective of the cell free DNA/RNA of the patient and of the healthy individual to so obtain patient and tumor specific information.
  • omics information can then be processed using pathway analysis (especially using PARADIGM) to identify any impact of any mutations on DNA repair pathways.
  • sequence data may be performed in numerous manners. In most preferred methods, however, analysis is performed in silico by location-guided synchronous alignment of cell free DNA/RNA of the patient and a healthy individual as, for example, disclosed in US 2012/0059670A1 and US 2012/0066001A1 using BAM files and BAM servers. Such analysis advantageously reduces false positive data and significantly reduces demands on memory and computational resources.
  • differential sequence object is generated by incremental synchronous alignment of BAM files representing genomic sequence information of the cell free DNA/RNA of the patient and a healthy individual.
  • particularly preferred methods include BAMBAM-based methods as described in US 2012/0059670 and US 2012/0066001.
  • a library or reference base for all cancer-related genes, inflammation-related genes, DNA repair-related genes, and/or other non-disease related housekeeping genes can be created using one or more omics data for each of those genes, and such library is particularly useful where the omics data are associated with one or more health parameter.
  • such library can provide a tool to generate a large cross-sectional database for all cancer-related gene activity, inflammation-related gene activity, DNA repair gene activity and housekeeping gene activity (as a control).
  • the large cross-sectional database can be a basis for generating a cancer matrix, based on which a prognosis of a cancer, a health status of the patient, a likelihood of outcome of treatment, an effectiveness of the treatment can be more reliably calculated.
  • analyses presented herein may be performed over specific and diverse populations to so obtain reference values for the specific populations, such as across various health associated states (e.g., healthy, diagnosed with a specific disease and/or disease state, which may or may not be inherited, or which may or may not be associated with impaired DNA repair, inflammation-related autoimmunity, etc.), a specific age or age bracket, a specific ethnic group that may or may not be associated with frequent occurrence of specific type of cancer.
  • populations may also be enlisted from databases with known omics information, and especially publically available omics information from cancer patients (e.g., TCGA, COSMIC, etc.) and proprietary databases from a large variety of individuals that may be healthy or diagnosed with a disease.
  • the population records may also be indexed over time for the same individual or group of individuals, which advantageously allows detection of shifts or changes in the genes and pathways associated with different types of cancers.
  • a cancer score can be established for one or more cancer-related genes, inflammation-related genes, a DNA-repair gene, a neoepitope, and a gene not associated with a disease and that the score may be reflective of or even prognostic for various types of cancer that are at least in part due to mutations in cancer-related genes and/or pathways.
  • suitable cancer scores may involve scores for one or more genes associated with one or more types of cancer (e.g., BRCA1, BRCA2, P53, etc.) relative to another gene that may or may not be associated with one type of cancer (e.g., housekeeping genes, etc.).
  • contemplated cancer scores may involve scores for one or more genes associated with one or more types of one or more types of cancer (e.g., BRCA1, BRCA2, P53, etc.) relative to an overall mutation rate (e.g., mutation rate of the genes not associated with a disease, etc.) to so better identify cancer relevant mutations over ‘background’ mutations.
  • an overall mutation rate e.g., mutation rate of the genes not associated with a disease, etc.
  • the omics data may be used to generate a general error status for an individual (or tumor within an individual), or to associate the number and/or type of alterations in cancer-related genes, inflammation-related genes, or a DNA-repair gene to identify a ‘tipping point’ for one or more gene mutations after which a general mutation rate skyrockets.
  • a rate or number of mutations in ERCC1 and other DNA repair genes could have only minor systemic consequence, addition of further mutations to TP53 may result in a catastrophic increase in mutation rates.
  • mutations in the genes associated with DNA may be used to estimate the risk of occurrence for a DNA damage-based disease, and especially cancer and age-related diseases.
  • omics information may be analyzed in one or more pathway analysis algorithms (e.g., PARADIGM) to so identify affected pathways and to so possibly adjust treatment where treatment employs DNA damaging agents.
  • Pathway analysis algorithms may also be used to in silico modulate expression of one or more DNA repair genes, which may results in desirable or even unexpected in silico treatment outcomes, which may be translated into the clinic.
  • the cancer score is typically a compound score reflecting status of a plurality of genes.
  • the cancer score can be calculated by counting any mutations (e.g., deletion, missense, nonsense, etc.) of any cancer-related genes, inflammation-related genes, and DNA-repair genes with one or more mutations as having a positive value, counting any changes in methylation or other modifications in DNA of counting any cancer-related genes, DNA-repair genes, counting any upregulation or downregulation in expression levels of RNA of any cancer-related genes, inflammation-related genes, and DNA-repair genes, counting any presence of tumor-specific, patient specific neoepitopes, counting any changes or ratios in RNA isotypes (splice variants) of counting any cancer-related genes and DNA-repair genes, and counting any changes in length of poly A tail of any cancer-related genes, inflammation-related genes, and DNA-repair genes.
  • any mutations e.g., deletion, missense, nonsense, etc.
  • each count may be weighed uniformly or biased, based on the significance of each count and then be assigned a value according to the weight of each count (e.g., each count corresponds to 1 point, some counts correspond to different scores such as 1 point, 3 points, 10 points, 100 points, etc.).
  • Some mutations in some cancer related genes may be ‘leading indicators’ or triggers to activate other tumorigenesis mechanism or metastasis. Identification of such triggers may advantageously allow for early diagnosis or intervention of the cancer.
  • a mutation in a cancer-specific gene among cancer-related genes, inflammation-related genes, or DNA-repair genes may be weighed higher than other cancer-related genes or DNA-repair genes (e.g., at least 3 times, at least 5 times, at least 10 times, at least 100 times, etc.) and can be assigned to higher values accordingly.
  • the cancer-specific gene refers any gene or mutation of the gene that is a known genetic disposition (e.g., significantly increase a susceptibility to the disease) of specific types of cancer (e.g., BRCA1 and BRCA2 for breast cancer and ovarian cancer, etc.).
  • each gene in any cancer-related pathway or DNA-repair pathway may be differently weighed (e.g., most significant, significant, moderate, less significant, insignificant, etc.) and any mutation of a such gene that has any or no impact (e.g., adversely affect the pathway stream, etc.) on any cancer-related pathway or DNA-repair pathway may be weighed differently based on the significance of the impact.
  • gene A encoding a significant, unreplaceable protein A in a cancer pathway may be weighed heavier than another gene B encoding a redundant protein (replaceable with other proteins).
  • a nonsense mutation in gene A that results in nonfunctional protein may be weighed at least 3 times, at least 5 times, at least 10 times, at least 100 times than a silent mutation in gene A or a missense mutation which does not affect the function of protein A and can be assigned to higher values accordingly.
  • some countings may weigh equally or differently based on the significance of each counting and then be assigned to a negative value according to the weight of each counting (e.g., each counting corresponds to ⁇ 1 point, some countings correspond to different scores such as ⁇ 1 point, ⁇ 3 points, ⁇ 10 points, ⁇ 100 points, etc.).
  • upregulation of mRNA of gene C which can compensate the loss of function of gene A, can be assigned to a negative value (e.g., ⁇ 10 points) such that it can compensate the positive value of mutation of gene A (e.g., +10 points).
  • some countings may be differently weighed based on the degree of changes in expression level of some RNAs. For example, when the expression level of RNA “X” increases at least twice, at least 5 times, at least 10 times, at least 20 times, while other RNA expression level change is below 50% at best, then the increase of expression level of RNA “X” may be weighed at least 3 times, at least 5 times, at least 10 times, at least 100 times than other genes.
  • the cancer score is compound score that is a total sum of all values assigned to all counts.
  • the cancer score can be a total sum of all values assigned to all counts (all omics data).
  • the cancer score can be a total sum of a selected number of values assigned to some counts (e.g., corresponding to specific pathways, specific types of genes, specific groups of mechanisms, etc.).
  • the cancer score increases as more cancer-related genes or DNA-repair genes possess one or more mutations.
  • each mutation and/or change may be counted separately such that cancer scores may further increase where one or more cancer-related genes or DNA-repair genes show multiple mutations in a single gene.
  • cancer score may further increase when such multiple mutations in a single gene may further affect the function of the cancer-related genes or DNA-repair genes such that the multiple mutations drive the cells more cancer-prone, or more cancerous, or drive the cancer microenvironment more immune-resistant, and so on.
  • the cancer score can be presented as a trajectory with one or more counts as its vectors, where a few numbers of variables and/or factors dominantly govern in determination of cancer prognosis.
  • Each of variables and/or factors can be presented as a vector, whose amplitude is corresponding to the point of each weighted counting, and the addition of those vectors provides a trajectory indicating the prognosis of the disease.
  • multiple analyses over time can be prepared for the same patient, and that changes over time (e.g., with or without treatment) may be assigned specific values that will yet again generate a time-dependent score. Such scores or changes over time may be classified and serve as leading indicator for treatment outcome, drug response, etc.
  • the cancer score can be calculated with health information other than cf/ct nucleic acid data obtained from the patient's blood.
  • the health information may include expression levels/concentrations of several types of cytokines (e.g., IL-2, TNF-a, etc.) related to tumorigenesis/inflammation/immune response against the tumor, hormone levels (e.g., estrogen, progesterone, growth hormone, etc.), blood sugar level, alanine transaminase level (for liver function), creatine level (for kidney function), blood pressure, types and quantity of tumor cell-secreted proteins (e.g., soluble ligands of immune cell receptor, etc.) or foreign antigenic proteins (e.g., for virus or bacterial infection, etc.).
  • cytokines e.g., IL-2, TNF-a, etc.
  • hormone levels e.g., estrogen, progesterone, growth hormone, etc.
  • blood sugar level e.g., alanine transamina
  • the so obtained cancer score can be used to provide a diagnosis of cancer or risk of having or developing a cancer.
  • the calculated cancer score of a patient can be compared with an average cancer score of healthy individuals to determine the difference between two scores.
  • the patient may be diagnosed to have a tumor, or has a high risk to have a tumor.
  • the calculated cancer score of a patient can be compared with a predetermined threshold score.
  • the predetermined threshold score can be a predetermined score, which may vary depending on patient's ethnicity, age, gender, or other health status.
  • the predetermined threshold score can a dynamic score that can be changed based on a previous cancer score and a diagnosis or treatment performed to the patient.
  • the so obtained cancer score can be used to provide a prognosis of the cancer.
  • the cancer scores can be calculated based on omics data obtained in month 1, month 3, month 6, and month 12 after the patient got diagnosed with a first stage of lung cancer, and each cancer score can be compared with a predetermined threshold score corresponding to the month 1, 3, 6, and 12.
  • the cancer scores are about 120% of the threshold score in month 1 and 3, and the cancer score is about 180% in month 6, and 230% of the threshold score month 12.
  • the cancer score can be calculated by highly weighing the presence of neoepitopes that are tumor-specific and patient-specific.
  • the cancer scores can be calculated based on omics data obtained in month 1, month 3, month 6, and month 12 after the patient got diagnosed with a first stage of lung cancer, and each cancer score is calculated by highly weighing the presence/appearance of new epitope that is tumor/tissue specific.
  • the cancer scores are about 120% of the threshold score in month 1 and 3, and the cancer score is about 140% in month 6, and 230% of the threshold score month 12.
  • the cancer scores can provide an indicator for treatment options.
  • the treatment option may be a prophylactic treatment where the compound score is below the threshold value, indicating that the patient is unlikely to have a tumor for now or at least has low risk of developing a tumor.
  • a cancer-related gene A e.g., over a threshold such as at least 10%, at least 20%, at least 30%, at least 50%, etc.
  • the cancer score can be used to provide the treatment option that may use a drug inhibiting the activity of cancer-related gene A (e.g., a blocker of protein A, etc.).
  • cancer score when the cancer score is above the threshold value and a majority portion of the cancer score highly weighted was overexpression of a gene encoding a receptor of an immune cell or a ligand of the receptor, then the cancer score can be used to provide the immunotherapy using the receptor or ligand of the immune cells. Also, when the cancer score is above the threshold value and a majority portion of the cancer score highly weighted was overexpression of a specific neoepitope, then the cancer score can be used to provide the immunotherapy using the neoepitope as a bait or a surgery/a radiation therapy to physically remove local tumors. Also such cancer scores may be an indicative of likelihood of success for the treatment option. However, if the portion of the cancer score highly weighted was overexpression of a cancer-related gene A is below the threshold, then the treatment option using a drug inhibiting the activity of cancer-related gene A may be predicted less effective.
  • the patient can be treated with at least one of the treatment options based on the patient's cancer (compound) score.
  • the treatment option can be selected to include a recombinant virus (or yeast or bacteria) comprising a nucleic acid encoding the specific neoepitope.
  • the recombinant virus can be administered to the patient in a dose and schedule effective to treat the tumor and/or effective to reduce the cancer score of the patient for at least 10%, at least 20%, at least 30%, at least in 2 weeks, at least in 4 weeks, at least in 8 weeks, at least in 12 weeks after the administration or a series of administrations.
  • the patient's cancer score can be compared with one or more other patients having same type of cancer and having a treatment history to provide a treatment option and predicted outcome. For example, where other patients' history indicates that the drug treatment is effective only when the cancer score is below 200 (as absolute score), or less than 180% of the healthy individual's score, and the patient's cancer score has been increasing from 140 to 160 for the last 2 weeks, a recommendation to proceed with drug treatment no later than 2 weeks can be provided based on the other patients' history and cancer scores.
  • the calculated cancer score can also be an indicator of an effectiveness of a cancer treatment, especially when the omics data includes information of at least one or more genes encoding a target/indicator of the cancer treatment.
  • cancer scores can be calculated based on omics data obtained before the cancer treatment, 7 days after, 2 weeks, 1 month, and 6 months of the cancer treatment.
  • the cancer score of 7 days after the treatment is 80% of the cancer score before the treatment, and the cancer score of 2 weeks and 1 month after the treatment is 50% of the cancer score before the treatment, and the cancer score of 6 months after the treatment is 150% of the cancer score before the treatment.
  • the cancer scores before and after treatment can be compared with a predetermined threshold value to determine the effectiveness of the treatment. For example, if the cancer score is 200 before the treatment and 130 after the treatment where the threshold cancer score is 100, then the treatment can be determined “effective” as the cancer score drops below the threshold after the treatment. However, if the cancer score is 200 before the treatment and 160 after the treatment where the threshold cancer score is 150, then the treatment can be determined “not effective” as the cancer score stays above the threshold after the treatment even though the absolute value of the cancer score is decreased.
  • the inventors further contemplate that the patient continues with administering the treatment option (e.g., immune therapy, etc.) when the treatment can be determined “effective”, when the cancer score after the treatment is lower than the predetermined threshold, when the cancer score after the treatment is at most 5%, at most 10% higher than the predetermined threshold, or when the cancer score after the treatment is at least 5%, at least 10%, at least 15% lower than the predetermined threshold.
  • the treatment option e.g., immune therapy, etc.
  • the effectives of some cancer treatments can be determined by analyzing omics data including foreign DNA/RNA originated from a carrier of the immune therapy (e.g., virus, bacteria, yeast, etc.).
  • a carrier of the immune therapy e.g., virus, bacteria, yeast, etc.
  • the virus is a carrier to deliver a recombinant nucleic acid encoding recombinant killer activation receptor (KAR)
  • KAR recombinant killer activation receptor

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medical Informatics (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Primary Health Care (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Biomedical Technology (AREA)
  • Data Mining & Analysis (AREA)
  • Chemical & Material Sciences (AREA)
  • Pathology (AREA)
  • Databases & Information Systems (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Theoretical Computer Science (AREA)
  • Spectroscopy & Molecular Physics (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Evolutionary Biology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Surgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Methods for analyzing omics data and using the omics data to determine prognosis of a cancer, to predict an outcome of a treatment, and/or to determine an effectiveness of a treatment are presented. In preferred methods, blood from a patient having a cancer or suspected to have a cancer is obtained and blood omics data for a plurality of cancer-related, inflammation-related, or DNA repair-related genes are obtained. A cancer score can be calculated based on the omics data, which then can be used to provide a cancer prognosis, a therapeutic recommendation, an effectiveness of a treatment.

Description

  • This application is a continuation application of allowed US application having Ser. No. 16/754,088, which was filed Apr. 6, 2020, and which is a 371 application of PCT/US2018/055481, which was filed Oct. 11, 2018, and which claims priority to US provisional application having the Ser. No. 62/571,414, filed Oct. 12, 2017, all of which are incorporated by reference in their entirety herein.
  • FIELD OF THE INVENTION
  • The field of the invention is profiling of omics data as they relate to cancer, especially as it relates to the generation of indicators for cancer prognosis, prediction of treatment outcomes, and/or effectiveness of cancer treatments.
  • BACKGROUND OF THE INVENTION
  • The background description includes information that may be useful in understanding the present invention. It is not an admission that any of the information provided herein is prior art or relevant to the presently claimed invention, or that any publication specifically or implicitly referenced is prior art.
  • All publications and patent applications herein are incorporated by reference to the same extent as if each individual publication or patent application were specifically and individually indicated to be incorporated by reference. Where a definition or use of a term in an incorporated reference is inconsistent or contrary to the definition of that term provided herein, the definition of that term provided herein applies and the definition of that term in the reference does not apply.
  • Cancer is a multifactorial disease where many diverse genetic and environmental factors interplay and contribute to the development and outcome of the disease. In addition, genetic and environmental factors often affect the patient's prognosis in various degrees such that individual patients may show different responses to the same therapeutic and/or prophylactic treatment. Such complexity and diversity render traditional prediction of prognosis, identification of optimal treatments, and prediction of likelihood of success of the treatments based on a single or few factors (e.g., serum level of inflammation-related proteins, etc.), often unreliable. Further, many traditional methods of examining such factors are invasive as they require tumor biopsy samples for histology of tumor cells and tissues.
  • More recently, DNA or RNA populations present in the peripheral blood have drawn attention for analyzing genetic abnormalities associated with the cancer status. For example, U.S. Pat. No. 9,422,592 discloses the measurement of cell free RNA (cfRNA) of formulpeptide receptor gene (FPR1) and its association with the patient's risk for having lung cancer or non-small cell lung cancer (NSCLC). Yet, such studies are limited to a few numbers of genes, which are typically weighed equally in determining the cancer status. As multiple factors affect to various degrees prognosis of most cancers, oversimplification may cause inaccurate prognosis and/or prediction of treatment outcome.
  • Thus, even though some examples of using cell free nucleic acid in determining cancer status are known, differentially weighed, multi-factor approaches in determining cancer status using cell free nucleic acid are largely unexplored. Thus, there remains a need for improved methods of analyzing omics data of cell free nucleic acids in determining status, prognosis of a cancer as well as likelihood of treatment outcome or effectiveness of the treatment.
  • SUMMARY OF THE INVENTION
  • The inventive subject matter is directed to methods of using various omics data of cell free nucleic acids to calculate a composite cancer score that can be used to determine the status, prognosis of a cancer as well as likelihood of treatment outcome and/or effectiveness of current treatments. Thus, one aspect of the subject matter includes a method of analyzing omics data. In this method, blood is obtained from a patient having or suspected to have a cancer. From the blood, omics data for a plurality of cancer-related genes are obtained. Most preferably, the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data. From the omics data, a composite score is calculated which can then be associated with at least one of a health status, an omics error status, a cancer prognosis, a therapeutic recommendation, and an effectiveness of a treatment.
  • In some embodiments, the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status. Optionally, the DNA sequence data is obtained from circulating free DNA. In other embodiments, the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA. Optionally, the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • Typically, the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease. Preferably, the neoepitope is tumor-specific and patient-specific. In some embodiments, the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In such embodiments, it is preferred that the presence of the mutation in the cancer-specific gene weighs more than the presence of the mutation in the cancer-related genes other than the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio between or among a plurality of splice variants of the cancer gene.
  • In some embodiments, the method further comprises a step of comparing the score with a threshold value to thereby determine the therapeutic recommendation. In such embodiments, it is preferred that the therapeutic recommendation is a prophylactic treatment if the score is below the threshold value. Alternatively and/or additionally, the method further comprises a step of comparing the omics error status with a threshold value to thereby determine a risk score.
  • In another aspect of the inventive subject matter, the inventors contemplate a method of determining prognosis of a cancer of a patient. In this method, blood is obtained from a patient having or suspected to have a cancer. From the blood, omics data for a plurality of cancer genes are obtained. Preferably, the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data. From the omics data, a cancer prognosis score is calculated, and the prognosis of the cancer is provided based on the cancer prognosis score. IN some embodiments, the prognosis comprises a progress of metastasis.
  • In some embodiments, the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status. Optionally, the DNA sequence data is obtained from circulating free DNA. In other embodiments, the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA. Optionally, the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • Typically, the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease. Preferably, the neoepitope is tumor-specific and patient-specific. In some embodiments, the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio among or between a plurality of splice variants of the cancer gene.
  • In some embodiments, the omics data is a plurality of sets of omics data obtained at a different time points during a time period, and the prognosis is provided based on a plurality of scores from the plurality of sets of omics data. In such embodiments, it is preferred that the prognosis is represented by a change of a plurality of scores during the time period, wherein the change is over a predetermined threshold value.
  • Still another aspect of inventive subject matter is directed towards a method of predicting an outcome of a treatment for a cancer patient. In this method, blood is obtained from a patient having a cancer. From the blood, omics data for a plurality of cancer genes are obtained. Preferably, the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data. From the omics data, a cancer gene score is calculated, and a predicted outcome of the treatment is provided based on the cancer prognosis score. Preferably, the predicted outcome is determined by comparing the cancer gene score with a predetermined threshold value.
  • In some embodiments, the treatment is a drug, and at least one of the plurality of cancer gene is a predicted target of the drug. In other embodiments, the treatment is an immune therapy, and at least one of the plurality of cancer gene is a receptor of an immune cell or a ligand of the receptor. In still other embodiments, the treatment is a surgery or a radiation therapy, and at least one of the plurality of cancer gene is a neoepitope that is tumor-specific and patient-specific.
  • In some embodiments, the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status. Optionally, the DNA sequence data is obtained from circulating free DNA. In other embodiments, the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA. Optionally, the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • Typically, the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease. Preferably, the neoepitope is tumor-specific and patient-specific. In some embodiments, the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio between a plurality of splice variants of the cancer gene.
  • In still another aspect of the inventive subject matter, the inventors contemplate a method of evaluating an effectiveness of a treatment for a cancer patient. In this method, blood is obtained from a patient having a cancer. From the blood, omics data for a plurality of cancer genes are obtained before and after the treatment. Preferably, the omics data include at least one of DNA sequence data, RNA sequence data, and RNA expression level data. From the omics data, at least two cancer gene scores corresponding to the omics data before and after the treatment, respectively, are generated, and the effectiveness of the treatment is provided based on the comparison of the at least two cancer gene scores. In some embodiments, the effectiveness of the treatment can be determined by a difference between the cancer gene score before and after the treatment. In such embodiments, it is preferred that the treatment is determined effective when the difference is higher than a predetermined threshold value.
  • In some embodiments, the treatment is a drug, and at least one of the plurality of cancer gene is a predicted target of the drug. In other embodiments, the treatment is an immune therapy, and at least one of the plurality of cancer gene is a receptor of an immune cell or a ligand of the receptor. In still other embodiments, the treatment is a surgery or a radiation therapy, and at least one of the plurality of cancer gene is a neoepitope that is tumor-specific and patient-specific.
  • In some embodiments, the DNA sequence data v selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status. Optionally, the DNA sequence data is obtained from circulating free DNA. In other embodiments, the RNA sequence data is selected from the group consisting of mRNA sequence data and splice variant data, and/or the RNA expression level data is selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA. Optionally, the RNA sequence data is obtained from the group consisting of circulating tumor RNA and circulating free RNA.
  • Typically, the plurality of cancer-related genes comprises at least one of a cancer-related gene, a cancer-specific gene, a DNA-repair gene, a neoepitope, and a gene not associated with a disease. Preferably, the neoepitope is tumor-specific and patient-specific. In some embodiments, the plurality of cancer-related genes includes a cancer-specific gene, and the score is calculated based on a presence or an absence of a mutation in the cancer-specific gene. In other embodiments, the score is calculated based on a type of a splice variant of the cancer gene or a ratio between a plurality of splice variants of the cancer gene.
  • Various objects, features, aspects and advantages of the inventive subject matter will become more apparent from the following detailed description of preferred embodiments.
  • DETAILED DESCRIPTION
  • The inventors discovered that the status and/or prognosis of a cancer can be more reliably determined in a less invasive and quick manner using a compound score that is generated based on multiple factors associated with the cancer. The inventors also discovered that the compound score can be used to reliably predict a likelihood of outcome of a cancer treatment, and further, effectiveness of a particular cancer treatment. Viewed from a different perspective, the inventors discovered that a compound score can be generated from the patient's omics data obtained from nucleic acids in the patient's blood. Typically the omics data include omics data of various cancer-related genes, which can be differentially weighed based on the type and timing of the sampling. The compound score can be a reliable indicator to determine cancer status and/or prognosis of a cancer, a likelihood of outcome of a cancer treatment. Further, the compound scores generated based on omics data obtained before and after a cancer treatment can be compared to determine the effectiveness of a cancer treatment.
  • As used herein, the term “tumor” refers to, and is interchangeably used with one or more cancer cells, cancer tissues, malignant tumor cells, or malignant tumor tissue, that can be placed or found in one or more anatomical locations in a human body.
  • It should be noted that the term “patient” as used herein includes both individuals that are diagnosed with a condition (e.g., cancer) as well as individuals undergoing examination and/or testing for the purpose of detecting or identifying a condition. Thus, a patient having a tumor refers to both individuals that are diagnosed with a cancer as well as individuals that are suspected to have a cancer.
  • As used herein, the term “provide” or “providing” refers to and includes any acts of manufacturing, generating, placing, enabling to use, transferring, or making ready to use.
  • Cell-Free DNA/RNA
  • The inventors contemplate that tumor cells and/or some immune cells interacting or surrounding the tumor cells release cell free DNA/RNA to the patient's bodily fluid, and thus may increase the quantity of the specific cell free DNA/RNA in the patient's bodily fluid as compared to a healthy individual. As used herein, the patient's bodily fluid includes, but is not limited to, blood, serum, plasma, mucus, cerebrospinal fluid, ascites fluid, saliva, and urine of the patient. Alternatively, it should be noted that various other bodily fluids are also deemed appropriate so long as cell free DNA/RNA is present in such fluids. The patient's bodily fluid may be fresh or preserved/frozen. Appropriate fluids include saliva, ascites fluid, spinal fluid, urine, etc., which may be fresh or preserved/frozen.
  • The cell free RNA may include any types of DNA/RNA that are circulating in the bodily fluid of a person without being enclosed in a cell body or a nucleus. Most typically, the source of the cell free DNA/RNA is the tumor cells. However, it is also contemplated that the source of the cell free DNA/RNA is an immune cell (e.g., NK cells, T cells, macrophages, etc.). Thus, the cell free DNA/RNA can be circulating tumor DNA/RNA (ctDNA/RNA) and/or circulating free DNA/RNA (cf DNA/RNA, circulating nucleic acids that do not derive from a tumor). While not wishing to be bound by a particular theory, it is contemplated that release of cell free DNA/RNA originating from a tumor cell can be increased when the tumor cell interacts with an immune cell or when the tumor cells undergo cell death (e.g., necrosis, apoptosis, autophagy, etc.). Thus, in some embodiments, the cell free DNA/RNA may be enclosed in a vesicular structure (e.g., via exosomal release of cytoplasmic substances) so that it can be protected from nuclease (e.g., RNAase) activity in some type of bodily fluid. Yet, it is also contemplated that in other aspects, the cell free DNA/RNA is a naked DNA/RNA without being enclosed in any membranous structure, but may be in a stable form by itself or be stabilized via interaction with one or more non-nucleotide molecules (e.g., any RNA binding proteins, etc.).
  • It is contemplated that the cell free DNA/RNA can be any type of DNA/RNA which can be released from either cancer cells or immune cell. Thus, the cell free DNA may include any whole or fragmented genomic DNA, or mitochondrial DNA, and the cell free RNA may include mRNA, tRNA, microRNA, small interfering RNA, long non-coding RNA (lncRNA). Most typically, the cell free DNA is a fragmented DNA typically with a length of at least 50 base pair (bp), 100 base pair (bp), 200 bp, 500 bp, or 1 kbp. Also, it is contemplated that the cell free RNA is a full length or a fragment of mRNA (e.g., at least 70% of full-length, at least 50% of full length, at least 30% of full length, etc.). While cell free DNA/RNA may include any type of DNA/RNA encoding any cellular, extracellular proteins or non-protein elements, it is preferred that at least some of cell free DNA/RNA encodes one or more cancer-related proteins, or inflammation-related proteins. For example, the cell free DNA/mRNA may be full-length or fragments of (or derived from the) cancer related genes including, but not limited to ABL1, ABL2, ACTB, ACVR1B, AKT1, AKT2, AKT3, ALK, AMER11, APC, AR, ARAF, ARFRP1, ARID1A, ARID1B, ASXL1, ATF1, ATM, ATR, ATRX, AURKA, AURKB, AXIN1, AXL, BAP1, BARD1, BCL2, BCL2L1, BCL2L2, BCL6, BCOR, BCORL1, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRD4, BRIP1, BTG1, BTK, EMSY, CARD11, CBFB, CBL, CCND1, CCND2, CCND3, CCNE1, CD274, CD79A, CD79B, CDC73, CDH1, CDK12, CDK4, CDK6, CDK8, CDKN1A, CDKN1B, CDKN2A, CDKN2B, CDKN2C, CEA, CEBPA, CHD2, CHD4, CHEK1, CHEK2, CIC, CREBBP, CRKL, CRLF2, CSF1R, CTCF, CTLA4, CTNNA1, CTNNB1, CUL3, CYLD, DAXX, DDR2, DEPTOR, DICER1, DNMT3A, DOT1L, EGFR, EP300, EPCAM, EPHA3, EPHA5, EPHA7, EPHB1, ERBB2, ERBB3, ERBB4, EREG, ERG, ERRFI1, ESR1, EWSR1, EZH2, FAM46C, FANCA, FANCC, FANCD2, FANCE, FANCF, FANCG, FANCL, FAS, FAT1, FBXW7, FGF10, FGF14, FGF19, FGF23, FGF3, FGF4, FGF6, FGFR1, FGFR2, FGFR3, FGFR4, FH, FLCN, FLI1, FLT1, FLT3, FLT4, FOLH1, FOXL2, FOXP1, FRS2, FUBP1, GABRA6, GATA1, GATA2, GATA3, GATA4, GATA6, GID4, GLI1, GNA11, GNA13, GNAQ, GNAS, GPR124, GRIN2A, GRM3, GSK3B, H3F3A, HAVCR2, HGF, HMGB1, HMGB2, HMGB3, HNF1A, HRAS, HSD3B1, HSP90AA1, IDH1, IDH2, IDO, IGF1R, IGF2, IKBKE, IKZF1, IL7R, INHBA, INPP4B, IRF2, IRF4, IRS2, JAK1, JAK2, JAK3, JUN, MYST3, KDM5A, KDM5C, KDM6A, KDR, KEAP, KEL, KIT, KLHL6, KLK3, MLL, MLL2, MLL3, KRAS, LAG3, LMO1, LRP1B, LYN, LZTR1, MAGI2, MAP2K1, MAP2K2, MAP2K4, MAP3K1, MCL1, MDM2, MDM4, MED12, MEF2B, MEN1, MET, MITF, MLH1, MPL, MRE11A, MSH2, MSH6, MTOR, MUC1, MUTYH, MYC, MYCL, MYCN, MYD88, MYH, NF1, NF2, NFE2L2, NFKB1A, NKX2-1, NOTCH1, NOTCH2, NOTCH3, NPM1, NRAS, NSD1, NTRK1, NTRK2, NTRK3, NUP93, PAK3, PALB2, PARK2, PAX3, PAX, PBRM1, PDGFRA, PDCD1, PDCD1LG2, PDGFRB, PDK1, PGR, PIK3C2B, PIK3CA, PIK3CB, PIK3CG, PIK3R1, PIK3R2, PLCG2, PMS2, POLD1, POLE, PPP2R1A, PREX2, PRKAR1A, PRKC1, PRKDC, PRSS8, PTCH1, PTEN, PTPN11, QK1, RAC1, RAD50, RAD51, RAF1, RANBP1, RARA, RB1, RBM10, RET, RICTOR, RIT1, RNF43, ROS1, RPTOR, RUNX1, RUNX1T1, SDHA, SDHB, SDHC, SDHD, SETD2, SF3B1, SLIT2, SMAD2, SMAD3, SMAD4, SMARCA4, SMARCB1, SMO, SNCAIP, SOCS1, SOX10, SOX2, SOX9, SPEN, SPOP, SPTA1, SRC, STAG2, STAT3, STAT4, STK11, SUFU, SYK, T (BRACHYURY), TAF1, TBX3, TERC, TERT, TET2, TGFRB2, TNFAIP3, TNFRSF14, TOP1, TOP2A, TP53, TSC1, TSC2, TSHR, U2AF1, VEGFA, VHL, WISP3, WT1, XPO1, ZBTB2, ZNF217, ZNF703, CD26, CD49F, CD44, CD49F, CD13, CD15, CD29, CD151, CD138, CD166, CD133, CD45, CD90, CD24, CD44, CD38, CD47, CD96, CD 45, CD90, ABCB5, ABCG2, ALCAM, ALPHA-FETOPROTEIN, DLL1, DLL3, DLL4, ENDOGLIN, GJA1, OVASTACIN, AMACR, NESTIN, STRO-1, MICL, ALDH, BMI-1, GLI-2, CXCR1, CXCR2, CX3CR1, CX3CL1, CXCR4, PON1, TROP1, LGR5, MSI-1, C-MAF, TNFRSF7, TNFRSF16, SOX2, PODOPLANIN, L1CAM, HIF-2 ALPHA, TFRC, ERCC1, TUBB3, TOP1, TOP2A, TOP2B, ENOX2, TYMP, TYMS, FOLR1, GPNMB, PAPPA, GART, EBNA1, EBNA2, LMP1, BAGE, BAGE2, BCMA, C10ORF54, CD4, CD8, CD19, CD20, CD25, CD30, CD33, CD80, CD86, CD123, CD276, CCL1, CCL2, CCL3, CCL4, CCL5, CCL7, CCL8, CCL11, CCL13, CCL14, CCL15, CCL16, CCL17, CCL18, CCL19, CCL20, CCL21, CCL22, CCL23, CCL24, CCL25, CCL26, CCL27, CCL28, CCR1, CCR2, CCR3, CCR4, CCR5, CCR6, CCR7, CCR8, CCR9, CCR10, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL9, CXCL10, CXCL11, CXCL12, CXCL13, CXCL14, CXCL16, CXCL17, CXCR3, CXCR5, CXCR6, CTAG1B, CTAG2, CTAG1, CTAG4, CTAG5, CTAG6, CTAG9, CAGE1, GAGE1, GAGE2A, GAGE2B, GAGE2C, GAGE2D, GAGE2E, GAGE4, GAGE10, GAGE12D, GAGE12F, GAGE12J, GAGE13, HHLA2, ICOSLG, LAG1, MAGEA10, MAGEA12, MAGEA1, MAGEA2, MAGEA3, MAGEA4, MAGEA4, MAGEA5, MAGEA6, MAGEA7, MAGEA8, MAGEA9, MAGEB1, MAGEB2, MAGEB3, MAGEB4, MAGEB6, MAGEB10, MAGEB16, MAGEB18, MAGEC1, MAGEC2, MAGEC3, MAGED1, MAGED2, MAGED4, MAGED4B, MAGEE1, MAGEE2, MAGEF1, MAGEH1, MAGEL2, NCR3LG1, SLAMF7, SPAG1, SPAG4, SPAG5, SPAG6, SPAG7, SPAG8, SPAG9, SPAG11A, SPAG11B, SPAG16, SPAG17, VTCN1, XAGE1D, XAGE2, XAGE3, XAGE5, XCL1, XCL2, and XCR1. Of course, it should be appreciated that the above genes may be wild type or mutated versions, including missense or nonsense mutations, insertions, deletions, fusions, and/or translocations, all of which may or may not cause formation of full-length mRNA when transcribed.
  • For another example, some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of inflammation-related proteins, including, but not limited to, HMGB1, HMGB2, HMGB3, MUC1, VWF, MMP, CRP, PBEF1, TNF-α, TGF-β, PDGFA, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, Eotaxin, FGF, G-CSF, GM-CSF, IFN-7, IP-10, MCP-1, PDGF, and hTERT, and in yet another example, the cell free mRNA encoded a full length or a fragment of HMGB1.
  • For still another example, some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of DNA repair-related proteins or RNA repair-related proteins. Table 1 provides an exemplary collection of predominant RNA repair genes and their associated repair pathways contemplated herein, but it should be recognized that numerous other genes associated with DNA repair and repair pathways are also expressly contemplated herein, and Tables 2 and 3 illustrate further exemplary genes for analysis and their associated function in DNA repair.
  • TABLE 1
    Repair mechanism Predominant DNA Repair genes
    Base excision DNA glycosylase, APE1, XRCC1, PNKP, Tdp1,
    repair (BER) APTX, DNA polymerase β, FEN1, DNA
    polymerase δ or ε, PCNA-RFC, PARP
    Mismatch repair MutSα (MSH2-MSH6), Mutsβ (MSH2-MSH3),
    (MMR) MutLα (MLH1-PMS2), MutLβ (MLH1-PMS2),
    MutLγ (MLH1-MLH3), Exo1, PCNA-RFC
    Nucleotide XPC-Rad23B-CEN2, UV-DDB (DDB1-XPE), CSA,
    excision CSB, TFIIH, XPB, XPD, XPA, RPA, XPG,
    repair (NER) ERCC1- XPF, DNA polymerase δ or ε
    Homologous Mre11-Rad50-Nbs1, CtIP, RPA, Rad51, Rad52,
    recombination BRCA1, BRCA2, Exo1, BLM-TopIIIα,
    (HR) GEN1-Yen1, Slx1-Slx4, Mus81/Eme1
    Non-homologous Ku70-Ku80, DNA-PKc, XRCC4-DNA ligase IV,
    end-joining XLF
    (NHEJ)
  • TABLE 2
    Accession
    Gene name (synonyms) Activity number
    Base excision repair (BER)
    DNA glycosylases: major altered base
    released
    UNG U excision NM_003362
    SMUG1 U excision NM_014311
    MBD4 U or T opposite G at CpG sequences NM_003925
    TDG U, T or ethenoC opposite G NM_003211
    OGG1 8-oxoG opposite C NM_002542
    MYH A opposite 8-oxoG NM_012222
    NTH1 Ring-saturated or fragmented NM_002528
    pyrimidines
    MPG 3-meA, ethenoA, hypoxanthine NM_002434
    Other BER factors
    APE1 (HAP1, APEX, REF1) AP endonuclease NM_001641
    APE2 (APEXL2) AP endonuclease NM_014481
    LIG3 Main ligation function NM_013975
    XRCC1 Main ligation function NM_006297
    Poly(ADP-ribose) polymerase (PARP)
    enzymes
    ADPRT Protects strand interruptions NM_001618
    ADPRTL2 PARP-like enzyme NM_005485
    ADPRTL3 PARP-like enzyme AF085734
    Direct reversal of damage
    MGMT O6-meG alkyltransferase NM_002412
    Mismatch excision repair
    (MMR)
    MSH2 Mismatch and loop recognition NM_000251
    MSH3 Mismatch and loop recognition NM_002439
    MSH6 Mismatch recognition NM_000179
    MSH4 MutS homolog specialized for meiosis NM_002440
    MSH5 MutS homolog specialized for meiosis NM_002441
    PMS1 Mitochondrial MutL homolog NM_000534
    MLH1 MutL homolog NM_000249
    PMS2 MutL homolog NM_000535
    MLH3 MutL homolog of unknown function NM_014381
    PMS2L3 MutL homolog of unknown function D38437
    PMS2L4 MutL homolog of unknown function D38438
    Nucleotide excision repair
    (NER)
    XPC Binds damaged DNA as complex NM_004628
    RAD23B (HR23B) Binds damaged DNA as complex NM_002874
    CETN2 Binds damaged DNA as complex NM_004344
    RAD23A (HR23A) Substitutes for HR23B NM_005053
    χPA Binds damaged DNA in preincisioncomplex NM_000380
    RPA1 Binds DNA in preincision complex NM_002945
    RPA2 Binds DNA in preincision complex NM_002946
    RPA3 Binds DNA in preincision complex NM_002947
    TFIIH Catalyzes unwinding in preincisioncomplex
    XPB (ERCC3) 3′ to 5′ DNA helicase NM_000122
    XPD (ERCC2) 5′ to 3′ DNA helicase X52221
    GTF2H1 Core TFIIH subunit p62 NM_005316
    GTF2H2 Core TFIIH subunit p44 NM_001515
    GTF2H3 Core TFIIH subunit p34 NM_001516
    GTF2H4 Core TFIIH subunit p52 NM_001517
    CDK7 Kinase subunit of TFIIH NM_001799
    CCNH Kinase subunit of TFIIH NM_001239
    MNAT1 Kinase subunit of TFIIH NM_002431
    XPG (ERCC5) 3′ incision NM_000123
    ERCC1 5′ incision subunit NM_001983
    XPF (ERCC4) 5′ incision subunit NM_005236
    LIG1 DNA joining NM_000234
    NER-related
    CSA (CKN1) Cockayne syndrome; needed for NM_000082
    transcription-coupled NER
    CSB (ERCC6) Cockayne syndrome; needed for NM_000124
    transcription-coupled NER
    XAB2 (HCNP) Cockayne syndrome; needed for NM_020196
    transcription-coupled NER
    DDB1 Complex defective in XP group E NM_001923
    DDB2 Mutated in XP group E NM_000107
    MMS19 Transcription and NER AW852889
    Homologous recombination
    RAD51 Homologous pairing NM_002875
    RAD51L1 (RAD51B) Rad51 homolog U84138
    RAD51C Rad51 homolog NM_002876
    RAD51L3 (RAD51D) Rad51 homolog NM_002878
    DMC1 Rad51 homolog, meiosis NM_007068
    XRCC2 DNA break and cross-link repair NM_005431
    XRCC3 DNA break and cross-link repair NM_005432
    RAD52 Accessory factor for recombination NM_002879
    RAD54L Accessory factor for recombination NM_003579
    RAD54B Accessory factor for recombination NM_012415
    BRCA1 Accessory factor for transcription NM_007295
    and recombination
    BRCA2 Cooperation with RAD51, essential NM_000059
    function
    RAD50 ATPase in complex with MRE11A, NBS1 NM_005732
    MRE11A 3′ exonuclease NM_005590
    NBS1 Mutated in Nijmegen breakage syndrome NM_002485
    Nonhomologous end-joining
    Ku70 (G22P1) DNA end binding NM_001469
    Ku80 (XRCC5) DNA end binding M30938
    PRKDC DNA-dependent protein kinase NM_006904
    catalytic subunit
    LIG4 Nonhomologous end-joining NM_002312
    XRCC4 Nonhomologous end-joining NM_003401
    Sanitization of nucleotide pools
    MTH1 (NUDT1) 8-oxoGTPase NM_002452
    DUT dUTPase NM_001948
    DNA polymerases (catalytic subunits)
    POLB BER in nuclear DNA NM_002690
    POLG BER in mitochondrial DNA NM_002693
    POLD1 NER and MMR NM_002691
    POLE1 NER and MMR NM_006231
    PCNA Sliding clamp for pol delta and pol NM_002592
    epsilon
    REV3L (POLZ) DNA pol zeta catalytic subunit, NM_002912
    essential function
    REV7 (MAD2L2) DNA pol zeta subunit NM_006341
    REV1 dCMP transferase NM_016316
    POLH XP variant NM_006502
    POLI (RAD30B) Lesion bypass NM_007195
    POLQ DNA cross-link repair NM_006596
    DINB1 (POLK) Lesion bypass NM_016218
    POLL Meiotic function NM_013274
    POLM Presumed specialized lymphoid NM_013284
    function
    TRF4-1 Sister-chromatid cohesion AF089896
    TRF4-2 Sister-chromatid cohesion AF089897
    Editing and processing nucleases
    FEN1 (DNase IV) 5′ nuclease NM_004111
    TREX1 (DNase III) 3′ exonuclease NM_007248
    TREX2 3′ exonuclease NM_007205
    EX01 (HEX1) 5′ exonuclease NM_003686
    SPO11 endonuclease NM_012444
    Rad6 pathway
    UBE2A (RAD6A) Ubiquitin-conjugating enzyme NM_003336
    UBE2B (RAD6B) Ubiquitin-conjugating enzyme NM_003337
    RAD18 Assists repair or replication of damaged AB035274
    DNA
    UBE2VE (MMS2) Ubiquitin-conjugating complex AF049140
    UBE2N (UBC13, BTG1) Ubiquitin-conjugating complex NM_003348
    Genes defective in diseases
    associated with sensitivity to
    DNA damaging agents
    BLM Bloom syndrome helicase NM_000057
    WRN Werner syndrome helicase/3′- NM_000553
    exonuclease
    RECQL4 Rothmund-Thompson syndrome NM_004260
    ATM Ataxia telangiectasia NM_000051
    Fanconi anemia
    FANCA Involved in tolerance or repair of DNA NM_000135
    cross-links
    FANCB Involved in tolerance or repair of DNA N/A
    cross-links
    FANCC Involved in tolerance or repair of DNA NM_000136
    cross-links
    FANCD Involved in tolerance or repair of DNA N/A
    cross-links
    FANCE Involved in tolerance or repair of DNA NM_021922
    cross-links
    FANCF Involved in tolerance or repair of DNA AF181994
    cross-links
    FANCG (XRCC9) Involved in tolerance or repair of DNA NM_004629
    cross-links
    Other identified genes with a
    suspected DNA repair function
    SNM1 (PS02) DNA cross-link repair D42045
    SNM1B Related to SNM1 AL137856
    SNM1C Related to SNM1 AA315885
    RPA4 Similar to RPA2 NM_013347
    ABH (ALKB) Resistance to alkylation damage X91992
    PNKP Converts some DNA breaks to ligatable NM_007254
    ends
    Other conserved DNA
    damage response genes
    ATR ATM- and PI-3K-like essential kinase NM_001184
    RAD1 (S. pombe) homolog PCNA-like DNA damage sensor NM_002853
    RAD9 (S. pombe) homolog PCNA-like DNA damage sensor NM_004584
    HUS1 (S. pombe) homolog PCNA-like DNA damage sensor NM_004507
    RAD17 (RAD24) RFC-like DNA damage sensor NM_002873
    TP53BP1 BRCT protein NM_005657
    CHEK1 Effector kinase NM_001274
    CHK2 (Rad53) Effector kinase NM_007194
  • TABLE 3
    Gene Name Gene Title Biological Activity
    RFC2 replication factor C (activator 1) 2, DNA replication
    40 kDa
    XRCC6 X-ray repair complementing DNA ligation /// DNA repair /// double-strand break
    defective repair in Chinese hamster repair via nonhomologous end-joining /// DNA
    cells 6 (Ku autoantigen, 70 kDa) recombination /// positive regulation of
    transcription, DNA-dependent /// double-strand
    break repair via nonhomologous end-joining ///
    response to DNA damage stimulus /// DNA recombination
    APOBEC apolipoprotein B mRNA editing For all of APOBEC1, APOBEC2, APOBEC3A-H,
    enzyme, catalytic polypeptide-like and APOBEC4, cytidine deaminases.
    POLD2 polymerase (DNA directed), delta 2, DNA replication /// DNA replication
    regulatory subunit 50 kDa
    PCNA proliferating cell nuclear antigen regulation of progression through cell cycle /// DNA
    replication /// regulation of DNA replication ///
    DNA repair /// cell proliferation ///
    phosphoinositide-mediated signaling /// DNA replication
    RPA1 replication protein A1, 70 kDa DNA-dependent DNA replication /// DNA repair ///
    DNA recombination /// DNA replication
    RPA1 replication protein A1, 70 kDa DNA-dependent DNA replication /// DNA repair ///
    DNA recombination /// DNA replication
    RPA2 replication protein A2, 32 kDa DNA replication /// DNA-dependent DNA replication
    ERCC3 excision repair cross-complementing DNA topological change /// transcription-coupled
    rodent repair deficiency, nucleotide-excision repair /// transcription ///
    complementation group 3 (xeroderma regulation of transcription, DNA-dependent ///
    pigmentosum group B transcription from RNA polymerase II promoter ///
    complementing) induction of apoptosis /// sensory perception of
    sound /// DNA repair /// nucleotide-excision repair ///
    response to DNA damage stimulus /// DNA repair
    UNG uracil-DNA glycosylase carbohydrate metabolism /// DNA repair ///
    base-excision repair /// response to DNA damage
    stimulus /// DNA repair /// DNA repair
    ERCC5 excision repair cross-complementing transcription-coupled nucleotide-excision repair ///
    rodent repair deficiency, nucleotide-excision repair /// sensory perception of
    complementation group 5 (xeroderma sound /// DNA repair /// response to DNA damage
    pigmentosum, complementation stimulus /// nucleotide-excision repair
    group G (Cockayne syndrome))
    MLH1 mutL homolog 1, colon cancer, mismatch repair /// cell cycle /// negative regulation
    nonpolyposis type 2 (E. coli) of progression through cell cycle /// DNA repair ///
    mismatch repair /// response to DNA damage stimulus
    LIG1 ligase I, DNA, ATP-dependent DNA replication /// DNA repair /// DNA
    recombination /// cell cycle /// morphogenesis ///
    cell division /// DNA repair /// response to DNA
    damage stimulus /// DNA metabolism
    NBN nibrin DNA damage checkpoint /// cell cycle checkpoint ///
    double-strand break repair
    NBN nibrin DNA damage checkpoint /// cell cycle checkpoint ///
    double-strand break repair
    NBN nibrin DNA damage checkpoint /// cell cycle checkpoint ///
    double-strand break repair
    MSH6 mutS homolog 6 (E. coli) mismatch repair /// DNA metabolism /// DNA repair ///
    mismatch repair /// response to DNA damage stimulus
    POLD4 polymerase (DNA-directed), delta 4 DNA replication /// DNA replication
    RFC5 replication factor C (activator 1) 5, DNA replication /// DNA repair /// DNA replication
    36.5 kDa
    RFC5 replication factor C (activator 1) 5, DNA replication /// DNA repair /// DNA replication
    36.5 kDa
    DDB2 /// damage-specific DNA binding nucleotide-excision repair /// regulation of
    LHX3 protein 2, 48 kDa /// LIM homeobox 3 transcription, DNA-dependent /// organ
    morphogenesis /// DNA repair /// response to DNA
    damage stimulus /// DNA repair /// transcription ///
    regulation of transcription
    POLD1 polymerase (DNA directed), delta 1, DNA replication /// DNA repair /// response to UV ///
    catalytic subunit 125 kDa DNA replication
    FANCG Fanconi anemia, complementation cell cycle checkpoint /// DNA repair /// DNA repair ///
    group G response to DNA damage stimulus /// regulation
    of progression through cell cycle
    POLB polymerase (DNA directed), beta DNA-dependent DNA replication /// DNA repair ///
    DNA replication /// DNA repair /// response to DNA
    damage stimulus
    XRCC1 X-ray repair complementing single strand break repair
    defective repair in Chinese hamster
    cells 1
    MPG N-methylpurine-DNA glycosylase base-excision repair /// DNA dealkylation /// DNA
    repair /// base-excision repair /// response to DNA
    damage stimulus
    RFC2 replication factor C (activator 1) 2, DNA replication
    40 kDa
    ERCC1 excision repair cross-complementing nucleotide-excision repair /// morphogenesis ///
    rodent repair deficiency, nucleotide-excision repair /// DNA repair ///
    complementation group 1 (includes response to DNA damage stimulus
    overlapping antisense sequence)
    TDG thymine-DNA glycosylase carbohydrate metabolism /// base-excision repair ///
    DNA repair /// response to DNA damage stimulus
    TDG thymine-DNA glycosylase carbohydrate metabolism /// base-excision repair ///
    DNA repair /// response to DNA damage stimulus
    FANCA Fanconi anemia, complementation DNA repair /// protein complex assembly /// DNA
    group A /// Fanconi anemia, repair /// response to DNA damage stimulus
    complementation group A
    RFC4 replication factor C (activator 1) 4, DNA replication /// DNA strand elongation /// DNA
    37 kDa repair /// phosphoinositide-mediated signaling ///
    DNA replication
    RFC3 replication factor C (activator 1) 3, DNA replication /// DNA strand elongation
    38 kDa
    RFC3 replication factor C (activator 1) 3, DNA replication /// DNA strand elongation
    38 kDa
    APEX2 APEX nuclease DNA repair /// response to DNA damage stimulus
    (apurinic/apyrimidinic endonuclease) 2
    RAD1 RAD1 homolog (S. pombe) DNA repair /// cell cycle checkpoint /// cell cycle
    checkpoint /// DNA damage checkpoint /// DNA
    repair /// response to DNA damage stimulus ///
    meiotic prophase I
    RAD1 RAD1 homolog (S. pombe) DNA repair /// cell cycle checkpoint /// cell cycle
    checkpoint /// DNA damage checkpoint /// DNA
    repair /// response to DNA damage stimulus ///
    meiotic prophase I
    BRCA1 breast cancer 1, early onset regulation of transcription from RNA polymerase II
    promoter /// regulation of transcription from RNA
    polymerase III promoter /// DNA damage response,
    signal transduction by p53 class mediator resulting
    in transcription of p21 class mediator /// cell cycle ///
    protein ubiquitination /// androgen receptor
    signaling pathway /// regulation of cell proliferation ///
    regulation of apoptosis /// positive regulation of
    DNA repair /// negative regulation of progression
    through cell cycle /// positive regulation of
    transcription, DNA-dependent /// negative
    regulation of centriole replication /// DNA damage
    response, signal transduction resulting in induction
    of apoptosis /// DNA repair /// response to DNA
    damage stimulus /// protein ubiquitination /// DNA
    repair /// regulation of DNA repair /// apoptosis ///
    response to DNA damage stimulus
    EXO1 exonuclease 1 DNA repair /// DNA repair /// mismatch repair ///
    DNA recombination
    FEN1 flap structure-specific endonuclease 1 DNA replication /// double-strand break repair ///
    UV protection /// phosphoinositide-mediated
    signaling /// DNA repair /// DNA replication ///
    DNA repair /// DNA repair
    FEN1 flap structure-specific endonuclease 1 DNA replication /// double-strand break repair ///
    UV protection /// phosphoinositide-mediated
    signaling /// DNA repair /// DNA replication ///
    DNA repair /// DNA repair
    MLH3 mutL homolog 3 (E. coli) mismatch repair /// meiotic recombination /// DNA
    repair /// mismatch repair /// response to DNA
    damage stimulus /// mismatch repair
    MGMT O-6-methylguanine-DNA DNA ligation /// DNA repair /// response to DNA
    methyltransferase damage stimulus
    RAD51 RAD51 homolog (RecA homolog, double-strand break repair via homologous
    E. coli) (S. cerevisiae) recombination /// DNA unwinding during
    replication /// DNA repair /// mitotic recombination ///
    meiosis /// meiotic recombination /// positive
    regulation of DNA ligation /// protein
    homooligomerization /// response to DNA damage
    stimulus /// DNA metabolism /// DNA repair ///
    response to DNA damage stimulus /// DNA repair ///
    DNA recombination /// meiotic recombination ///
    double-strand break repair via homologous
    recombination /// DNA unwinding during replication
    RAD51 RAD51 homolog (RecA homolog, double-strand break repair via homologous
    E. coli) (S. cerevisiae) recombination /// DNA unwinding during
    replication /// DNA repair /// mitotic recombination ///
    meiosis /// meiotic recombination /// positive
    regulation of DNA ligation /// protein
    homooligomerization /// response to DNA damage
    stimulus /// DNA metabolism /// DNA repair ///
    response to DNA damage stimulus /// DNA repair ///
    DNA recombination /// meiotic recombination ///
    double-strand break repair via homologous
    recombination /// DNA unwinding during replication
    XRCC4 X-ray repair complementing DNA repair /// double-strand break repair /// DNA
    defective repair in Chinese hamster recombination /// DNA recombination /// response
    cells 4 to DNA damage stimulus
    XRCC4 X-ray repair complementing DNA repair /// double-strand break repair /// DNA
    defective repair in Chinese hamster recombination /// DNA recombination /// response
    cells 4 to DNA damage stimulus
    RECQL RecQ protein-like (DNA helicase DNA repair /// DNA metabolism
    Q1-like)
    ERCC8 excision repair cross-complementing DNA repair /// transcription /// regulation of
    rodent repair deficiency, transcription, DNA-dependent /// sensory perception
    complementation group 8 of sound /// transcription-coupled nucleotide-
    excision repair
    FANCC Fanconi anemia, complementation DNA repair /// DNA repair /// protein complex
    group C assembly /// response to DNA damage stimulus
    OGG1 8-oxoguanine DNA glycosylase carbohydrate metabolism /// base-excision repair /// DNA
    repair /// base-excision repair /// response to DNA damage
    stimulus /// DNA repair
    MRE11A MRE11 meiotic recombination 11 regulation of mitotic recombination /// double-
    homolog A (S. cerevisiae) strand break repair via nonhomologous end-joining ///
    telomerase-dependent telomere maintenance ///
    meiosis /// meiotic recombination /// DNA
    metabolism /// DNA repair /// double-strand break
    repair /// response to DNA damage stimulus ///
    DNA repair /// double-strand break repair /// DNA
    recombination
    RAD52 RAD52 homolog (S. cerevisiae) double-strand break repair /// mitotic recombination ///
    meiotic recombination /// DNA repair /// DNA
    recombination /// response to DNA damage stimulus
    WRN Werner syndrome DNA metabolism /// aging
    XPA xeroderma pigmentosum, nucleotide-excision repair /// DNA repair ///
    complementation group A response to DNA damage stimulus /// DNA repair ///
    nucleotide-excision repair
    BLM Bloom syndrome DNA replication /// DNA repair /// DNA
    recombination /// antimicrobial humoral response
    (sensu Vertebrata) /// DNA metabolism /// DNA
    replication
    OGG1 8-oxoguanine DNA glycosylase carbohydrate metabolism /// base-excision repair ///
    DNA repair /// base-excision repair /// response to
    DNA damage stimulus /// DNA repair
    MSH3 mutS homolog 3 (E. coli) mismatch repair /// DNA metabolism /// DNA repair ///
    mismatch repair /// response to DNA damage stimulus
    POLE2 polymerase (DNA directed), epsilon DNA replication /// DNA repair /// DNA replication
    2 (p59 subunit)
    RAD51C RAD51 homolog C (S. cerevisiae) DNA repair /// DNA recombination /// DNA
    metabolism /// DNA repair /// DNA recombination ///
    response to DNA damage stimulus
    LIG4 ligase IV, DNA, ATP-dependent single strand break repair /// DNA replication ///
    DNA recombination /// cell cycle /// cell division ///
    DNA repair /// response to DNA damage stimulus
    ERCC6 excision repair cross-complementing DNA repair /// transcription /// regulation of
    rodent repair deficiency, transcription, DNA-dependent /// transcription from
    complementation group 6 RNA polymerase II promoter /// sensory perception
    of sound
    LIG3 ligase III, DNA, ATP-dependent DNA replication /// DNA repair /// cell cycle ///
    meiotic recombination /// spermatogenesis /// cell
    division /// DNA repair /// DNA recombination ///
    response to DNA damage stimulus
    RAD17 RAD17 homolog (S. pombe) DNA replication /// DNA repair /// cell cycle ///
    response to DNA damage stimulus
    XRCC2 X-ray repair complementing DNA repair /// DNA recombination /// meiosis ///
    defective repair in Chinese hamster DNA metabolism /// DNA repair /// response to
    cells 2 DNA damage stimulus
    MUTYH mutY homolog (E. coli) carbohydrate metabolism /// base-excision repair ///
    mismatch repair /// cell cycle /// negative regulation
    of progression through cell cycle /// DNA repair ///
    response to DNA damage stimulus /// DNA repair
    RFC1 replication factor C (activator 1) 1, DNA-dependent DNA replication /// transcription ///
    145 kDa /// replication factor C regulation of transcription, DNA-dependent ///
    (activator 1) 1, 145 kDa telomerase-dependent telomere maintenance ///
    DNA replication /// DNA repair
    RFC1 replication factor C (activator 1) 1, DNA-dependent DNA replication /// transcription ///
    145 kDa regulation of transcription, DNA-dependent ///
    telomerase-dependent telomere maintenance ///
    DNA replication /// DNA repair
    BRCA2 breast cancer 2, early onset regulation of progression through cell cycle ///
    double-strand break repair via homologous
    recombination /// DNA repair /// establishment
    and/or maintenance of chromatin architecture ///
    chromatin remodeling /// regulation of S phase of
    mitotic cell cycle /// mitotic checkpoint ///
    regulation of transcription /// response to DNA
    damage stimulus
    RAD50 RAD50 homolog (S. cerevisiae) regulation of mitotic recombination /// double-
    strand break repair /// telomerase-dependent
    telomere maintenance /// cell cycle /// meiosis ///
    meiotic recombination /// chromosome organization
    and biogenesis /// telomere maintenance /// DNA
    repair /// response to DNA damage stimulus ///
    DNA repair /// DNA recombination
    DDB1 damage-specific DNA binding nucleotide-excision repair /// ubiquitin cycle ///
    protein 1, 127 kDa DNA repair /// response to DNA damage stimulus ///
    DNA repair
    XRCC5 X-ray repair complementing double-strand break repair via nonhomologous end-
    defective repair in Chinese hamster joining /// DNA recombination /// DNA repair ///
    cells 5 (double-strand-break DNA recombination /// response to DNA damage
    rejoining; Ku autoantigen, 80 kDa) stimulus /// double-strand break repair
    XRCC5 X-ray repair complementing double-strand break repair via nonhomologous end-
    defective repair in Chinese hamster joining /// DNA recombination /// DNA repair ///
    cells 5 (double-strand-break DNA recombination /// response to DNA damage
    rejoining; Ku autoantigen, 80 kDa) stimulus /// double-strand break repair
    PARP1 poly (ADP-ribose) polymerase DNA repair /// transcription from RNA polymerase
    family, member 1 II promoter /// protein amino acid ADP-ribosylation ///
    DNA metabolism /// DNA repair /// protein amino acid
    ADP-ribosylation /// response to DNA damage stimulus
    POLE3 polymerase (DNA directed), epsilon DNA replication
    3 (p17 subunit)
    RFC1 replication factor C (activator 1) 1, DNA-dependent DNA replication /// transcription ///
    145 kDa regulation of transcription, DNA-dependent ///
    telomerase-dependent telomere maintenance ///
    DNA replication /// DNA repair
    RAD50 RAD50 homolog (S. cerevisiae) regulation of mitotic recombination /// double-
    strand break repair /// telomerase-dependent
    telomere maintenance /// cell cycle /// meiosis ///
    meiotic recombination /// chromosome organization
    and biogenesis /// telomere maintenance /// DNA
    repair /// response to DNA damage stimulus ///
    DNA repair /// DNA recombination
    XPC xeroderma pigmentosum, nucleotide-excision repair /// DNA repair ///
    complementation group C nucleotide-excision repair /// response to DNA
    damage stimulus /// DNA repair
    MSH2 mutS homolog 2, colon cancer, mismatch repair /// postreplication repair /// cell
    nonpolyposis type 1 (E. coli) cycle /// negative regulation of progression through
    cell cycle /// DNA metabolism /// DNA repair ///
    mismatch repair /// response to DNA damage
    stimulus /// DNA repair
    RPA3 replication protein A3, 14 kDa DNA replication /// DNA repair /// DNA replication
    MBD4 methyl-CpG binding domain protein 4 base-excision repair /// DNA repair /// response to
    DNA damage stimulus /// DNA repair
    MBD4 methyl-CpG binding domain protein 4 base-excision repair /// DNA repair /// response to
    DNA damage stimulus /// DNA repair
    NTHL1 nth endonuclease III-like 1 (E. coli) carbohydrate metabolism /// base-excision repair ///
    nucleotide-excision repair, DNA incision, 5′-to
    lesion /// DNA repair /// response to DNA damage
    stimulus
    PMS2 /// PMS2 postmeiotic segregation mismatch repair /// cell cycle /// negative regulation
    PMS2CL increased 2 (S. cerevisiae) /// of progression through cell cycle /// DNA repair ///
    PMS2-C terminal-like mismatch repair /// response to DNA damage
    stimulus /// mismatch repair
    RAD51C RAD51 homolog C (S. cerevisiae) DNA repair /// DNA recombination /// DNA
    metabolism /// DNA repair /// DNA recombination ///
    response to DNA damage stimulus
    UNG2 uracil-DNA glycosylase 2 regulation of progression through cell cycle ///
    carbohydrate metabolism /// base-excision repair ///
    DNA repair /// response to DNA damage stimulus
    APEX1 APEX nuclease (multifunctional base-excision repair /// transcription from RNA
    DNA repair enzyme) 1 polymerase II promoter /// regulation of DNA
    binding /// DNA repair /// response to DNA damage
    stimulus
    ERCC4 excision repair cross-complementing nucleotide-excision repair /// nucleotide-excision
    rodent repair deficiency, repair /// DNA metabolism /// DNA repair ///
    complementation group 4 response to DNA damage stimulus
    RAD1 RAD1 homolog (S. pombe) DNA repair /// cell cycle checkpoint /// cell cycle
    checkpoint /// DNA damage checkpoint /// DNA
    repair /// response to DNA damage stimulus ///
    meiotic prophase I
    RECQL5 RecQ protein-like 5 DNA repair /// DNA metabolism /// DNA
    metabolism
    MSH5 mutS homolog 5 (E. coli) DNA metabolism /// mismatch repair /// mismatch
    repair /// meiosis /// meiotic recombination ///
    meiotic prophase II /// meiosis
    RECQL RecQ protein-like (DNA helicase DNA repair /// DNA metabolism
    Q1-like)
    RAD52 RAD52 homolog (S. cerevisiae) double-strand break repair /// mitotic recombination ///
    meiotic recombination /// DNA repair /// DNA
    recombination /// response to DNA damage stimulus
    XRCC4 X-ray repair complementing DNA repair /// double-strand break repair /// DNA
    defective repair in Chinese hamster recombination /// DNA recombination /// response
    cells 4 to DNA damage stimulus
    XRCC4 X-ray repair complementing DNA repair /// double-strand break repair /// DNA
    defective repair in Chinese hamster recombination /// DNA recombination /// response
    cells 4 to DNA damage stimulus
    RAD17 RAD17 homolog (S. pombe) DNA replication /// DNA repair /// cell cycle ///
    response to DNA damage stimulus
    MSH3 mutS homolog 3 (E. coli) mismatch repair /// DNA metabolism /// DNA repair ///
    mismatch repair /// response to DNA damage stimulus
    MRE11A MRE11 meiotic recombination 11 regulation of mitotic recombination /// double-
    homolog A (S. cerevisiae) strand break repair via nonhomologous end-joining ///
    telomerase-dependent telomere maintenance ///
    meiosis /// meiotic recombination /// DNA
    metabolism /// DNA repair /// double-strand break
    repair /// response to DNA damage stimulus ///
    DNA repair /// double-strand break repair /// DNA
    recombination
    MSH6 mutS homolog 6 (E. coli) mismatch repair /// DNA metabolism /// DNA repair ///
    mismatch repair /// response to DNA damage stimulus
    MSH6 mutS homolog 6 (E. coli) mismatch repair /// DNA metabolism /// DNA repair ///
    mismatch repair /// response to DNA damage stimulus
    RECQL5 RecQ protein-like 5 DNA repair /// DNA metabolism /// DNA metabolism
    BRCA1 breast cancer 1, early onset regulation of transcription from RNA polymerase II
    promoter /// regulation of transcription from RNA
    polymerase III promoter /// DNA damage response,
    signal transduction by p53 class mediator resulting
    in transcription of p21 class mediator /// cell cycle ///
    protein ubiquitination /// androgen receptor
    signaling pathway /// regulation of cell proliferation ///
    regulation of apoptosis /// positive regulation of
    DNA repair /// negative regulation of progression
    through cell cycle /// positive regulation of
    transcription, DNA-dependent /// negative
    regulation of centriole replication /// DNA damage
    response, signal transduction resulting in induction
    of apoptosis /// DNA repair /// response to DNA
    damage stimulus /// protein ubiquitination /// DNA
    repair /// regulation of DNA repair /// apoptosis ///
    response to DNA damage stimulus
    RAD52 RAD52 homolog (S. cerevisiae) double-strand break repair /// mitotic recombination ///
    meiotic recombination /// DNA repair /// DNA
    recombination /// response to DNA damage stimulus
    POLD3 polymerase (DNA-directed), delta 3, DNA synthesis during DNA repair /// mismatch
    accessory subunit repair /// DNA replication
    MSH5 mutS homolog 5 (E. coli) DNA metabolism /// mismatch repair /// mismatch
    repair /// meiosis /// meiotic recombination ///
    meiotic prophase II /// meiosis
    ERCC2 excision repair cross-complementing transcription-coupled nucleotide-excision repair ///
    rodent repair deficiency, transcription /// regulation of transcription, DNA-
    complementation group 2 (xeroderma dependent /// transcription from RNA polymerase II
    pigmentosum D) promoter /// induction of apoptosis /// sensory
    perception of sound /// nucleobase, nucleoside,
    nucleotide and nucleic acid metabolism ///
    nucleotide-excision repair
    RECQL4 RecQ protein-like 4 DNA repair /// development /// DNA metabolism
    PMS1 PMS1 postmeiotic segregation mismatch repair /// regulation of transcription,
    increased 1 (S. cerevisiae) DNA-dependent /// cell cycle /// negative regulation
    of progression through cell cycle /// mismatch repair ///
    DNA repair /// response to DNA damage stimulus
    ZFP276 zinc finger protein 276 homolog transcription /// regulation of transcription,
    (mouse) DNA-dependent
    MBD4 methyl-CpG binding domain protein 4 base-excision repair /// DNA repair /// response to
    DNA damage stimulus /// DNA repair
    MBD4 methyl-CpG binding domain protein 4 base-excision repair /// DNA repair /// response to
    DNA damage stimulus /// DNA repair
    MLH3 mutL homolog 3 (E. coli) mismatch repair /// meiotic recombination /// DNA
    repair /// mismatch repair /// response to DNA
    damage stimulus /// mismatch repair
    FANCA Fanconi anemia, complementation DNA repair /// protein complex assembly /// DNA
    group A repair /// response to DNA damage stimulus
    POLE polymerase (DNA directed), epsilon DNA replication /// DNA repair /// DNA replication ///
    response to DNA damage stimulus
    XRCC3 X-ray repair complementing DNA repair /// DNA recombination /// DNA
    defective repair in Chinese hamster metabolism /// DNA repair /// DNA recombination ///
    cells 3 response to DNA damage stimulus /// response to
    DNA damage stimulus
    MLH3 mutL homolog 3 (E. coli) mismatch repair /// meiotic recombination /// DNA
    repair /// mismatch repair /// response to DNA
    damage stimulus /// mismatch repair
    NBN nibrin DNA damage checkpoint /// cell cycle checkpoint ///
    double-strand break repair
    SMUG1 single-strand selective carbohydrate metabolism /// DNA repair /// response
    monofunctional uracil DNA to DNA damage stimulus
    glycosylase
    FANCF Fanconi anemia, complementation DNA repair /// response to DNA damage stimulus
    group F
    NEIL1 nei endonuclease VIII-like 1 (E. coli) carbohydrate metabolism /// DNA repair /// response
    to DNA damage stimulus
    FANCE Fanconi anemia, complementation DNA repair /// response to DNA damage stimulus
    group E
    MSH5 mutS homolog 5 (E. coli) DNA metabolism /// mismatch repair /// mismatch
    repair /// meiosis /// meiotic recombination ///
    meiotic prophase II /// meiosis
    RECQL5 RecQ protein-like 5 DNA repair /// DNA metabolism /// DNA metabolism
  • For still another example, some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of a gene not associated with a disease (e.g., housekeeping genes), including, but not limited to, those related to transcription factors (e.g., ATF1, ATF2, ATF4, ATF6, ATF7, ATFIP, BTF3, E2F4, ERH, HMGB1, ILF2, IER2, JUND, TCEB2, etc.), repressors (e.g., PUF60), RNA splicing (e.g., BAT1, HNRPD, HNRPK, PABPN1, SRSF3, etc.), translation factors (EIF1, EIF1AD, EIF1B, EIF2A, EIF2AK1, EIF2AK3, EIF2AK4, EIF2B2, EIF2B3, EIF2B4, EIF2S2, EIF3A, etc.), tRNA synthetases (e.g., AARS, CARS, DARS, FARS, GARS, HARS, IARS, KARS, MARS, etc.), RNA binding protein (e.g., ELAVL1, etc.), ribosomal proteins (e.g., RPL5, RPL8, RPL9, RPL10, RPL11, RPL14, RPL25, etc.), mitochondrial ribosomal proteins (e.g., MRPL9, MRPL1, MRPL10, MRPL11, MRPL12, MRPL13, MRPL14, etc.), RNA polymerase (e.g., POLR1C, POLR1D, POLR1E, POLR2A, POLR2B, POLR2C, POLR2D, POLR3C, etc.), protein processing (e.g., PPID, PPI3, PPIF, CANX, CAPN1, NACA, PFDN2, SNX2, SS41, SUMO1, etc.), heat shock proteins (e.g., HSPA4, HSPA5, HSBP1, etc.), histone (e.g., HIST1HSBC, H1FX, etc.), cell cycle (e.g., ARHGAP35, RAB10, RAB11A, CCNY, CCNL, PPP1CA, RAD1, RAD17, etc.), carbohydrate metabolism (e.g., ALDOA, GSK3A, PGK1, PGAM5, etc.), lipid metabolism (e.g., HADHA), citric acid cycle (e.g., SDHA, SDHB, etc.), amino acid metabolism (e.g., COMT, etc.), NADH dehydrogenase (e.g., NDUFA2, etc.), cytochrome c oxidase (e.g., COX5B, COX8, COX11, etc.), ATPase (e.g. ATP2C1, ATP5F1, etc.), lysosome (e.g., CTSD, CSTB, LAMP1, etc.), proteasome (e.g., PSMA1, UBA1, etc.), cytoskeletal proteins (e.g., ANXA6, ARPC2, etc.), and organelle synthesis (e.g., BLOC1S1, AP2A1, etc.).
  • In still another example, some cell free DNAs/mRNAs are fragments of or those encoding a full length or a fragment of a neoepitope specific to the tumor. With respect to neoepitope, it should be appreciated that neoepitopes can be characterized as random mutations in tumor cells that create unique and tumor specific antigens. Therefore, high-throughput genome sequencing should allow for rapid and specific identification of patient specific neoepitopes where the analysis also considers matched normal tissue of the same patient. In some embodiments, neoepitopes may be identified from a patient tumor in a first step by whole genome analysis of a tumor biopsy (or lymph biopsy or biopsy of a metastatic site) and matched normal tissue (i.e., non-diseased tissue from the same patient) via synchronous comparison of the so obtained omics information. While not limiting to the inventive subject matter, it is typically preferred that the data are patient matched tumor data (e.g., tumor versus same patient normal), and that the data format is in SAM, BAM, GAR, or VCF format. However, non-matched or matched versus other reference (e.g., prior same patient normal or prior same patient tumor, or Homo statisticus) are also deemed suitable for use herein. Therefore, the omics data may be ‘fresh’ omics data or omics data that were obtained from a prior procedure (or even different patient). However, and especially where genomics ctDNA is analyzed, the neoepitope-coding sequence need not necessarily be expressed.
  • In particularly preferred aspects, the nucleic acid encoding a neoepitope may encode a neoepitope that is also a suitable target for immune therapy. Therefore, neoepitopes can then be further filtered for a match to the patient's HLA type to thereby increase likelihood of antigen presentation of the neoepitope. Most preferably, and as further discussed below, such matching can be done in silico. Most typically, the patient-specific epitopes are unique to the patient, but may also in at least some cases include tumor type-specific neoepitopes (e.g., Her-2, PSA, brachyury) or cancer-associated neoepitopes (e.g., CEA, MUC-1, CYPB1).
  • It is contemplated that cell free DNA/mRNA may present in modified forms or different isoforms. For example, the cell free DNA may be present in methylated or hydroxyl methylated, and the methylation level of some genes (e.g., GSTP1, p16, APC, etc.) may be a hallmark of specific types of cancer (e.g., colorectal cancer, etc.). The cell free mRNA may be present in a plurality of isoforms (e.g., splicing variants, etc.) that may be associated with different cell types and/or location. Preferably, different isoforms of mRNA may be a hallmark of specific tissues (e.g., brain, intestine, adipose tissue, muscle, etc.), or may be a hallmark of cancer (e.g., different isoform is present in the cancer cell compared to corresponding normal cell, or the ratio of different isoforms is different in the cancer cell compared to corresponding normal cell, etc.). For example, mRNA encoding HMGB1 are present in 18 different alternative splicing variants and 2 unspliced forms. Those isoforms are expected to express in different tissues/locations of the patient's body (e.g., isoform A is specific to prostate, isoform B is specific to brain, isoform C is specific to spleen, etc.). Thus, in these embodiments, identifying the isoforms of cell free mRNA in the patient's bodily fluid can provide information on the origin (e.g., cell type, tissue type, etc.) of the cell free mRNA.
  • The inventors contemplate that the quantities and/or isoforms (or subtypes) or regulatory noncoding RNA (e.g., microRNA, small interfering RNA, long non-coding RNA (lncRNA)) can vary and fluctuate by presence of a tumor or immune response against the tumor. Without wishing to be bound by any specific theory, varied expression of regulatory noncoding RNA in a cancer patient's bodily fluid may due to genetic modification of the cancer cell (e.g., deletion, translocation of parts of a chromosome, etc.), and/or inflammations at the cancer tissue by immune system (e.g., regulation of miR-29 family by activation of interferon signaling and/or virus infection, etc.). Thus, in some embodiments, the cell free RNA can be a regulatory noncoding RNA that modulates expression (e.g., downregulates, silences, etc.) of mRNA encoding a cancer-related protein or an inflammation-related protein (e.g., HMGB1, HMGB2, HMGB3, MUC1, VWF, MMP, CRP, PBEF1, TNF-α, TGF-β, PDGFA, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, Eotaxin, FGF, G-CSF, GM-CSF, IFN-7, IP-10, MCP-1, PDGF, hTERT, etc.).
  • It is also contemplated that some cell free regulatory noncoding RNA may be present in a plurality of isoforms or members (e.g., members of miR-29 family, etc.) that may be associated with different cell types and/or location. Preferably, different isoforms or members of regulatory noncoding RNA may be a hallmark of specific tissues (e.g., brain, intestine, adipose tissue, muscle, etc.), or may be a hallmark of cancer (e.g., different isoform is present in the cancer cell compared to corresponding normal cell, or the ratio of different isoforms is different in the cancer cell compared to corresponding normal cell, etc.). For example, higher expression level of miR-155 in the bodily fluid can be associated with the presence of breast tumor, and the reduced expression level of miR-155 can be associated with reduced size of breast tumor. Thus, in these embodiments, identifying the isoforms of cell free regulatory noncoding RNA in the patient's bodily fluid can provide information on the origin (e.g., cell type, tissue type, etc.) of the cell free regulatory noncoding RNA.
  • Isolation and Amplification of Cell Free DNA/RNA
  • Any suitable methods to isolate and amplify cell free DNA/RNA are contemplated. Most typically, cell free DNA/RNA is isolated from a bodily fluid (e.g., whole blood) that is processed under a suitable conditions, including a condition that stabilizes cell free RNA. Preferably, both cell free DNA and RNA are isolated simultaneously from the same badge of the patient's bodily fluid. Yet, it is also contemplated that the bodily fluid sample can be divided into two or more smaller samples from which DNA or RNA can be isolated separately. Once separated from the non-nucleic acid components, cell free RNA are then quantified, preferably using real time, quantitative PCR or real time, quantitative RT-PCR.
  • The bodily fluid of the patient can be obtained at any desired time point(s) depending on the purpose of the omics analysis. For example, the bodily fluid of the patient can be obtained before and/or after the patient is confirmed to have a tumor and/or periodically thereafter (e.g., every week, every month, etc.) in order to associate the cell free DNA/RNA data with the prognosis of the cancer. In some embodiments, the bodily fluid of the patient can be obtained from a patient before and after the cancer treatment (e.g., chemotherapy, radiotherapy, drug treatment, cancer immunotherapy, etc.). While it may vary depending on the type of treatments and/or the type of cancer, the bodily fluid of the patient can be obtained at least 24 hours, at least 3 days, at least 7 days after the cancer treatment. For more accurate comparison, the bodily fluid from the patient before the cancer treatment can be obtained less than 1 hour, less than 6 hours before, less than 24 hours before, less than a week before the beginning of the cancer treatment. In addition, a plurality of samples of the bodily fluid of the patient can be obtained during a period before and/or after the cancer treatment (e.g., once a day after 24 hours for 7 days, etc.).
  • Additionally or alternatively, the bodily fluid of a healthy individual can be obtained to compare the sequence/modification of cell free DNA, and/or quantity/subtype expression of cell free RNA. As used herein, a healthy individual refers an individual without a tumor. Preferably, the healthy individual can be chosen among group of people shares characteristics with the patient (e.g., age, gender, ethnicity, diet, living environment, family history, etc.).
  • Any suitable methods for isolating cell free DNA/RNA are contemplated. For example, in one exemplary method of DNA isolation, specimens were accepted as 10 ml of whole blood drawn into a test tube. Cell free DNA can be isolated from other from mono-nucleosomal and di-nucleosomal complexes using magnetic beads that can separate out cell free DNA at a size between 100-300 bps. For another example, in one exemplary method of RNA isolation, specimens were accepted as 10 ml of whole blood drawn into cell-free RNA BCT® tubes or cell-free DNA BCT® tubes containing RNA stabilizers, respectively. Advantageously, cell free RNA is stable in whole blood in the cell-free RNA BCT tubes for seven days while cell free RNA is stable in whole blood in the cell-free DNA BCT Tubes for fourteen days, allowing time for shipping of patient samples from world-wide locations without the degradation of cell free RNA. Moreover, it is generally preferred that the cell free RNA is isolated using RNA stabilization agents that will not or substantially not (e.g., equal or less than 1%, or equal or less than 0.1%, or equal or less than 0.01%, or equal or less than 0.001%) lyse blood cells. Viewed from a different perspective, the RNA stabilization reagents will not lead to a substantial increase (e.g., increase in total RNA no more than 10%, or no more than 5%, or no more than 2%, or no more than 1%) in RNA quantities in serum or plasma after the reagents are combined with blood. Likewise, these reagents will also preserve physical integrity of the cells in the blood to reduce or even eliminate release of cellular RNA found in blood cell. Such preservation may be in form of collected blood that may or may not have been separated. In less preferred aspects, contemplated reagents will stabilize cell free RNA in a collected tissue other than blood for at 2 days, more preferably at least 5 days, and most preferably at least 7 days. Of course, it should be recognized that numerous other collection modalities are also deemed appropriate, and that the cell free RNA can be at least partially purified or adsorbed to a solid phase to so increase stability prior to further processing.
  • As will be readily appreciated, fractionation of plasma and extraction of cell free DNA/RNA can be done in numerous manners. In one exemplary preferred aspect, whole blood in 10 mL tubes is centrifuged to fractionate plasma at 1600 rcf for 20 minutes. The so obtained plasma is then separated and centrifuged at 16,000 rcf for 10 minutes to remove cell debris. Of course, various alternative centrifugal protocols are also deemed suitable so long as the centrifugation will not lead to substantial cell lysis (e.g., lysis of no more than 1%, or no more than 0.1%, or no more than 0.01%, or no more than 0.001% of all cells). Cell free RNA is extracted from 2 mL of plasma using Qiagen reagents. The extraction protocol was designed to remove potential contaminating blood cells, other impurities, and maintain stability of the nucleic acids during the extraction. All nucleic acids were kept in bar-coded matrix storage tubes, with DNA stored at −4° C. and RNA stored at −80° C. or reverse-transcribed to cDNA that is then stored at −4° C. Notably, so isolated cell free RNA can be frozen prior to further processing.
  • Omics Data Processing
  • Once cell free DNA/RNA is isolated, various types of omics data can be obtained using any suitable methods. DNA sequence data will not only include the presence or absence of a gene that is associated with cancer or inflammation, but also take into account mutation data where the gene is mutated, the copy number (e.g., to identify duplication, loss of allele or heterozygosity), and epigenetic status (e.g., methylation, histone phosphorylation, nucleosome positioning, etc.). With respect to RNA sequence data it should be noted that contemplated RNA sequence data include mRNA sequence data, splice variant data, polyadenylation information, etc. Moreover, it is generally preferred that the RNA sequence data also include a metric for the transcription strength (e.g., number of transcripts of a damage repair gene per million total transcripts, number of transcripts of a damage repair gene per total number of transcripts for all damage repair genes, number of transcripts of a damage repair gene per number of transcripts for actin or other household gene RNA, etc.), and for the transcript stability (e.g., a length of poly A tail, etc.).
  • With respect to the transcription strength (expression level), transcription strength of the cell free RNA can be examined by quantifying the cell free RNA. Quantification of cell free RNA can be performed in numerous manners, however, expression of analytes is preferably measured by quantitative real-time RT-PCR of cell free RNA using primers specific for each gene. For example, amplification can be performed using an assay in a 10 μL reaction mix containing 2 μL cell free RNA, primers, and probe. mRNA of α-actin can be used as an internal control for the input level of cell free RNA. A standard curve of samples with known concentrations of each analyte was included in each PCR plate as well as positive and negative controls for each gene. Test samples were identified by scanning the 2D barcode on the matrix tubes containing the nucleic acids. Delta Ct (dCT) was calculated from the Ct value derived from quantitative PCR (qPCR) amplification for each analyte subtracted by the Ct value of actin for each individual patient's blood sample. Relative expression of patient specimens is calculated using a standard curve of delta Cts of serial dilutions of Universal Human Reference RNA set at a gene expression value of 10 (when the delta CTs were plotted against the log concentration of each analyte).
  • Alternatively, where discovery or scanning for new mutations or changes in expression of a particular gene is desired, real time quantitative PCR may be replaced by RNAseq to so cover at least part of a patient transcriptome. Moreover, it should be appreciated that analysis can be performed static or over a time course with repeated sampling to obtain a dynamic picture without the need for biopsy of the tumor or a metastasis.
  • Thus, omics data of cell free DNA/RNA preferably comprise a genomic data set that includes genomic sequence information. Most typically, the genomic sequence information comprises DNA sequence information of cell free DNA of the patient and optionally cell free DNA of a healthy individual. The sequence data sets may include unprocessed or processed data sets, and exemplary data sets include those having BAM format, SAM format, FASTQ format, or FASTA format. However, it is especially preferred that the data sets are provided in BAM format or as BAMBAM diff objects (see e.g., US2012/0059670A1 and US2012/0066001A1). Moreover, it should be noted that the data sets are reflective of the cell free DNA/RNA of the patient and of the healthy individual to so obtain patient and tumor specific information. Thus, genetic germ line alterations not giving rise to the diseased cells (e.g., silent mutation, SNP, etc.) can be excluded. Further, so obtained omics information can then be processed using pathway analysis (especially using PARADIGM) to identify any impact of any mutations on DNA repair pathways.
  • Likewise, computational analysis of the sequence data may be performed in numerous manners. In most preferred methods, however, analysis is performed in silico by location-guided synchronous alignment of cell free DNA/RNA of the patient and a healthy individual as, for example, disclosed in US 2012/0059670A1 and US 2012/0066001A1 using BAM files and BAM servers. Such analysis advantageously reduces false positive data and significantly reduces demands on memory and computational resources.
  • With respect to the analysis of cell free DNA/RNA of the patient and a healthy individual, numerous manners are deemed suitable for use herein so long as such methods will be able to generate a differential sequence object. However, it is especially preferred that the differential sequence object is generated by incremental synchronous alignment of BAM files representing genomic sequence information of the cell free DNA/RNA of the patient and a healthy individual. For example, particularly preferred methods include BAMBAM-based methods as described in US 2012/0059670 and US 2012/0066001.
  • Omics Data Analysis: Calculation of a Score
  • For calculation of a score, it should be appreciated that all data from ct/cf nucleic acids are deemed suitable for use herein and may therefore be specific to a particular tumor and/or patient and/or specific to a cancer. Furthermore, such data may be further normalized or otherwise preprocessed to adjust for age, treatment, gender, stage of disease, etc.
  • For example, in one aspect of the inventive subject matter the inventors contemplate that a library or reference base for all cancer-related genes, inflammation-related genes, DNA repair-related genes, and/or other non-disease related housekeeping genes can be created using one or more omics data for each of those genes, and such library is particularly useful where the omics data are associated with one or more health parameter. Viewed from a different perspective, while traditional methods of determining cancer prognosis or predicting treatment outcome have been based on a few number of genes, such library can provide a tool to generate a large cross-sectional database for all cancer-related gene activity, inflammation-related gene activity, DNA repair gene activity and housekeeping gene activity (as a control). The large cross-sectional database can be a basis for generating a cancer matrix, based on which a prognosis of a cancer, a health status of the patient, a likelihood of outcome of treatment, an effectiveness of the treatment can be more reliably calculated.
  • Of course, it should be appreciated that analyses presented herein may be performed over specific and diverse populations to so obtain reference values for the specific populations, such as across various health associated states (e.g., healthy, diagnosed with a specific disease and/or disease state, which may or may not be inherited, or which may or may not be associated with impaired DNA repair, inflammation-related autoimmunity, etc.), a specific age or age bracket, a specific ethnic group that may or may not be associated with frequent occurrence of specific type of cancer. Of course, populations may also be enlisted from databases with known omics information, and especially publically available omics information from cancer patients (e.g., TCGA, COSMIC, etc.) and proprietary databases from a large variety of individuals that may be healthy or diagnosed with a disease. Likewise, it should be appreciated that the population records may also be indexed over time for the same individual or group of individuals, which advantageously allows detection of shifts or changes in the genes and pathways associated with different types of cancers.
  • In further particularly preferred aspects, it is contemplated that a cancer score can be established for one or more cancer-related genes, inflammation-related genes, a DNA-repair gene, a neoepitope, and a gene not associated with a disease and that the score may be reflective of or even prognostic for various types of cancer that are at least in part due to mutations in cancer-related genes and/or pathways. For example, especially suitable cancer scores may involve scores for one or more genes associated with one or more types of cancer (e.g., BRCA1, BRCA2, P53, etc.) relative to another gene that may or may not be associated with one type of cancer (e.g., housekeeping genes, etc.). In another example, contemplated cancer scores may involve scores for one or more genes associated with one or more types of one or more types of cancer (e.g., BRCA1, BRCA2, P53, etc.) relative to an overall mutation rate (e.g., mutation rate of the genes not associated with a disease, etc.) to so better identify cancer relevant mutations over ‘background’ mutations.
  • Additionally, the omics data may be used to generate a general error status for an individual (or tumor within an individual), or to associate the number and/or type of alterations in cancer-related genes, inflammation-related genes, or a DNA-repair gene to identify a ‘tipping point’ for one or more gene mutations after which a general mutation rate skyrockets. For example, where a rate or number of mutations in ERCC1 and other DNA repair genes could have only minor systemic consequence, addition of further mutations to TP53 may result in a catastrophic increase in mutation rates. Thus, and viewed from a different perspective, mutations in the genes associated with DNA may be used to estimate the risk of occurrence for a DNA damage-based disease, and especially cancer and age-related diseases. In still further contemplated uses, so obtained omics information may be analyzed in one or more pathway analysis algorithms (e.g., PARADIGM) to so identify affected pathways and to so possibly adjust treatment where treatment employs DNA damaging agents. Pathway analysis algorithms may also be used to in silico modulate expression of one or more DNA repair genes, which may results in desirable or even unexpected in silico treatment outcomes, which may be translated into the clinic.
  • With respect to calculation, the inventors contemplate that the cancer score is typically a compound score reflecting status of a plurality of genes. For example, the cancer score can be calculated by counting any mutations (e.g., deletion, missense, nonsense, etc.) of any cancer-related genes, inflammation-related genes, and DNA-repair genes with one or more mutations as having a positive value, counting any changes in methylation or other modifications in DNA of counting any cancer-related genes, DNA-repair genes, counting any upregulation or downregulation in expression levels of RNA of any cancer-related genes, inflammation-related genes, and DNA-repair genes, counting any presence of tumor-specific, patient specific neoepitopes, counting any changes or ratios in RNA isotypes (splice variants) of counting any cancer-related genes and DNA-repair genes, and counting any changes in length of poly A tail of any cancer-related genes, inflammation-related genes, and DNA-repair genes.
  • The inventors further contemplate that each count may be weighed uniformly or biased, based on the significance of each count and then be assigned a value according to the weight of each count (e.g., each count corresponds to 1 point, some counts correspond to different scores such as 1 point, 3 points, 10 points, 100 points, etc.). Some mutations in some cancer related genes may be ‘leading indicators’ or triggers to activate other tumorigenesis mechanism or metastasis. Identification of such triggers may advantageously allow for early diagnosis or intervention of the cancer. Thus, for example, a mutation in a cancer-specific gene among cancer-related genes, inflammation-related genes, or DNA-repair genes may be weighed higher than other cancer-related genes or DNA-repair genes (e.g., at least 3 times, at least 5 times, at least 10 times, at least 100 times, etc.) and can be assigned to higher values accordingly. As used herein the cancer-specific gene refers any gene or mutation of the gene that is a known genetic disposition (e.g., significantly increase a susceptibility to the disease) of specific types of cancer (e.g., BRCA1 and BRCA2 for breast cancer and ovarian cancer, etc.). In another example, each gene in any cancer-related pathway or DNA-repair pathway may be differently weighed (e.g., most significant, significant, moderate, less significant, insignificant, etc.) and any mutation of a such gene that has any or no impact (e.g., adversely affect the pathway stream, etc.) on any cancer-related pathway or DNA-repair pathway may be weighed differently based on the significance of the impact. Thus, for example, gene A encoding a significant, unreplaceable protein A in a cancer pathway may be weighed heavier than another gene B encoding a redundant protein (replaceable with other proteins). Also, a nonsense mutation in gene A that results in nonfunctional protein may be weighed at least 3 times, at least 5 times, at least 10 times, at least 100 times than a silent mutation in gene A or a missense mutation which does not affect the function of protein A and can be assigned to higher values accordingly.
  • In some embodiments, some countings may weigh equally or differently based on the significance of each counting and then be assigned to a negative value according to the weight of each counting (e.g., each counting corresponds to −1 point, some countings correspond to different scores such as −1 point, −3 points, −10 points, −100 points, etc.). For example, upregulation of mRNA of gene C, which can compensate the loss of function of gene A, can be assigned to a negative value (e.g., −10 points) such that it can compensate the positive value of mutation of gene A (e.g., +10 points).
  • It is also contemplated that some countings may be differently weighed based on the degree of changes in expression level of some RNAs. For example, when the expression level of RNA “X” increases at least twice, at least 5 times, at least 10 times, at least 20 times, while other RNA expression level change is below 50% at best, then the increase of expression level of RNA “X” may be weighed at least 3 times, at least 5 times, at least 10 times, at least 100 times than other genes.
  • Most typically, the cancer score is compound score that is a total sum of all values assigned to all counts. In some embodiments, the cancer score can be a total sum of all values assigned to all counts (all omics data). In other embodiments, the cancer score can be a total sum of a selected number of values assigned to some counts (e.g., corresponding to specific pathways, specific types of genes, specific groups of mechanisms, etc.). Thus, the cancer score increases as more cancer-related genes or DNA-repair genes possess one or more mutations. In some embodiments, each mutation and/or change may be counted separately such that cancer scores may further increase where one or more cancer-related genes or DNA-repair genes show multiple mutations in a single gene. In other embodiments, cancer score may further increase when such multiple mutations in a single gene may further affect the function of the cancer-related genes or DNA-repair genes such that the multiple mutations drive the cells more cancer-prone, or more cancerous, or drive the cancer microenvironment more immune-resistant, and so on.
  • Alternatively or additionally, the cancer score can be presented as a trajectory with one or more counts as its vectors, where a few numbers of variables and/or factors dominantly govern in determination of cancer prognosis. Each of variables and/or factors can be presented as a vector, whose amplitude is corresponding to the point of each weighted counting, and the addition of those vectors provides a trajectory indicating the prognosis of the disease. Viewed form a different perspective, it should be appreciated that multiple analyses over time can be prepared for the same patient, and that changes over time (e.g., with or without treatment) may be assigned specific values that will yet again generate a time-dependent score. Such scores or changes over time may be classified and serve as leading indicator for treatment outcome, drug response, etc.
  • Additionally, it is also contemplated that the cancer score can be calculated with health information other than cf/ct nucleic acid data obtained from the patient's blood. For example, the health information may include expression levels/concentrations of several types of cytokines (e.g., IL-2, TNF-a, etc.) related to tumorigenesis/inflammation/immune response against the tumor, hormone levels (e.g., estrogen, progesterone, growth hormone, etc.), blood sugar level, alanine transaminase level (for liver function), creatine level (for kidney function), blood pressure, types and quantity of tumor cell-secreted proteins (e.g., soluble ligands of immune cell receptor, etc.) or foreign antigenic proteins (e.g., for virus or bacterial infection, etc.).
  • The inventors contemplated that the so obtained cancer score can be used to provide a diagnosis of cancer or risk of having or developing a cancer. In some embodiments, the calculated cancer score of a patient can be compared with an average cancer score of healthy individuals to determine the difference between two scores. Preferably, when the difference between two scores is above a threshold value, the patient may be diagnosed to have a tumor, or has a high risk to have a tumor. In other embodiments, the calculated cancer score of a patient can be compared with a predetermined threshold score. The predetermined threshold score can be a predetermined score, which may vary depending on patient's ethnicity, age, gender, or other health status. In other embodiments, the predetermined threshold score can a dynamic score that can be changed based on a previous cancer score and a diagnosis or treatment performed to the patient.
  • The inventors also contemplate that the so obtained cancer score can be used to provide a prognosis of the cancer. For example, the cancer scores can be calculated based on omics data obtained in month 1, month 3, month 6, and month 12 after the patient got diagnosed with a first stage of lung cancer, and each cancer score can be compared with a predetermined threshold score corresponding to the month 1, 3, 6, and 12. The cancer scores are about 120% of the threshold score in month 1 and 3, and the cancer score is about 180% in month 6, and 230% of the threshold score month 12. Such progress indicates that the prognosis of the lung cancer of the patient is not optimistic if the progress is not intervened. In another example, the cancer score can be calculated by highly weighing the presence of neoepitopes that are tumor-specific and patient-specific. In this example, the cancer scores can be calculated based on omics data obtained in month 1, month 3, month 6, and month 12 after the patient got diagnosed with a first stage of lung cancer, and each cancer score is calculated by highly weighing the presence/appearance of new epitope that is tumor/tissue specific. The cancer scores are about 120% of the threshold score in month 1 and 3, and the cancer score is about 140% in month 6, and 230% of the threshold score month 12. Such progress indicates a possible metastasis of the tumor to another organ (releasing different type of neoepitope) or development of different type of tumor in the same organ (releasing different type of neoepitope).
  • In a further example, the cancer scores can provide an indicator for treatment options. The treatment option may be a prophylactic treatment where the compound score is below the threshold value, indicating that the patient is unlikely to have a tumor for now or at least has low risk of developing a tumor. When the cancer score is above the threshold value and a majority portion of the cancer score highly weighted was overexpression of a cancer-related gene A (e.g., over a threshold such as at least 10%, at least 20%, at least 30%, at least 50%, etc.), then the cancer score can be used to provide the treatment option that may use a drug inhibiting the activity of cancer-related gene A (e.g., a blocker of protein A, etc.). Similarly, when the cancer score is above the threshold value and a majority portion of the cancer score highly weighted was overexpression of a gene encoding a receptor of an immune cell or a ligand of the receptor, then the cancer score can be used to provide the immunotherapy using the receptor or ligand of the immune cells. Also, when the cancer score is above the threshold value and a majority portion of the cancer score highly weighted was overexpression of a specific neoepitope, then the cancer score can be used to provide the immunotherapy using the neoepitope as a bait or a surgery/a radiation therapy to physically remove local tumors. Also such cancer scores may be an indicative of likelihood of success for the treatment option. However, if the portion of the cancer score highly weighted was overexpression of a cancer-related gene A is below the threshold, then the treatment option using a drug inhibiting the activity of cancer-related gene A may be predicted less effective.
  • Consequently, the patient can be treated with at least one of the treatment options based on the patient's cancer (compound) score. For example, above the threshold value and a majority portion of the cancer score highly weighted was overexpression of a specific neoepitope, the treatment option can be selected to include a recombinant virus (or yeast or bacteria) comprising a nucleic acid encoding the specific neoepitope. Then, the recombinant virus can be administered to the patient in a dose and schedule effective to treat the tumor and/or effective to reduce the cancer score of the patient for at least 10%, at least 20%, at least 30%, at least in 2 weeks, at least in 4 weeks, at least in 8 weeks, at least in 12 weeks after the administration or a series of administrations.
  • It is also contemplated that the patient's cancer score can be compared with one or more other patients having same type of cancer and having a treatment history to provide a treatment option and predicted outcome. For example, where other patients' history indicates that the drug treatment is effective only when the cancer score is below 200 (as absolute score), or less than 180% of the healthy individual's score, and the patient's cancer score has been increasing from 140 to 160 for the last 2 weeks, a recommendation to proceed with drug treatment no later than 2 weeks can be provided based on the other patients' history and cancer scores.
  • The calculated cancer score can also be an indicator of an effectiveness of a cancer treatment, especially when the omics data includes information of at least one or more genes encoding a target/indicator of the cancer treatment. For example, cancer scores can be calculated based on omics data obtained before the cancer treatment, 7 days after, 2 weeks, 1 month, and 6 months of the cancer treatment. The cancer score of 7 days after the treatment is 80% of the cancer score before the treatment, and the cancer score of 2 weeks and 1 month after the treatment is 50% of the cancer score before the treatment, and the cancer score of 6 months after the treatment is 150% of the cancer score before the treatment. Such progress indicates that the treatment was effective at least for a short term (e.g., up to 1 month), yet the effectiveness is decreased over time and may not effective at all in 6 months after the treatment. In some embodiments, the cancer scores before and after treatment can be compared with a predetermined threshold value to determine the effectiveness of the treatment. For example, if the cancer score is 200 before the treatment and 130 after the treatment where the threshold cancer score is 100, then the treatment can be determined “effective” as the cancer score drops below the threshold after the treatment. However, if the cancer score is 200 before the treatment and 160 after the treatment where the threshold cancer score is 150, then the treatment can be determined “not effective” as the cancer score stays above the threshold after the treatment even though the absolute value of the cancer score is decreased. Consequently, the inventors further contemplate that the patient continues with administering the treatment option (e.g., immune therapy, etc.) when the treatment can be determined “effective”, when the cancer score after the treatment is lower than the predetermined threshold, when the cancer score after the treatment is at most 5%, at most 10% higher than the predetermined threshold, or when the cancer score after the treatment is at least 5%, at least 10%, at least 15% lower than the predetermined threshold. s
  • The inventors also contemplate that the effectives of some cancer treatments can be determined by analyzing omics data including foreign DNA/RNA originated from a carrier of the immune therapy (e.g., virus, bacteria, yeast, etc.). For example, where the virus is a carrier to deliver a recombinant nucleic acid encoding recombinant killer activation receptor (KAR), the level of cell free DNA/RNA of recombinant KAR in the patient blood can be an indicator of an effectiveness of infection of the virus.
  • It should be apparent to those skilled in the art that many more modifications besides those already described are possible without departing from the inventive concepts herein. The inventive subject matter, therefore, is not to be restricted except in the scope of the appended claims. Moreover, in interpreting both the specification and the claims, all terms should be interpreted in the broadest possible manner consistent with the context. In particular, the terms “comprises” and “comprising” should be interpreted as referring to elements, components, or steps in a non-exclusive manner, indicating that the referenced elements, components, or steps may be present, or utilized, or combined with other elements, components, or steps that are not expressly referenced. As used in the description herein and throughout the claims that follow, the meaning of “a,” “an,” and “the” includes plural reference unless the context clearly dictates otherwise. Also, as used in the description herein, the meaning of “in” includes “in” and “on” unless the context clearly dictates otherwise. Where the specification claims refers to at least one of something selected from the group consisting of A, B, C . . . and N, the text should be interpreted as requiring only one element from the group, not A plus N, or B plus N, etc.

Claims (18)

What is claimed is:
1. A method of selecting a treatment option for a cancer patient, comprising:
obtaining blood from a patient having a cancer;
obtaining from the blood omics data of the cancer patient for a plurality of cancer genes, wherein the omics data comprise at least one of DNA sequence data, RNA sequence data, and RNA expression level;
wherein the omics data comprise an expression level of a cancer related gene, an expression level of an immune therapy related gene, and an expression level of a DNA or RNA sequence encoding a neoepitope;
providing an omics record computer system that includes at least one processor and at least one computer readable memory coupled to the processor and configured to digitally store the omics data for the plurality of cancer-related genes in the at least one memory;
analyzing, in silico, the omics data to generate a digital cancer gene score, wherein the digital cancer gene score is calculated in silico using the omics data; and
administering, when (1) the digital cancer gene score exceeds a threshold level and (2) the majority portion of the digital cancer gene score is weighted for the cancer related gene, the immune therapy related gene, or the DNA or RNA sequence encoding a neoepitope, a therapeutic agent that targets the cancer related gene, the immune therapy related gene, or the DNA or RNA sequence encoding the neoepitope.
2. The method of claim 1, wherein the blood omics data are obtained from DNA/RNA that is enclosed in a vesicular structure or bound to non-nucleotide molecule.
3. The method of claim 1, wherein the blood omics data are obtained from cell-free DNA or cell-free RNA.
4. The method of claim 1, wherein the DNA sequence data are selected from the group consisting of mutation data, copy number data duplication, loss of heterozygosity data, and epigenetic status.
5. The method of claim 1, wherein the RNA sequence data are selected from the group consisting of mRNA sequence data and splice variant data.
6. The method of claim 1, wherein the RNA expression level data are selected from the group consisting of a quantity of RNA transcript and a quantity of a small noncoding RNA.
7. The method of claim 1, wherein DNA sequence data are obtained from circulating free DNA.
8. The method of claim 1, wherein the RNA sequence data are obtained from the group consisting of circulating tumor RNA and circulating free RNA.
9. The method of claim 1, wherein the cancer related gene is selected form the group consisting of a gene encoding a protein kinase, a cancer-specific gene, a cancer associated gene, a DNA polymerase gene, a nuclease gene, a replicated associated gene,
10. The method of claim 1, wherein the immune therapy related gene is selected form the group consisting of a DNA repair gene, an RNA repair gene, an inflammation-related gene, a chemokine gene, a cytokine gene, a chemokine receptor gene, a cytokine receptor gene, a homologous recombination gene, non-homologous end joining gene,
11. The method of claim 1, wherein the DNA or RNA sequence encoding the neoepitope is a DNA or RNA sequence that encodes a patient-and tumor specific neoepitope, is a sequence that encodes a tumor type-specific neoepitope, or is a sequence that encodes a cancer-associated neoepitope.
12. The method of claim 1, wherein the neoepitope is matched to the patient's HLA type.
13. The method of claim 1, wherein the cancer related gene or the immune therapy related gene is selected form the group of genes presented in Table 1, Table 2, and Table 3.
14. The method of claim 1, further comprising a step of processing the omics data using pathway analysis to thereby identify one or more pathways affected by the expression level.
15. The method of claim 1, wherein the cancer score is a compound score reflecting status of the cancer related gene, the immune therapy related gene, and the DNA or RNA sequence encoding a neoepitope.
16. The method of claim 1, wherein the therapeutic agent is an enzyme inhibitor, a receptor ligand.
17. The method of claim 1, wherein the therapeutic agent is an immune therapy.
18. The method of claim 1, wherein the treatment option is surgery or radiation therapy.
US18/474,797 2017-10-12 2023-09-26 Cancer Score for Assessment and Response Prediction from Biological Fluids Pending US20240021314A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/474,797 US20240021314A1 (en) 2017-10-12 2023-09-26 Cancer Score for Assessment and Response Prediction from Biological Fluids

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201762571414P 2017-10-12 2017-10-12
PCT/US2018/055481 WO2019075251A2 (en) 2017-10-12 2018-10-11 Cancer score for assessment and response prediction from biological fluids
US202016754088A 2020-04-06 2020-04-06
US18/474,797 US20240021314A1 (en) 2017-10-12 2023-09-26 Cancer Score for Assessment and Response Prediction from Biological Fluids

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2018/055481 Continuation WO2019075251A2 (en) 2017-10-12 2018-10-11 Cancer score for assessment and response prediction from biological fluids
US16/754,088 Continuation US11810672B2 (en) 2017-10-12 2018-10-11 Cancer score for assessment and response prediction from biological fluids

Publications (1)

Publication Number Publication Date
US20240021314A1 true US20240021314A1 (en) 2024-01-18

Family

ID=66101142

Family Applications (3)

Application Number Title Priority Date Filing Date
US16/754,088 Active 2039-09-14 US11810672B2 (en) 2017-10-12 2018-10-11 Cancer score for assessment and response prediction from biological fluids
US18/465,868 Pending US20230420137A1 (en) 2017-10-12 2023-09-12 Cancer Score for Assessment and Response Prediction from Biological Fluids
US18/474,797 Pending US20240021314A1 (en) 2017-10-12 2023-09-26 Cancer Score for Assessment and Response Prediction from Biological Fluids

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US16/754,088 Active 2039-09-14 US11810672B2 (en) 2017-10-12 2018-10-11 Cancer score for assessment and response prediction from biological fluids
US18/465,868 Pending US20230420137A1 (en) 2017-10-12 2023-09-12 Cancer Score for Assessment and Response Prediction from Biological Fluids

Country Status (6)

Country Link
US (3) US11810672B2 (en)
CN (1) CN111213209A (en)
AU (1) AU2018348249A1 (en)
CA (1) CA3077221A1 (en)
SG (1) SG11202002711WA (en)
WO (1) WO2019075251A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111213209A (en) 2017-10-12 2020-05-29 南托米克斯有限责任公司 Cancer scoring and response prediction from assessment of biological fluids
US11168356B2 (en) * 2017-11-02 2021-11-09 The Chinese University Of Hong Kong Using nucleic acid size range for noninvasive cancer detection
EP3977477A1 (en) * 2019-05-24 2022-04-06 Yeda Research and Development Co. Ltd Method and system for predicting gestational diabetes
JP7168270B2 (en) * 2019-12-24 2022-11-09 イーソリューションズ株式会社 Health management system and health management method
US20220042109A1 (en) * 2020-08-06 2022-02-10 Agendia NV Methods of assessing breast cancer using circulating hormone receptor transcripts
CN116917733A (en) * 2020-12-17 2023-10-20 匹兹堡大学高等教育联邦体系 Multiple-set of learning methods for precision medicine
WO2023196390A1 (en) * 2022-04-06 2023-10-12 Foundation Medicine, Inc. Aneuploidy biomarkers associated with response to anti-cancer therapies
CN114959026A (en) * 2022-04-15 2022-08-30 深圳市陆为生物技术有限公司 Application of reagent for detecting gene in preparation of product for evaluating recurrence risk of breast cancer patient
CN115132327B (en) * 2022-05-25 2023-03-24 中国医学科学院肿瘤医院 Microsatellite instability prediction system, construction method thereof, terminal equipment and medium

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2797645C (en) 2010-05-25 2020-09-22 The Regents Of The University Of California Bambam: parallel comparative analysis of high-throughput sequencing data
US9646134B2 (en) 2010-05-25 2017-05-09 The Regents Of The University Of California Bambam: parallel comparative analysis of high-throughput sequencing data
EP2721180A4 (en) * 2011-06-16 2015-09-02 Decode Genetics Ehf Genetic variants for predicting risk of breast cancer
US9422592B2 (en) 2012-01-06 2016-08-23 Viomics, Inc. System and method of detecting RNAS altered by cancer in peripheral blood
WO2013190089A1 (en) * 2012-06-21 2013-12-27 Pangaea Biotech, S.L. Molecular biomarkers for predicting outcome in lung cancer
CN113337604A (en) 2013-03-15 2021-09-03 莱兰斯坦福初级大学评议会 Identification and use of circulating nucleic acid tumor markers
ES2829415T3 (en) * 2013-05-30 2021-05-31 Genomic Health Inc Gene expression profiling algorithm to calculate a recurrence score for a kidney cancer patient
CN107002122B (en) * 2014-07-25 2023-09-19 华盛顿大学 Method for determining tissue and/or cell type leading to the production of cell-free DNA and method for identifying diseases or disorders using the same
MA40737A (en) * 2014-11-21 2017-07-04 Memorial Sloan Kettering Cancer Center DETERMINANTS OF CANCER RESPONSE TO PD-1 BLOCKED IMMUNOTHERAPY
EP3384050A4 (en) 2015-12-03 2019-07-31 Alfred Health Monitoring treatment or progression of myeloma
US10982286B2 (en) 2016-01-22 2021-04-20 Mayo Foundation For Medical Education And Research Algorithmic approach for determining the plasma genome abnormality PGA and the urine genome abnormality UGA scores based on cell free cfDNA copy number variations in plasma and urine
MX2019000733A (en) * 2016-07-20 2019-05-02 Biontech Rna Pharmaceuticals Gmbh Selecting neoepitopes as disease-specific targets for therapy with enhanced efficacy.
WO2018231860A1 (en) * 2017-06-12 2018-12-20 Grail, Inc. Alignment free filtering for identifying fusions
CN111213209A (en) 2017-10-12 2020-05-29 南托米克斯有限责任公司 Cancer scoring and response prediction from assessment of biological fluids

Also Published As

Publication number Publication date
US20200335215A1 (en) 2020-10-22
SG11202002711WA (en) 2020-04-29
WO2019075251A2 (en) 2019-04-18
US11810672B2 (en) 2023-11-07
WO2019075251A3 (en) 2019-07-25
CN111213209A (en) 2020-05-29
AU2018348249A1 (en) 2020-04-16
US20230420137A1 (en) 2023-12-28
CA3077221A1 (en) 2019-04-18

Similar Documents

Publication Publication Date Title
US11810672B2 (en) Cancer score for assessment and response prediction from biological fluids
US20200165685A1 (en) Circulating rna for detection, prediction, and monitoring of cancer
US20180087114A1 (en) Early assessment of mechanism of action and efficacy of anti-cancer therapies using molecular markers in bodily fluid
US20220290252A1 (en) Method of isolating circulating nucleosomes
US20230323476A1 (en) Targeted cell free nucleic acid analysis
US20220399080A1 (en) Methods and products for minimal residual disease detection
US20190234955A1 (en) Exosome-guided treatment of cancer
TW201918560A (en) Circulating RNA for detection, prediction, and monitoring of cancer
US20200248267A1 (en) TUMOR VS. MATCHED NORMAL cfRNA
US20200234790A1 (en) Dna repair profiling and methods therefor
US20200385815A1 (en) Using cfRNA for Diagnosing Minimal Residual Disease
EP3778924A1 (en) Molecular predictors of patient response to radiotherapy treatment
US20230160881A1 (en) HMGB1 RNA And Methods Therefor
TW202317523A (en) Biomarkers for colorectal cancer treatment
US20240105279A1 (en) Methods and systems employing targeted next generation sequencing for classifying a tumor sample as having a level of homologous recombination deficiency similar to that associated with mutations in brca1 or brca2 genes

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION