DNA METHYLATION MARKERS FOR NONINVASIVE DETECTION OF CANCER AND USES THEREOF

Information

  • Patent Application
  • 20210171617
  • Publication Number
    20210171617
  • Date Filed
    July 09, 2019
    4 years ago
  • Date Published
    June 10, 2021
    3 years ago
Abstract
A “binary-categorical differentiation (BCD)” method for finding a combination of a small number (2-10) of exquisite DNA methylation positions in the human genome (CG IDs) for detecting cancer in DNA in biological material derived from a patient such as plasma, saliva, urine, feces, tissue biopsy, tissue swabs and tissue smears (such as pup smears) and distinguish it from other tissue cell free DNA and blood cells DNA. Another method for detecting tissue of origin of tumor DNA uses a combination of small number of unique DNA methylation positions in the human genome (CG IDs). Various novel combinations of CG IDs derived from tumor DNA are disclosed for detecting with high specificity and sensitivity a hepatocellular carcinoma (HCC), b. lung cancer, c. prostate cancer, d. breast cancer, e. colorectal cancer, f. pancreatic cancer, g. brain cancer (glioblastoma), h. gastric cancer i. ovarian cancer, j. cervical cancer k. head and neck squamous cell carcinoma (HNSC), l. esophageal cancer m. bladder cancer, n. renal cancer, o. testicular cancer, p. common solid tumors, q. blood cancers, r. acute myeloid leukemia (AML), s. melanoma by measuring the DNA methylation of a combination of specific CG IDs and deriving a “methylation score” disclosed herein. Kits for predicting cancer using CG IDs using multiplexed next generation sequencing methylation assays, pyrosequencing assays and methylation specific PCR from a small volume of plasma. Various methods using plasma, urine, feces, tissue biopsy or tissue swabs help lead to prediction of cancer in persons with no other clinical evidence for cancer.
Description
FIELD OF THE INVENTION

The present invention relates to DNA methylation signatures in human DNA, particularly in the field of molecular diagnos.ics.


BACKGROUND OF THE INVENTION

Cancer has become a major killer of humans. Early detection of cancer can significantly improve cure rates and reduce the horrific personal and financial cost to the patients their families and the health care system For example. Hepatocellular Carcinoma (HCC) is the fifth most common cancer world-wide (El-Serag, 2011). It is particularly prevalent in Asia, and its occurrence is highest in areas where hepatitis B is prevalent, indicating a possible causal relationship (Flores & Marrero, 2014). Follow up of high-risk populations such as chronic hepatitis patients and early diagnosis of transitions from chronic hepatitis to HCC wculd improve cure rates. The survival rate of hepatocellular carcinoma is currently extremely low because it is almost always diagnosed at the late stages. Liver cancer could be effectively treated with cure rates of >80% if diagnosed early. Advances in imaging have improved noninvasive detection of HCC (Tan, Low, & Thng, 2011; Valente et al., 2014). However, current diagnostic methods, which include imaging and immunoassays with single proteins such as alpha- fetoprotein often fail to diagnose HCC early (Flores & Marrero, 2014) These challenges are not limited to HCC but common to other cancers as well. For example, early detection of breast cancer and colorectal cancer could dramatically reduce morbidity and mortality and the cost to the public health system and insurance companies. Moreover, certain cancers such as pancreatic cancers are detected almost invariably late resulting in virtually certain mortality. Advances in imaging have improved early detection of Cancers, however high-resolution imaging such as MRI is expensive, requires highly trained personnel and is unavailable in many locations. It has not evolved yet to a method of screening of wide populations. To have an impact on reducing morbidity and mortality from cancer it is necessary to develop a noninvasive, robust but nevertheless low-cost method that could be used in wide geographic areas for routine screening of the population. The main challenge is that solid tumors hide in internal organs and evolve long before they exhibit clinical symptoms. It is however possible to obtain tumor material noninvasively.


It is widely established by now that tumor DNA is shed into the system and could be found in plasma (Warton & Samimi, 2015) and possibly other secreted body fluids such as urine and saliva, as well as feces. By measuring molecular characteristics of tumor DNA, it is possible to determine that the DNA found in body fluids originated in the tumor (Zhai et al., 2012). Although tumor cells develop mutations that could distinguish tumor DNA from normal cells DNA, the number of possible mutations is vast and common mutations don't occur in all tumors (Dominguez-Vigil, Moreno-Martinez, Wang, Roehrl, & Barrera-Saldana, 2018).


DNA methylation, a covalent modification of DNA, which is a primary mechanism of epigenetic regulation of genome function is ubiquitously altered in tumors (Aguirre-Ghiso, 2007; Baylin et al., 2001; Ehrlich, 2002; Issa et al., 1993). DNA methylation profiles of tumors are potentially robust tools for tumor classification, prognosis and prediction of response to chemotherapy (Stefansson et al., 2014). The major drawback for using tumor DNA methylation in early diagnosis is that it requires invasive procedures and anatomical visualization of the suspected tumor. Circulating tumor cells are a noninvasive source of tumor DNA and are used for measuring DNA methylation in tumor suppressor genes (Radpour et al., 2011). Hypomethylation of HCC DNA is detectable in patients' blood (Ramzy, Omran, Hamad, Shaker, & Abboud, 2011) and genome wide bisulfite sequencing was recently applied to detect hypomethylated DNA in plasma from HCC patients (Chan et al., 2013). However, this source is limited, particularly at early stages of cancer and the DNA methylation profiles are confounded by host DNA methylation profiles. Genome wide bisulfite sequencing is a relatively costly procedure and requires significant bioinformatics analysis which makes it unfeasible as a screening tool. The challenge is therefore to delineate a small number of CGs that could robustly differentiate tumor DNA from nontumor DNA and develop a high throughput low cost assay that will enable the screening of wide populations in broad and diverse geographic areas. More recently several groups have performed comparative analysis of genome wide DNA methylation maps of cancer and normal DNA and blood DNA (Zhai et al., 2012). However, the main challenge with these approaches is that they have not taken into account cell free DNA from other tissues that is found in blood at different levels that are unanticipated a priori. Contaminating DNA from another tissue that has a similar methylation profile as the cancer tissue could lead to false positives. In addition, past approaches have quantitatively compared DNA methylation in normal and cancer tissues. This quantitative difference is diluted when tumor DNA is mixed with different and unknown amounts of DNA from other untransformed tissues, which can cause false negatives. These deficiencies in current methods necessitate a different approach that is disclosed in the present inventive subject matter.


Further publications dealing with the use of systems and methods for detecting cancer are: Grigg G, Clark S. Sequencing 5-methylcytosine residues in genomic DNA. Bioessays. 1994 June; 16(6):431-6, 431; Zeschnigk M, Schmitz B, Dittrich B, Buiting K, Horsthemke B, Doerfler W. Imprinted segments in the human genome: different DNA methylation patterns in the Prader-Willi/Angelman syndrome region as determined by the genomic sequencing method. Hum Mol Genet. 1997 March; 6(3):387-95; Feil R, Charlton J, Bird A P, Walter J, Reik W. Methylation analysis on individual chromosomes: improved protocol for bisulphite genomic sequencing. Nucleic Acids Res. 1994 February 25; 22(4):695-6; Martin V, Ribieras S, Song-Wang X, Rio M C, Dante R. Genomic sequencing indicates a correlation between DNA hypomethylation in the 5′ region of the pS2 gene and its expression in human breast cancer cell lines. Gene. 1995 May 19; 157(1-2):261-4; WO 97 46705, WO 95 15373 and WO 45560.

  • Aguirre-Ghiso, J. A. (2007). Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer, 7(11), 834-846. doi:10.1038/nrc2256
  • Baylin, S. B., Esteller, M., Rountree, M. R., Bachman, K. E., Schuebel, K., & Herman, J. G. (2001). Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer. Hum Mol Genet, 10(7), 687-692.
  • Breitbach, S., Tug, S., Helmig, S., Zahn, D., Kubiak, T., Michal, M., . . . Simon, P. (2014). Direct quantification of cell-free, circulating DNA from unpurified plasma. PLoS One, 9(3), e87838. doi:10.1371/journal.pone.0087838
  • Chan, K. C., Jiang, P., Chan, C. W., Sun, K., Wong, J., Hui, E. P., . . . Lo, Y. M. (2013). Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc Natl Acad Sci USA, 110(47), 18761-18768. doi:10.1073/pnas.1313995110
  • Dominguez-Vigil, I. G., Moreno-Martinez, A. K., Wang, J. Y., Roehrl, M. H. A., & Barrera-Saldana, H. A. (2018). The dawn of the liquid biopsy in the fight against cancer. Oncotarget, 9(2), 2912-2922. doi:10.18632/oncotarget.23131
  • Ehrlich, M. (2002). DNA methylation in cancer: too much, but also too little. Oncogene, 21(35), 5400-5413.
  • El-Serag, H. B. (2011). Hepatocellular carcinoma. N Engl J Med, 365(12), 1118-1127. doi:10.1056/NEJMra1001683
  • Flores, A., & Marrero, J. A. (2014). Emerging trends in hepatocellular carcinoma: focus on diagnosis and therapeutics. Cin Med Insights Oncol, 8, 71-76. doi:10.4137/CMO.S9926
  • Issa, J. P., Vertino, P. M., Wu, J., Sazawal, S., Celano, P., Nelkin, B. D., . . . Baylin, S. B. (1993). Increased cytosine DNA-methyltransferase activity during colon cancer progression. J Natl Cancer Inst, 85(15), 1235-1240.
  • Luczak, M. W., & Jagodzinski, P. P. (2006). The role of DNA methylation in cancer development. Folia Histochem Cytobiol, 44(3), 143-154.
  • Radpour, R., Barekati, Z., Kohler, C., Lv, Q., Burki, N., Diesch, C., . . . Zhong, X. Y. (2011). Hypermethylation of tumor suppressor genes involved in critical regulatory pathways for developing a blood-based test in breast cancer. PLoS One, 6(1), e16080. doi:10.1371/journal.pone.0016080
  • Ramzy, II, Omran, D. A., Hamad, O., Shaker, O., & Abboud, A. (2011). Evaluation of serum LINE-1 hypomethylation as a prognostic marker for hepatocellular carcinoma. Arab J Gastroenterol, 12(3), 139-142. doi:10.1016/j.ajg.2011.07.002
  • Stefansson, O. A., Moran, S., Gomez, A., Sayols, S., Arribas-Jorba, C., Sandoval, J., . . . Esteller, M. (2014). A DNA methylation-based definition of biologically distinct breast cancer subtypes. Mol Oncol. doi:10.1016/j.molonc.2014.10.012
  • Tan, C. H., Low, S. C., & Thng, C. H. (2011). APASL and AASLD Consensus Guidelines on Imaging Diagnosis of Hepatocellular Carcinoma: A Review. Int J Hepatol, 2011, 519783. doi:10.4061/2011/519783
  • Valente, S., Liu, Y., Schnekenburger, M., Zwergel, C., Cosconati, S., Gros, C., . . . Mai, A. (2014). Selective non-nucleoside inhibitors of human DNA methyltransferases active in cancer including in cancer stem cells. J Med Chem, 57(3), 701-713. doi:10.1021/jm4012627
  • Warton, K., & Samimi, G. (2015). Methylation of cell-free circulating DNA in the diagnosis of cancer. Front Mol Biosci, 2, 13. doi:10.3389/fmolb.2015.00013
  • Xu, R. H., Wei, W., Krawczyk, M., Wang, W., Luo, H., Flagg, K., . . . Zhang, K. (2017). Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat Mater, 16(11), 1155-1161. doi:10.1038/nmat4997
  • Zhai, R., Zhao, Y., Su, L., Cassidy, L., Liu, G., & Christiani, D. C. (2012). Genome-wide DNA methylation profiling of cell-free serum DNA in esophageal adenocarcinoma and Barrett esophagus. Neoplasia, 14(1), 29-33.


SUMMARY OF THE INVENTION

Embodiments of the claimed subject matter show that cancer is associated with a set of “categorically” distinct DNA methylation signatures that are different from any normal tissue and blood cell DNA methylation profiles. These sites create a binary differentiation between cancer and other tissues, whereby these sites are only methylated in cancer and fully unmethylated in other cancers. Thus, it is possible using deep next generation sequencing to detect even a few molecules of cancer cells on the background of normal cell DNA profile of methylation. Embodiments of the inventive subject matter enable detection of cell free tumor DNA even on a high background of cell free DNA from other tissues and are thus particularly suitable for early detection of cancer using cell free (CF) DNA extracted from body fluids, for example saliva, plasma, urine, feces etc. Embodiments also allow for early detection of cancer in tissue smears such as pup smears as well as biopsies and needle biopsies. Previous analyses in the prior art only compared normal and cancer cells from the same tissue and blood and derived sites that are quantitatively different in their DNA methylation level (Xu et al., 2017). However, sites discovered by such prior art analyses can't detect CF tumor DNA when it is mixed with other tissue CF DNA (see FIG. 2 for ctDNA markers for HCC from Sun Yat Sen University Cancer hospital). One embodiment of the present claimed subject matter reveals a unique set of sites that are unmethylated in all tissues but methylated in specific cancers. Another embodiment reveals a method to discover categorically distinct methylation sites in cancers, other tissues and other diseases called the “binary-categorical differentiation (BCD) method” using different sources of genome wide DNA methylation data derived by next generation sequencing, MeDIP arrays, MeDIP sequencing, and the like. One embodiment reveals a combination of “Categorical” DNA methylation sites for detection of a. hepatocellular carcinoma (HCC), b. lung cancer, c. prostate cancer, d. breast cancer, e. colorectal cancer, f. head and neck squamous cell carcinoma (HNSC). g. pancreatic cancer, h. brain cancer (glioblastoma), i. gastric cancer j. ovarian cancer, k. cervical cancer, 1. esophageal carcinoma m. bladder cancer, n. renal cancer, o. testicular cancer, p. common solid tumors, q. blood cancer profiles in a discovery set of genome wide data. Another embodiment also reveals a combination of “Categorical” DNA methylation sites that differentiate tumors by their tissue of origin. This embodiment differentiates the assay from prior art methods for detecting methylated CF DNA which have low tissue specificity. Embodiments validate the polygenic DNA methylation assays for detection of cancer in DNA methylation data from hundreds of patients as well as the tissue of origins of the tumors with high sensitivity and specificity. The present invention discloses a method that accurately measures DNA methylation in a polygenic set of CG IDs in hundreds of people concurrently, by sequential amplification with target specific primers followed by barcoding primers and multiplexed sequencing in a single next generation Miseq sequencing reaction, data extraction and quantification of methylation from a small volume of body fluids such as plasma, saliva or urine. Another embodiments of the inventive subject matter also discloses measurement of methylation of the said DNA methylation CG IDs using pyrosequencing assays or methylation specific PCR. Another embodiment discloses the calculation of either a “categorical” or a polygenic weighted methylation score that differentiates people with cancer from healthy people. Another embodiment discloses a novel process leading from plasma, urine, feces, tissue biopsy or tissue swabs to prediction of cancer in a person with no other clinical evidence for cancer. Another embodiment could be used by any person skilled in the art to detect cancer as well as other diseases that involve cell death and release of CF DNA such Alzheimer disease and other neurodegenerative diseases for neurons, heart disease for heart muscle cells. The DNA methylation markers (CG IDs) described in the embodiments will be utilized for a. noninvasive early detection of cancer in otherwise “healthy” people through routine “checkup”; b. monitoring “high risk” people such as chronic hepatitis patients who are at high risk for HCC or smokers who are at high risk for lung cancer; c. monitoring response to therapy in patients undergoing cancer treatment to detect recurrence or metastasis.


Embodiments demonstrate the utility detecting cancer of unknown samples using polygenic or categorical scores based on the DNA methylation measurement methods disclosed herein. The disclosed embodiments could be used by any person skilled in the art to detect cancer in body fluids, feces, urine and tissues of any cancer or diseased tissue using any method for methylation analysis that are available to those skilled in the art such as for example next generation bisulfite sequencing, Illumina Epic microarrays, capture sequencing, methylated DNA Immunoprecipitation (MeDIP) methylation specific PCR and any methylation measurements that becomes available.


Embodiments also disclose the potential for discovery of new “polygenic” categorical DNA methylation markers for other cancers and diseases using any method available to people skilled in the art for genome wide sequencing such as next generation bisulfite sequencing, MeDip sequencing, ion torrent sequencing, Epic microarrays etc. followed by binary-categorical differentiation (BCD) method of analysis for discovering specific and sensitive markers that will be used for noninvasive detection of disease Embodiments of the present inventive subject matter include:


In a first aspect, embodiments provide polygenic DNA methylation markers of cancer in cell free DNA in body fluids such as plasma for early detection of cancer, said polygenic DNA methylation markers set being derived using “binary-categorical differentiation (BCD) analysis” as disclosed herein on genome wide DNA methylation derived by mapping methods such as Illumina 450K or EPIC arrays, genome wide bisulfite sequencing, methylated DNA Immunoprecipitation (MeDIP) sequencing or hybridization with oligonucleotide arrays.


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below (or a short subset of this list such as the example listed below Table 1) for early detection of Liver cancer hepatocellular carcinoma (HCC) using plasma CF DNA or other body fluid CF DNA.









TABLE 1





Liver_detect




















cg00370303
cg10900437
cg02012576
cg16460359
cg04035559
cg17419241


cg00931619
cg11223367
cg03768777
cg16977570
cg04085025
cg18607529


cg05040544
cg19289599
cg06233293
cg24804544
cg26523670
cg09992116


cg05739190
cg24599205










Subset for detect:


cg02012576, cg03768777, cg24804544, cg05739190


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example shown below Table 2) for specifying the origin of the cancer as HCC and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 2





Liver_spec




















cg12137206
cg14126493
cg06105778
cg22076972
cg13341720
cg03705926


cg09363194
cg07036412
cg02702614
cg10181419
cg05876864
cg11068343


cg17167468
cg15375239
cg00026222
cg17283781
cg16147221
cg26386472


cg14570307
cg06207432
cg07610192
cg03422204
cg11684022
cg23693289


cg21107197
cg04920951
cg20385508
cg25296314
cg20707679
cg26703661


cg00456086
cg05009389
cg19388016
cg08460435
cg04739306
cg04221886


cg26797073
cg04109768
cg05337743
cg00483503
cg18668780
cg10604002


cg27650175
cg05684891
cg26026416
cg00177496
cg14221460
cg16551483


cg13438961
cg24432073
cg21059834
cg23305567
cg04809136
cg21105227










Subset for specificity:


cg14126493


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 3) for early detection of lung cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 3





Lung_detect




















cg04223424
cg06530490
cg08944430
cg09463882
cg11017065
cg12405785


cg16405026
cg21410080
cg23141355
cg25024074










Subset for detect:


cg04223424, cg23141355


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 4) for specifying the origin of the cancer as lung cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 4





Lung_spec




















cg05917792
cg02682457
cg23407396
cg23601095
cg23141355
cg15386964


cg02578368
cg24631970
cg27487839
cg16405026


cg23141355
cg06530490
cg04223424
cg25470077
cg07138603
cg23460835


cg20678442
cg15436096










Subset for spec:


cg05917732, cg25470077


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below (or a short subset of this list such as the example listed below Table 5) for early detection of prostate cancer as well as for specifying the origin of the cancer as prostate cancer and discriminating from other 16 common solid tumor cancers using plasma CF DNA or other body fluid CF DNA.









TABLE 5





Prostate_detect_spec


















cg14283569
cg18085998
cg13303553
cg17929627










Subset for detect_spec:


cg14283569


[it is a subset of the 4 listed in the table above]


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 6) for early detection of breast cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 6





Breast_detect




















cg13031251
cg19595750
cg16842053
cg18565473
cg09734791
cg25694349


cg09695735
cg03637878
cg26261793
cg18800085
cg16132520
cg05617413


cg17228900
cg06945936
cg08406370
cg24427504
cg26937500
cg11297107


cg02215070
cg14140647
cg05377226
cg07070305
cg24899571
cg07844931










Subset for detect:


cg13031251, cg09734791, cg09695735, cg03637878


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 7) for specifying the origin of the cancer as breast cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 7





Breast_Spec



















cg00467244
cg04194674
cg06879394
cg10720997
cg19966212


cg00722320
cg04751811
cg06998282
cg11498607
cg24525457


cg01308827
cg06282270
cg08066035
cg14862207
cg26228266


cg03113878
cg06405186
cg08296680
cg17945153
cg20180843










Subset for spec:


cg03113878, cg20180843


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 8) for early detection of colorectal cancer (CRC) as well as for specifying the origin of the cancer as colorectal cancer and discriminating from other 16 common solid tumor cancers using plasma CF DNA or other body fluid CF DNA.









TABLE 8





CRC_detect_spec




















cg08808128
cg03788131
cg09854653
cg21627760
cg08169901
cg07494047


cg01566242
cg13788592
cg24102266
cg17716617
cg16733654










Subset for detect-spec


cg09854653, cg01566242


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 9) for early detection of pancreatic cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 9





Pancreas_detect




















cg11017065
cg23833588
cg25024074
cg21277995
cg22286978
cg06241792


cg11591516
cg10096177
cg12035092
cg03611007
cg17996329
cg13807970


cg16678602
cg15386964
cg01423964
cg07900968
cg19118812
cg06728579


cg16232979
cg08406370










Subset for detect:


cg25024074, cg15386964, cg16232979


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 10) for specifying the origin of the cancer as pancreatic cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 10





Pancreas_Spec




















cg01237565
cg08182975
cg15323936
cg19102272
cg01311909
cg26927232


cg22290704
cg15832577
cg29983577
cg26466027
cg09796911
cg15850155


cg15510118
cg25591377
cg16165258
cg25595541
cg18151519
cg19749445


cg14870128
cg13441142










Subset for spec:


cg01237565, cg08182975, cg20983577, cg25591377


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 11) for early detection of brain cancer (glioblastoma) as well as for specifying the origin of the cancer as brain cancer (glioblastoma) and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid CF DNA.










TABLE 11





Brain_detect-spec




















cg06887260
cg19116006
cg10938374
cg03663746
cg25568243
cg27449131


cg24917627
cg04134305
cg09797645
cg02892595
cg13231951
cg26269703


cg09183941
cg16842053
cg26551897
cg26988692
cg07849581
cg25026703


cg06798642
cg22963915
cg04245373
cg25533556
cg24917627
cg27659841


cg04692993
cg06045408
cg06887260
cg11345323
cg17167076
cg17526812











cg19929355
cg22513169
cg22865720










Subset for spec-detect


Cg19929355

In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 12) for early detection of stomach (gastric) cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 12





Stomach_detect




















cg04125371
cg05377226
cg05611779
cg06241792
cg07900968
cg09734791


cg11017065
cg12510981
cg13807970
cg15760257
cg18323466
cg19118812


cg19419279
cg19769760
cg20334243
cg26261793










Subset for detect:


cg05611779, cg09734791, cg15760257


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example shown below Table 13) for specifying the origin of the cancer as gastric cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 13





Stomach_Spec




















cg17187167
cg07350470
cg05000488
cg25612145
cg09851120
cg00904726


cg04911739
cg18192294
cg11861709
cg19452853
cg02706110
cg03768513


cg05611779
cg06981182
cg06118999
cg04812509
cg10131095
cg05339066


cg19235339










Subset for spec:


cg05611779, cg19235339


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example shown below Table 14) for early detection of ovarian cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 14





Ovarian_detect




















cg24339193
cg04008429
cg06849719
cg22694153
cg11252337
cg15804105


cg12479674
cg06961071
cg21210985
cg01556502
cg02537149
cg23983315


cg03597143
cg27209395










Subset for detect:


cg24339193, cg22694153, cg11252337, cg21210985


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 15) for specifying the origin of the cancer as ovarian cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 15





Ovarian_spec




















cg00895834
cg01159194
cg01961086
cg02649698
cg03345116
cg03456771


cg09392827
cg10178270
cg10581012
cg13459217
cg15701612
cg17130982


cg05901462
cg07068768
cg08389588
cg09173621
cg19276014
cg19846609


cg18476766
cg19129687










Subset for spec:


cg07068768, cg19846609


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example shown below Table 16) for early detection of cervical cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 16





Cervix_detect




















cg00578154
cg13644629
cg01423964
cg15745619
cg04522671
cg21621906


cg00757182
cg14962363
cg01601746
cg17228900
cg07126167
cg22806837


cg08134106
cg22922289
cg09260640
cg24625128
cg11259628
cg27420520


cg08535260
cg23141355
cg09734791
cg25024074










Subset for detect:


cg00757182, cg01601746


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 17) for specifying the origin of the cancer as cervical cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 17





Cervix_spec




















cg00829990
cg02401399
cg04996219
cg07066594
cg07195011
cg07576142


cg09260640
cg12961842
cg13668618
cg18543270










Subset for spec:


cg07066594, cg09260640, cg12961842


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 18) for early detection of head and neck squamous cell carcinoma (HNSC) carcinoma using plasma CF DNA or other body fluid CF DNA.










TABLE 18





HNSC_detect




















cg01613638
cg02776314
cg03280624
cg04524120
cg05151803
cg07746323


cg07900968
cg08406370
cg11108676
cg12083965
cg15397448
cg17428324











cg18403606
cg20334243
cg26770917
cg27009208
cg27420520










Subset for detect:


cg07900968, cg20334243, cg27420520


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 19) for specifying the origin of the cancer as head and neck squamous cell carcinoma (HNSC) and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 19





HNSC_spec




















cg01217080
cg03522799
cg06672120
cg09136346
cg10155875
cg18006328


cg18443253
cg19287220










Subset for spec:


cg18006328, cg19287220


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or in a short subset of this list (such as the example listed below Table 20) for early detection of esophageal carcinoma using plasma CF DNA or other body fluid CF DNA.










TABLE 20





Esophagus_detect




















cg03280624
cg03735888
cg06530490
cg06963053
cg08944430
cg09734791


cg11017065
cg12035092
cg18344922
cg19118812
cg20334243
cg22128431











cg23141355
cg24740531
cg27009208
cg27420520










Subset for detect:


cg03280624, cg03735888, cg09734791, cg27420520


In one embodiment, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 21) for specifying the origin of the cancer as esophageal carcinoma and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 21





Esophagus_spec




















cg00532449
cg02763101
cg04743654
cg08055087
cg08932440
cg09556952


cg10608333
cg12473285
cg12966367
cg17579667
cg17949440
cg18723937


cg21554552
cg22647407
cg23286646
cg23730575
cg27569446










Subset for spec:


Cg09556952, cg12473285


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 22) for early detection of bladder cancer using plasma CF DNA or other body fluid CF DNA.









TABLE 22





Bladder_detect




















cg01423964
cg04223424
cg01556502
cg23141355
cg10723962
cg09734791


cg25024074
cg21039778
cg19113641
cg04342821
cg09008417
cg05873285










Subset for detect:


cg04223424, cg10723962, cg25024074


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (as shown in the example listed below Table 23) for specifying the origin of the cancer as bladder cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid DNA.









TABLE 23





Bladder_spec




















cg14773260
cg08827322
cg05085809
cg13544006
cg02293936
cg10153335


cg05039004
cg14718568
cg07790170
cg12995090
cg16495265
cg01508045


cg04933208
cg03231163
cg06708937
cg00675569
cg06312813
cg14126688


cg12984729
cg02384661
cg08307030
cg26884027
cg20540209
cg04355159


cg08857479
cg23395715
cg22006640
cg26279336
cg19898108
cg17446010


cg12911122










Subset for spec:


cg13544006


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as the example listed below Table 24) for early detection of renal (kidney) cancer for specifying the origin of the cancer as renal cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid CF DNA.









TABLE 24





Kidney_detect_spec




















cg08884571
cg00011225
cg14535274
cg05507490
cg10367244
cg07190763


cg02971546
cg19990785
cg03084949
cg08765317
cg02820958
cg23946709


cg26642667










Subset for detect spec:


cg08884571, cg00011225, cg00011225


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list (such as shown in the example listed below Table 25) for early detection of testicular cancer for specifying the origin of the cancer as testicular cancer and discriminating from other 10 common solid tumor cancers using plasma CF DNA or other body fluid CF DNA.










TABLE 25





Testicular_detect_spec





















cg14531093
cg11777290
cg13283877
cg03966099
cg26939375
cg26297530


cg16157016
cg14002772
cg09890687
cg05134918
cg09719850
cg06456125


cg17978367
cg20100671
cg26627956
cg19789755
cg14393609
cg21634331


cg02039634
cg25159927
cg01895439










Subset for detect and spec:


cg14531093, cg25159927


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs in the list below or a short subset of this list such (as shown in the example listed below Table 26) for early detection of one of 13 most common solid tumors using plasma CF DNA or other body fluid CF DNA.









TABLE 26





Pan-cancer_detect




















cg01423964
cg04223424
cg06530490
cg06543087
cg07900968
cg08406370


cg08848774
cg09734791
cg10723962
cg11017065
cg15759056
cg15760257


cg16405026
cg17228900
cg21277995
cg22286978
cg23141355
cg24427504


cg25024074
cg27420520










Subset for detect:


cg10723962, cg15759056, cg24427504, cg25024074


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs delineated by the BCD method on genome wide DNA methylation data as shown in Table 27 (or a short subset of this combination as shown below Table 27) for early detection of blood cancers such as AML, CLL, etc. using white blood cells, plasma CF DNA or other body fluid CF DNA.









TABLE 27





AML_detect-spec




















cg00594866
cg05608159
cg18658397
cg18780412
cg20430388
cg22828045


cg25375340










Subset for detect-spec:


cg18658397, cg18780412, cg20439288, cg22828045, cg25375340


In other embodiments, the polygenic DNA methylation markers are a combination of CG IDs shown in the list below (or a short subset of this list shown in the example listed below Table 28) for early detection of Melanoma for specifying the origin of the cancer as melanoma and discriminating from other 16 common solid tumor cancers using plasma CF DNA or other body fluid CF DNA.









TABLE 28





Melanoma_detect-spec




















cg00325866
cg01228636
cg04616691
cg04824711
cg05569109
cg06019853


cg08611163
cg10830649
cg12593303
cg15307891
cg18866529
cg19530886


cg19634213
cg20146030
cg21755725
cg22280705
cg24217704
cg24678095


cg26127345
cg27084903










Subset for detect-spec:


cg15307891, cg18866529, cg27084903


In another aspect of the inventive subject matter, there is provided a kit and a process for detecting cancer, comprising means and reagents for detecting DNA methylation measurements of polygenic DNA methylation markers.


In one embodiment, a kit is provided for detecting hepatocellular carcinoma comprising means and reagents for DNA methylation measurements of the CG IDs of table 1 and 2.


In another embodiment, a kit is provided for detecting lung cancer comprising means and reagents for DNA methylation measurements of the CG IDs of table 3 and 4.


In another embodiment, a kit is provided for detecting prostate cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 5.


In another embodiment, a kit is provided for detecting breast cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 6 and 7.


In another embodiment, a kit is provided for detecting colorectal cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 8.


In another embodiment, a kit is provided for detecting pancreatic cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 9 and 10.


In yet another embodiment, a kit is provided for detecting brain cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 11.


In another embodiment, a kit is provided for detecting gastric cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 12 and 13.


In another embodiment, a kit is provided for detecting ovarian cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 14 and 15.


In another embodiment, a kit is provided for detecting cervical cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 16 and 17.


In another embodiment, a kit is provided for detecting head and neck squamous carcinoma (HNSC) comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 18 and 19.


In another embodiment, a kit is provided for detecting esophageal carcinoma comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 20 and 21.


In another embodiment, a kit is provided for detecting bladder cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 22 and 23.


In another embodiment, a kit is provided for detecting renal cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 24.


In another embodiment, a kit is provided for detecting testicular cancer comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 25


In other embodiments, a kit is provided for detecting one of 13 common cancers (bladder, brain, breast, cervical, colorectal, esophageal, HNSC, HCC (liver), lung, ovarian, pancreatic, prostate, stomach) comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 26.


In another embodiment, a kit is provided for detecting blood cancers such as AML and CLL comprising means and reagents for detecting DNA methylation measurements of the CG IDs detected by the BCD method that are specific for different subtypes of blood cancer Table 27


In another embodiment, a kit is provided for detecting melanoma comprising means and reagents for detecting DNA methylation measurements of the CG IDs of table 28.


In another embodiment, DNA pyrosequencing methylation assays are used for predicting HCC in body fluids such as plasma CF DNA by using CG IDs listed above, for example by using the below disclosed primers and standard conditions of pyrosequencing reactions:











cg02012576



Forward:



GGTAGTTAGGAAGTTTAGAGGTTGTAGTA






Reverse (biotinylated):



ACCACTACCCCAACCCAACCCTA






Sequence:



GGTTTTAGGATGTTTG






cg03768777 (VASH2)



Forward:



AGAATAATATTAGAGAATGGGATATGGAA






Reverse (biotinylated):



ACAACTCCAAAATCCTACCT






Sequence:



GAATGGGATATGGAATGA






cg05739190 (CCNJ)



Forward:



GTTTAGGAGTTGGGTTTTAGTTGAG






Reverse (biotinylated):



ACCCCACCCTAACTCCCTTACC






Sequence:



TGGGTTTTAGTTGAGG






cg24804544 (GRID2IP)



Forward(biotinylated):



GGGTAGGGGAGGGTTTTGAAATA






Reverse:



TAACCCCCCCTCCAACCTCATTC






Sequence:



CACCCAACTTCTCAAT






The specificity of the tissue of origin of the cancer is determined by measuring the DNA methylation of the following CGID cg02012576 (HPX)











Forward(biotinylated):



ATTTTTATGGGTATTAGTTTTAGGGAGAA






Reverse (biotinylated):



CCAAAACTATCCTATAACCTCTACAACTCA






Sequence:



ACCATTACCACCCCT






In another embodiment, a polygenic multiplexed amplicon bisulfite sequencing DNA methylation assay is used for predicting cancer in body fluids such as plasma CF DNA by using CG IDs listed above. For example, predicting prostate cancer using the below disclosed primers and standard conditions that involve bisulfite conversion, sequential amplification with target specific primers (PCR 1) followed by barcoding primers (PCR 2) and multiplexed sequencing in a single next generation MiSeq sequencer (Illumina), demultiplexing using Illumina software, data extraction and quantification of methylation using standard methods for methylation analysis such as Methylkit, followed by calculation of the weighted DNA methylation score and prediction of cancer from a small volume of body fluids such as plasma, saliva or urine.


The steps to detect prostate cancer the first PCR is performed as follows:


For CGID cg02879662











Forward primer:



5′ACACTCTTTCCCTACACgACgCTCTTCCgA






TCTNNNNNGGTAGGAGTTTTGGGAATTGG3′






Reverse primer:



5′gTgACTggAgTTCAgACgTgTgCTCT






TCCgATCTCCACCCCTACAATCCCTAA3′






For CGID cg16232979



Forward primer:



5′ACACTCTTTCCCTACACgACgCTCTTCCgAT






CTNNNNNYGGTTTYGGGTTTYGTATT3′






Reverse primer:



5′gTgACTggAgTTCAgACgTgTgCTCTTCCgA






TCTACRCAAAAATATAAATCRACRATC3′







To test that the cancer specifically originates in the prostate the first PCR is performed as follows:











For CGID: cg14041701 and cg14498227



Forward primer:



5′ACACTCTTTCCCTACACgACgCTCTTCCgATCTNNNNN






GTTTTGYGTTTYGGATTTGGGTT3′






Reverse primer:



5′gTgACTggAgTTCAgACgTgTgCTCTTCCgATCTCATAAA






CAACACCTTTAAATAAACACTAAA3′







To barcode the samples, use a second PCR reaction with the following primers:











Forward primer:



5′AATgATACggCgACCACCgAgATCT






ACACTCTTTCCCTACACgAC3′






Barcoding primer (reverse):



5′CAAgCAgAAgACggCATACgAgATAGTCAT






CGgTgACTggAgTTCAgACgTg3′






(red bases are the index; 200



variations of this index are used)






In other embodiments, Receiver operating characteristics (ROC) assays are used for detecting cancer by defining a threshold value between cancer and normal using weighted DNA methylation measurements of CG IDs. Samples above/below threshold will be classified as cancer. For example, CGIDs listed above for detecting HCC:



















cg02012576
cg03768777
cg05739190
cg24804544.









In another embodiment, hierarchical Clustering analysis assays are used for predicting cancer by using measurements of methylation of CG IDs listed above.


In another aspect of the inventive subject matter, methods for identifying DNA methylation markers for detecting cancer and other disease comprise the step of performing statistical analysis with the “binary-categorical differentiation (BCD)” method previously disclosed regarding DNA methylation measurements obtained from clinical samples.


In another embodiment, the method includes performing statistical analysis and the “binary-categorical differentiation (BCD)” method on DNA methylation measurements obtained from samples, with DNA methylation measurements obtained by performing Illumina Beadchip 450K or EPIC array of DNA extracted from at least one sample.


In another embodiment, the DNA methylation measurements are obtained by performing DNA pyrosequencing of DNA extracted from a sample followed by mass spectrometry based (Epityper™), PCR based methylation assays and targeted amplification of a region spanning the target CG IDs disclosed herein from bisulfite converted DNA followed by barcoding in a second set of amplification and indexed-multiplexed sequencing on an Illumina next generation sequencer.


In other embodiments, the statistical analysis includes Receiver operating characteristics (ROC) assays.


In other embodiments, the statistical analysis includes hierarchical clustering analysis assays.


Definitions

As used herein, the term “CG” refers to a di-nucleotide sequence in DNA containing cytosine and guanosine bases. These di-nucleotide sequences could become methylated in human and other animals' DNA. The CG ID reveals its position in the human genome as defined by the Illumina 450K manifest (The annotation of the CGs listed herein is publicly available at https://bioconductor.org/packages/release/data/annotation/html/IlluminaHumanMethylation405_db.html and installed as an R package IlluminaHumanMethylation450k.db IlluminaHumanMethlation450k.db: Illumina Human Methylation 450 k annotation data. R package version 2.0.9.).


As used herein, the term “beta-value” refers to an estimation of methylation level at a CG ID position derived by normalization and quantification of Illumina 450K arrays using the ratio of intensities between methylated and unmethylated probes using the formula beta value=methylated C intensity/(methylated C intensity+unmethylated C intensity) between 0 and 1 with 0 being fully unmethylated and 1 being fully methylated.


As used herein, the term “penalized regression” refers to a statistical method aimed at identifying the smallest number of predictors required to predict an outcome out of a larger list of biomarkers as implemented for example in the R statistical package “penalized” as described in Goeman, J. J., L1 penalized estimation in the Cox proportional hazards model. Biometrical Journal 52(1), 70-84.


As used herein, the term “clustering” refers to the grouping of a set of objects in such a way that objects in the same group (called a cluster) are more similar (in some sense or another) to each other than to those in other groups (clusters).


As used herein, the term “Hierarchical clustering” refers to a statistical method that builds a hierarchy of “clusters” based on how similar (close) or dissimilar (distant) are the clusters from each other as described for example in Kaufman, L.; Rousseeuw, P. J. (1990). Finding Groups in Data: An Introduction to Cluster Analysis (1 ed.). New York: John Wiley. ISBN 0-471-87876-6.


As used herein, the term “Receiver operating characteristics (ROC) assay” refers to a statistical method that creates a graphical plot that illustrates the performance of a predictor. The true positive rate of prediction is plotted against the false positive rate at various threshold settings for the predictor (i.e. different % of methylation) as described for example in Hanley, James A.; McNeil, Barbara J. (1982). “The Meaning and Use of the Area under a Receiver Operating Characteristic (ROC) Curve”. Radiology 143 (1): 29-36.


As used herein, the term “multivariate or polygenic linear regression” refers to a statistical method that estimates the relationship between multiple “independent variables” or “predictors” such as percentage of methylation in CG IDs, and a “dependent variable” such as cancer. This method determines the “weight” or coefficient of each CG IDs in predicting the “outcome” (dependent variable such as cancer) when several “independent variables” such as CG IDs are included in the model.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1. shows a shortlist of completely unmethylated sites across blood samples and normal tissues in hundreds of individuals. Illustration A. shows CG IDs across 17 tissues that are unmethylated in all individuals (<0.1) in Illumina450K genome wide methylation arrays (GSE50192) were overlapped with unmethylated CG IDs in genome wide DNA methylation arrays of blood samples from 312 individuals (GSE61496) to generate a list of 33477 CG IDs. B illustrates shortlisting the most robustly unmethylated CG IDs the list of 33477 CG IDs from A was overlapped with unmethylated CG IDs in DNA methylation arrays of blood samples from 656 individuals (females and male) aged from 19 to 101 years (GSE40279). Combined, these analyses generated a list of high confidence 28754 CG IDs that are unmethylated across tissues and blood samples of many individuals across all age spans. These 28754 positions were used for discovery of sites that are categorically methylated in cancer but not in other tissues using the “binary-categorical differentiation (BCD)” method disclosed by the present inventive subject matter.



FIG. 2 is an illustration showing the Lack of Tissue specificity of current Circulating DNA markers for HCC. The illustrated heatmap shows 10 CG IDs shortlisted in Xu et al., (Xu et al., 2017) as biomarkers of HCC and methylation levels for these sites in other normal tissues. Several of the CG IDs proposed as specific biomarkers of HCC are methylated in other tissues are well and show varying levels of methylation in blood DNA. (blue 0 methylation dark red 100% methylation)



FIG. 3 is an illustration showing the specificity of HCC DNA methylation markers discovered using the BCD method for cancer DNA. The illustrated heatmap shows 4 CG IDs selected as HCC DNA methylation markers by the BCD method described herein. Methylation levels are categorically different between cancer (HCC) and normal tissues and blood, whereby the sites are unmethylated in all individuals in blood and other tissues and measurably methylated in HCC.



FIG. 4 is an illustration showing the lack of cancer tissue of origin specificity of current DNA methylation markers for colorectal cancer and comparison with the “detect-spec” method according to embodiments of the inventive subject matter. Illustration A shows the CG sites in Sept9 gene as included in the “Epi-colon” CF DNA methylation marker for colorectal cancer (sold by Epigenomics Inc.) which can be used to detect many other cancers utilizing methylation data from the TCGA collection of cancer DNA methylation data and thus lack specificity for colorectal cancer (HKG-Colon (HKG-epiCRC), blue). The markers disclosed in the present inventive subject matter for detection of colorectal cancer (Table 9) discovered using the BCD method (HKG-Colon orange) (Table 10) are highly specific for colorectal cancer when tested against other common solid tumors cancers. Illustrations B and C are scatter-plots of DNA methylation values for tumor DNA from different individuals with different cancers using either the HKG-Colon (HKG-epiCRC) (B) or Epi-Colon (C) DNA methylation markers. Of note are the tight and categorical differences in DNA methylation between colorectal cancer and other cancers using the HKG-epiCRC markers (B) versus the scattered heterogenous profile of Epi-Colon markers (C).



FIG. 5 is an illustration showing a discovery of a polygenic DNA methylation marker for early detection of liver cancer (HCC). Illustration A shows a table which lists the source and number of patients whose methylation data was used for the discovery of a set of 4 CGIDs for detection of HCC according to an embodiment using the BCD method (Table 1) and CG IDs for determining the specific cancer of origin (Table 2). Illustration B at the bottom left panel of FIG. 5 (Detect) shows the combined methylation score for these CG IDs (Table 1) for each of the tested people listed from 1-145 (79 normal and 66 HCC). The polygenic score categorically differentiates between people with HCC and normal liver tissue. Illustration C at the bottom right panel shows the methylation score for the 1 CGID detecting the specific tumor origin (Table 2) using data on 8 different tumors (Table 2). The markers categorically differentiate between cancers from other origins and HCC.



FIG. 6 is an illustration of a validation of a polygenic DNA methylation marker for HCC (spec) using DNA methylation data from GSE76269 (n=227). Illustration A is a ROC plot showing area under the curve for the HCC DNA methylation markers using 227 liver cancer patient DNA methylation data and 10 normal. Illustration B of FIG. 6 shows the sensitivity, specificity and accuracy of HCC detection. Illustration C shows the prediction rate of detection of HCC in the validation dataset.



FIG. 7 is an illustration of a validation of the polygenic HKG-epiLiver-detect and spec markers accuracy and specificity for HCC versus other cancers in TCGA methylation data (n=4166). Illustration A of FIG. 7 shows a detection rate of the HKG-Liver detect/spec markers DNA methylation data of patients with different cancers. Note the almost perfect specificity for HCC. B. ROC plot of the HKG-Liver-detect markers specificity and sensitivity for HCC in 4166 patient DNA methylation data from TCGA. C. Sensitivity and specificity to HCC versus cancers from other origins.



FIG. 8 is an illustration of a discovery of a polygenic DNA methylation marker for lung cancer. Illustration A of FIG. 8 is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of lung cancer disclosed in embodiments using the BCD method (Table 3) as well as CG IDs for determining the specific cancer tissue of origin (Table 4). Illustration B at the bottom left panel of FIG. 8 (Detect) shows the combined methylation score for these CG IDs (Table 3) for each of the tested people listed from 1-20 (10 normal and 10 lung cancer). The polygenic score categorically differentiates between people with lung cancer and normal tissue. Illustration C at the bottom right panel of FIG. 8 shows the methylation score for the CGIDs (Table 4) detecting the specific tumor origin using data from people who have 8 different tumors (n=80). In these embodiments, the markers categorically differentiate between cancers from other origins and lung cancer.



FIG. 9 is an illustration of the validation of the polygenic HKG-epiLung-detect and spec markers accuracy and specificity for HCC versus other cancers in TCGA methylation data (n=4166). Illustration A of FIG. 9 shows a detection rate of the HKG-epiLung detect/spec markers using DNA methylation data of patients with different cancers. Of note is the specificity for lung cancer. Illustration B of FIG. 9 shows a ROC plot of the HKG-Lung-detect markers specificity and sensitivity for lung cancer on 4166 patient DNA methylation data from TCGA. Illustration C of FIG. 9 shows a sensitivity and specificity to lung cancer versus cancers from other origins.



FIG. 10 is an illustration of the discovery of a polygenic DNA methylation marker for prostate cancer. Illustration A of FIG. 10 is a table listing the source and number of a patient whose methylation data was used for discovery of a set of CGIDs for detection of prostate cancer disclosed in embodiments using the BCD method (Table 5) and CGIDs for determining the specific cancer tissue of origin (Table 6). Illustration B at the bottom left panel of FIG. 10 (Detect) shows the combined methylation score for these CGIDs (Table 5) for each of the tested people listed from 1-15 (5 normal and 10 prostate cancer). The polygenic score categorically differentiates between people with prostate cancer and normal. Illustration C at the bottom right panel of FIG. 10 shows the methylation score for the CGs detecting the specific tumor tissue of origin (Table 6) using data from people who have 8 different tumors (n=80). In these embodiments, the markers categorically differentiate between cancers from other origins and prostate cancer.



FIG. 11 is an illustration of a validation of the polygenic HKG-epiProstate-detect and spec markers accuracy and specificity for prostate versus other cancers in TCGA methylation data (n=4166). Illustration A of FIG. 11 shows the detection rate of the HKG-Prostate detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for prostate cancer. Illustration B of FIG. 11 is a ROC plot of the HKG-Prostate-detect markers specificity and sensitivity for lung cancer using DNA methylation data from 4166 patients in TCGA. Illustration C of FIG. 11 shows the sensitivity and specificity to prostate versus cancers from other origins.



FIG. 12 is an illustration of a discovery of a polygenic DNA methylation marker for breast cancer. Illustration A of FIG. 12 is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGs for detection of breast cancer disclosed in embodiments using the BCD method (Table 7) and CGIDs for determining the specific cancer of origin (Table 8). Illustration B at the bottom left panel of FIG. 12 (Detect) shows the combined methylation score for these CGIDs (Table 7) for each of the tested people listed from 1-27 (17 normal and 10 breast cancer). The polygenic score categorically differentiates between people with breast cancer and normal breast tissue. Illustration C at the bottom right panel of FIG. 12 shows the methylation score for the CGIDs detecting the specific origin of the tumor (Table 8) using data from people who have 8 different tumors (n=80). In these embodiments, the markers categorically differentiate between cancers from other origins and breast cancer.



FIG. 13 is an illustration of a HKG-epiBreast-detect polygenic DNA methylation marker detect noninvasive as well as invasive breast cancer in validation cohort GSE60185 (n=285). Illustration A of FIG. 13 is a ROC plot showing area under the curve for the breast cancer polygenic DNA methylation marker using 239 breast cancer patient DNA methylation data, 17 mamoplastic surgery patients with no breast cancer and 29 adjacent tissues. The sensitivity, specificity and accuracy for all breast cancer are listed in B and the prediction rate of DCIS (ductal carcinoma in situ), invasive and mixed breast cancer samples are shown in Illustration C of FIG. 13. Of note is that the breast cancer markers detect even very early breast cancers (DCIS).



FIG. 14 is an illustration of a validation of the polygenic HKG-epiBreast-detect and spec markers accuracy and specificity for breast versus other cancers in TCGA methylation data (n=4166). Illustration A of FIG. 14 shows a detection rate of the HKG-epiBreast detect/spec markers in DNA methylation data from patients with different cancers. Of note is the specificity for breast cancer. Illustration B of FIG. 14 is a ROC plot of the HKG-Breast-detect markers specificity and sensitivity for detecting breast cancer using DNA methylation data from 4166 patients in TCGA. Illustration C of FIG. 14 shows the sensitivity and specificity to breast versus cancers from other origins.



FIG. 15 is an illustration of a discovery of a polygenic DNA methylation marker for colorectal cancer (CRC). Illustration A of FIG. 15 is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of colorectal cancer disclosed in embodiments using the BCD method (Table 9) and CGIDs for determining the specific cancer of origin (Table 10). Illustration B at the bottom left panel of FIG. 15 (Detect) shows the combined methylation score for these CGIDs for each of the tested people listed from 1-75 (25 normal and 50 colorectal cancer). The polygenic score categorically differentiates between people with cancer and normal tissue. Illustration C at the bottom right panel of FIG. 15 shows the methylation score for the CGIDs detecting the specific origin of the tumor using DNA methylation data from people who have 8 different tumors (n=80). In these embodiments, the markers categorically differentiate between cancers from other origins and colorectal cancer.



FIG. 16 is an illustration validation of the polygenic HKG-epiCRC-detect and spec markers accuracy and specificity for CRC versus other cancers using TCGA DNA methylation data set (n=4166). Illustration A of FIG. 16 is a detection rate of the HKG-epiCRC detect/spec markers using DNA methylation data of patients with different cancers. Of note is the specificity for colorectal cancer. Illustration B of FIG. 16 is a ROC plot of the HKG-epiColon-detect markers specificity and sensitivity for colorectal cancer using DNA methylation data from 4166 patients in TCGA. Illustration C of FIG. 16 shows sensitivity and specificity to colorectal cancer versus cancers from other origins.



FIG. 17 illustrates a discovery of a polygenic DNA methylation marker for pancreatic cancer. Illustration A of FIG. 17 is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of pancreatic cancer disclosed in the present invention using the BCD method (Table 11) and CGIDs for determining the specific cancer of origin (Table 12). Illustration B at the bottom left panel of FIG. 17 (Detect) shows the combined methylation score for these CGIDs (Table 11) for each of the tested people listed from 1-32 (12 normal and 20 pancreatic cancer). The polygenic score categorically differentiates between people with pancreatic cancer and normal tissue. Illustration C at the bottom right panel of FIG. 17 shows the methylation score for the CGIDs detecting the specific origin of the tumor (Table 12) using data from people who have 10 different tumors (n=100). In these embodiments, the markers categorically differentiate between cancers from other origins and pancreatic cancer.



FIG. 18 is an illustration of a validation of the polygenic HKG-epiPancreas-detect and spec markers accuracy and specificity for pancreatic cancer versus other cancers in TCGA methylation data (n=4854). Illustration A of FIG. 18 is a detection rate of the HKG-epiPancreas detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for pancreatic cancer. Illustration B of FIG. 18 is a ROC plot of the HKG-epiPancreas-detect markers specificity and sensitivity for pancreatic cancer using DNA methylation data for 4854 patients in TCGA. Illustration C is the sensitivity and specificity to pancreatic cancer versus cancers from other origins.



FIG. 19 is an illustration of a discovery of a polygenic DNA methylation marker for brain cancer (glioblastoma). Illustration A of FIG. 19 is a table listing the source and number of patient whose methylation data was used for discovery of a set of CGIDs for detection of brain cancer disclosed in the present invention using the BCD method (Table 13) and CGIDs for determining the specific origin of the cancer (Table 13). Illustration B at the bottom left panel (Detect/spec) shows the combined methylation score for these CGIDs (Table 13) for each of the tested people listed from 1-16 (6 normal and 10 brain cancer). The polygenic score categorically differentiates between people with brain cancer, 110 other cancers, normal tissues.



FIG. 20 is an illustration of a validation of the polygenic HKG-epiBrain-detect and spec markers accuracy and specificity for breast versus other cancers in TCGA methylation data (n=4854). Illustration A is a detection rate of the HKG-epiBrain detect/spec markers using DNA methylation data from patients with different cancers. Note the specificity for brain cancer. Illustration B is a ROC plot of the specificity and sensitivity of the HKG-epiBrain-detect markers for brain cancer using DNA methylation data from 4854 patients in TCGA. Illustration C shows a sensitivity and specificity to brain cancer in TCGA data set (n=695).



FIG. 21 is an illustration of a discovery of a polygenic DNA methylation marker for detection of gastric (stomach) cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of gastric cancer disclosed in the present invention using the BCD method (Table 14) and CGIDs for determining the specific origin of the cancer (Table 15). Illustration B at the bottom left panel of FIG. 21 (Detect) shows the combined methylation score for these CGIDs (Table 14) for each of the tested people listed from 1-28 (14 normal and 20 gastric cancer). The polygenic score categorically differentiates between people with gastric cancer and normal tissue. Illustration C at the bottom right panel of FIG. 21 (Spec) shows the polygenic methylation scores for people who have 10 different tumors (n=100). In these embodiments, the markers categorically differentiate between cancers from other origins and gastric cancer.



FIG. 22 is an illustration of a validation of the polygenic HKG-Stomach-detect and spec markers accuracy and specificity for gastric cancer versus other cancers in TCGA methylation data (n=4817). Illustration A is a detection rate of the HKG-epiStomach detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for gastric cancer. Illustration B is a ROC plot specificity and sensitivity of the HKG-epiStomach-detect spec 1 markers for stomach (gastric cancer) using DNA methylation data from 4420 patients in TCGA. Illustration C is a ROC plot of specificity and sensitivity the HKG-epiStomach-spec 1 markers for gastric cancer using DNA methylation data from 4854 patients in TCGA. Of note is that there is a significant cross reactivity with colorectal and esophageal cancer which attests to a shared origin.



FIG. 23 is an illustration of a discovery of a polygenic DNA methylation marker for ovarian cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of ovarian cancer disclosed in the present invention using the BCD method (Table 16) and CGIDs for determining the specific cancer of origin (Table 17). Illustration B at the bottom left panel of FIG. 23 (Detect) shows the combined methylation score for these CGIDs for each of the tested people listed from 1-15 (5 normal and 10 ovarian cancer). The polygenic score categorically differentiates between people with ovarian cancer and normal tissue. Illustration C at the bottom right panel of FIG. 23 shows the methylation score for the CGIDs detecting the specific tumor origin using data from people who have 11 different tumors (n=110). In these embodiments, the markers categorically differentiate between cancers from other origins and ovarian cancer.



FIG. 24 is an illustration of a validation of the polygenic HKG-epiOvarian-detect and spec markers accuracy and specificity for cervical versus other cancers in TCGA methylation data (n=6522). Illustration A is a detection rate of the HKG-epiOvarian detect/spec markers using DNA methylation data from patients with different cancers. Note the specificity for ovarian cancer. Illustration B is a ROC plot specificity and sensitivity of the HKG-epiOvarian-detect and spec markers for ovarian cancer on DNA methylation data from 4723 patients in TCGA. Illustration C shows the sensitivity and specificity to ovarian cancer.



FIG. 25 is an illustration of a discovery of a polygenic DNA methylation marker for cervical cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of cervical cancer disclosed in the present invention using the BCD method (Table 18) and CGs for determining the specific cancer of origin (Table 19). Illustration B at the bottom left panel of FIG. 25 (Detect) shows the combined methylation score for these CGIDs (Table 18) for each of the tested people listed from 1-30 (20 normal and 10 ovarian cancer). The polygenic score categorically differentiates between cervical cancer and normal tissue. Illustration C at the bottom right panel of FIG. 25 shows the methylation score for the CG IDs detecting the specific origin of the tumor (Table 19) using data from people who have 8 different tumors (n=80). In these embodiments, the markers categorically differentiate between cancers from other origins and cervical cancer, note however some measurable detection of colorectal cancer.



FIG. 26 is an illustration of a validation of the polygenic HKG-Cervix-detect and spec markers accuracy and specificity for cervix versus other cancers in TCGA methylation data (n=6522). Illustration A shows a detection rate of the HKG-Cervix detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for cervical cancer. Illustration B is a ROC plot of specificity and sensitivity of the HKG-Cervix-detect spec markers for cervical cancer using DNA methylation data from 4420 patients in TCGA. Illustration C shows a sensitivity and specificity to cervical cancer.



FIG. 27 is an illustration of a discovery of a polygenic DNA methylation marker for Head and Neck squamous cell Carcinoma (HNSC). Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CG IDs for detection of HNSC disclosed in the present invention using the BCD method (Table 20) and CGs for determining the specific cancer of origin (Table 21). Illustration B at the bottom left panel of FIG. 27 shows the combined methylation score for these CG IDs (Table 20) for each of the tested people listed from 1-140 (10 cancer, 10 normal and 120 other cancers). Illustration C shows the polygenic score which categorically differentiates between HNSC and normal tissue samples in the embodiments as well as categorically differentiating between cancers from other origins and HNSC.



FIG. 28 is an illustration of a validation of the polygenic HKG-epiHNSC-detect/spec markers accuracy and specificity for HNSC versus other cancers in TCGA methylation data (n=4166). Illustration A is a detection rate of the HKG-epiHNSC detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for HNSC. Illustration B is a ROC plot of the specificity and sensitivity of HKG-epiHNSC-detect markers for HNSC on DNA methylation data from 4166 patients in TCGA. Illustration C shows the sensitivity and specificity for HNSC versus cancers from other origins.



FIG. 29 is an illustration of a discovery of a polygenic DNA methylation marker for esophageal cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of esophageal cancer disclosed in embodiments using the BCD method (Table 22) and CGIDs for determining the specific origin of the cancer (Table 23). Illustration B at the bottom left panel of FIG. 29 shows the combined methylation score for these CGIDs (Table 22) for each of the tested people listed from 1-15 (6 normal, 10 cancer). Illustration C shows the polygenic score categorically differentiating between esophageal cancer and normal tissue in the embodiments as well as categorically differentiating between cancers from other origins and esophageal cancer listed from 1-220 (20 cancer, 190 other cancer and 10 healthy blood).



FIG. 30 is an illustration of a validation of the polygenic HKG-epiEsophageal—detect/spec markers accuracy and specificity for esophageal cancer versus other cancers in TCGA methylation data (n=7102). Illustration A shows a detection rate of the HKG-epiEsophageal detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for esophageal cancer. Illustration B is a ROC plot of the specificity and sensitivity of the HKG-epiEsophageal-detect markers for HNSC on 4166 patient DNA methylation data in TCGA. Illustration C shows sensitivity and specificity to esophageal cancer versus cancers from other origins.



FIG. 31 is an illustration of a discovery of a polygenic DNA methylation marker for bladder cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of bladder cancer disclosed in embodiments using the BCD method (Table 24) and CGIDs for determining the specific origin of the cancer (Table 25). Illustration B at the bottom left panel of FIG. 31 (Detect) shows the combined methylation score for these CGIDs (Table 24) for each of the tested people listed from 1-15 (5 normal and 10 bladder cancer). Illustration C at the bottom right panel of FIG. 31 shows the methylation score for the CGIDs (Table 25) detecting the specific origin of the tumor using data from people who have 13 different tumors (n=130). In these embodiments, the markers differentiate between cancers from other origins and bladder cancer. Also of note are some measurable detection of colorectal cancer with these markers.



FIG. 32 is an illustration of a validation of accuracy and specificity of the polygenic HKG-epiBladder-detect and spec markers for bladder cancer versus other cancers in TCGA (n=4723). Illustration A shows a detection rate of the HKG-epiBladder spec (A) and detect markers (B) on DNA methylation data of patients with different cancers (A) and bladder cancer (B). Illustration C is a ROC plot of the specificity and sensitivity of HKG-epiBladder spec markers for bladder cancer using DNA methylation data from 4420 patients in TCGA. Illustration D is a ROC plot of the specificity and sensitivity of the HKG-epiBladder detect markers for bladder cancer (n=440).



FIG. 33 is an illustration of a discovery of a polygenic DNA methylation marker for kidney cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of kidney cancer disclosed in embodiments using the BCD (hypo) method and for determining the specific origin of the cancer (Table 26). Illustration B at the bottom left panel of FIG. 33 (Detect/spec) shows the combined methylation score for these CGIDs (Table 26) for each of the tested people listed from 1-226 (180 other cancers, 10 healthy blood, 6 normal kidney, 30 renal cancer). In these embodiments, the polygenic score categorically differentiates between kidney cancer, other cancers and normal blood.



FIG. 34 is an illustration of a validation of the accuracy and specificity of polygenic HKG-epiKidney-detect and spec markers for kidney cancer versus other cancers and normal tissues using TCGA DNA methylation data (n=7102). Illustration A is the detection rate of the HKG-epiKidney detect/spec markers using DNA methylation data from different cancers. Of note is the specificity for kidney cancer. Illustration B is a ROC plot of the specificity and sensitivity of HKG-Cervix-detect spec markers for renal cancer using DNA methylation data from 6367 cancers in TCGA. Illustration C is a sensitivity and specificity to renal (kidney) cancer. Of additional note is a crossover with brain, HCC and testicular cancer.



FIG. 35 is an illustration of a discovery of a polygenic DNA methylation marker for testicular cancer. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of testicular cancer disclosed in embodiments using the BCD (hypo) method and for determining the specific cancer of origin (Table 27). Illustration B at the bottom left panel of FIG. 35 (Detect/spec) shows the combined methylation score for these CG IDs (Table 27) for each of the tested people listed from 1-226 (10 testicular cancer, 180 other cancers, 10 normal blood). In these embodiments, the polygenic score categorically differentiates between testicular cancer and normal blood and other cancers.



FIG. 36 is an illustration of a validation of the accuracy and specificity of the polygenic HKG-epiTestis-detect and spec markers for testicular cancer versus other normal tissues and cancers in TCGA methylation data (n=7102). Illustration A shows the detection rate of the HKG-epiTesstis detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for Testis cancer. Illustration B is a ROC plot of the specificity and sensitivity of HKG-epiTestis-detect spec markers for testicular cancer using DNA methylation data from 6367 patients in TCGA. Illustration C is the sensitivity and specificity to testicular cancer.



FIG. 37 is an illustration of a discovery of a Pan cancer polygenic DNA methylation marker for 13 common cancers. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of 13 common cancers (Table 28) (bladder cancer, brain cancer, breast cancer, cervical cancer, colorectal cancer CRC, esophageal cancer, liver cancer, lung cancer. ovarian cancer, pancreatic cancer, prostate cancer, and stomach cancer) disclosed in embodiments using the BCD method (Table 28). Illustration B shows the combined methylation score for these CGIDs for each of the tested people listed from 1-310 (180 cancer and 10 normal). In these embodiments, the polygenic score differentiates between cancers and normal tissue.



FIG. 38 is an illustration of a validation of the polygenic HKG epiPancancer markers accuracy and specificity in TCGA methylation data (n=7102). Illustration A shows Methylation scores calculated using the epiPancancer polygenic DNA methylation markers in patients with 13 different cancers using TGCA data. Illustration B is a ROC plot of the specificity and sensitivity of HKG-epiPancancer detect and spec markers using DNA methylation data from for all cancers from 4878 patients in TCGA. Illustration C is a ROC plot of the epiPancancer polygenic markers describing specificity and sensitivity for detection of 13 common cancers. Illustration D shows the overall sensitivity and specificity of the pan cancer markers for detecting cancer. In these embodiments, one or more colors are used, for example orange (weighted methylation score) and blue (detection of one BCD marker per sample is scored as a positive cancer).



FIG. 39 is an illustration of a discovery of polygenic DNA methylation marker for Melanoma. Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of melanoma (Table 45) disclosed in embodiments using the BCD method (Table 45). Illustration B is the combined methylation score for these CGIDs for each of the tested people listed from 1-220 (other cancers and normal blood) and 10 patients with melanoma. In these embodiments, the polygenic score differentiates between melanoma, other cancers and normal tissue.



FIG. 40 is an illustration of a validation of the accuracy and specificity of the polygenic HKG-epiMelanoma-detect and spec markers for melanoma versus other normal tissues and cancers in TCGA methylation data (n=7102). Illustration A shows the detection rate of the HKG-epiMelanoma detect/spec markers using DNA methylation data from patients with different cancers. Of note is the specificity for melanoma (with overlapping detection of liver cancer brain and cancer and prostate cancer). Illustration B is a ROC plot of the specificity and sensitivity of HKG-Melanoma-detect spec markers for melanoma using DNA methylation data from 6367 patients in TCGA. Illustration C shows the sensitivity and specificity to melanoma.



FIG. 41 is an illustration of a discovery of polygenic DNA methylation marker for blood cancers (Acute Myeloid Leukemia (AML). Illustration A is a table listing the source and number of patients whose methylation data was used for discovery of a set of CGIDs for detection of blood cancer AML (Table 46) disclosed embodiments using the BCD method (Table 46). Illustration B is the combined methylation score for these CGIDs for each of the tested people listed from 1-10 (normal blood) and 10 patients with AML. In these embodiments, the polygenic score differentiates between AML and normal blood.



FIG. 42 is an illustration of a validation of the accuracy and specificity of the polygenic HKG-epiAML-detect and spec markers for AML in GSE86409 (n=79) and in TCGA (n−140) versus normal blood in GSE40279 and GSE61496 (n=968). Illustration A shows a detection rate of the HKG-epiAML detect/spec markers using DNA methylation data from patients with AML and healthy blood. Of note is the specificity for melanoma (with overlapping detection of liver cancer brain and cancer and prostate cancer). Illustration B is a ROC plot of the specificity and sensitivity of HKG-AML-detect spec markers for AML using DNA methylation data from GSE86409 (n=79), TCGA (n-140) GSE40279 and GSE61496 (n=968). Illustration C shows the sensitivity and specificity to AML.



FIG. 43 is an illustration of a validation that the primers selected for detecting different cancers exhibit BCD properties ˜0 methylation in plasma derived from normal people (each sample is a mixture of plasma from normal patients). The first PCR1 reaction targeting the specific CGs was performed using sequence targeted primers. Following a second PCR, the amplified fragments were purified and subjected to next generation sequencing. DNA methylation was quantified in each of the indicated CG ID positions.



FIG. 44 is an illustration of a validation that the indicated primers selected for detecting different cancers exhibit BCD properties ˜0 methylation in plasma derived from normal people (each sample is a mixture of plasma from normal patients).



FIG. 45 is an illustration of a primer design for multiplex amplification and sequencing. The first PCR reaction targets the specific regions of interest, note PCR1 primer has complementary sequence to second PCR2 primers. The second set of primers introduces the index for each patient as well as the reverse and forward sequencing primers.



FIG. 46 is an illustration of an optimization of PCR conditions for detecting prostate cancer. A multiplex PCR1 reaction using varying primer concentrations as indicated DNA for the three markers of prostate cancer HIF3A 232 bp, TPM4 213 bp, and CTTN 199 bp is shown on the right panel.



FIG. 47. is an illustration of a bioinformatics workflow for determining DNA methylation levels. PCR2 products are combined, quantified and purified and subjected to next generation sequencing on a Miseq Illumina sequencer. Sequence is demultiplexed, FASTQ files are generated for each patient and analyzed with the workflow shown in the scheme. DNA methylation scores are calculated for each patient.





DETAILED DESCRIPTION

All illustrations of the drawings are for the purpose of describing selected embodiments and are not intended to limit the scope of the claimed subject matter.


Embodiment 1. “Discovery of Categorically Unmethylated CGIDs Across Hundreds of Individuals in Normal Tissues and Blood DNA

Cell free DNA originating in tumors is known to be found in body fluids such as plasma, urine and in feces. It is also established that DNA methylation profiles of CF tumor DNA are similar to tumor DNA (Dominguez-Vigil et al., 2018). A vast body of data has established that tumor DNA is differentially methylated compared to normal tissues (Luczak & Jagodzinski, 2006). Therefore, many groups have tried to delineate by logistic regressions CGID positions in DNA (CG IDs in the Illumina 450K manifest) that are differentially methylated between cancerous and its normal tissue of origin for example, liver cancer versus adjacent liver tissue. However, since these methods measure quantitative differences between cancer and untransformed tissue rather than categorical qualitative differences, these quantitative differences between tumor and normal tissue would be diluted and erased by CF DNA from normal tissue, leading to false negatives and reduced sensitivity. In addition, other tissues that were not included in the analysis might have a DNA methylation profile similar to tumor DNA and since most studies only compare the tumor DNA to its untransformed counterparts and not to other tissues, this could lead to false positives. Varying and unpredictable quantities of DNA from different tissues have been detected in CF DNA (Breitbach et al., 2014) and thus the measured DNA methylation reflects a composite of unknown and unpredictable mixture of tissue DNA from different sources and tumor DNA. Thousands of tumor samples have been subjected to genome wide DNA methylation analysis using Illumina 450K arrays and are found in the public domain (TCGA). Examining the profiles of methylation of many normal tissues as well as cancer tissues, the inventors noticed that there is a significant group of CGs in the genome that are completely unmethylated in all normal tissues but methylated in DNA from tumors. A subset of these sites is unmethylated across numerous individuals whose DNA methylation was profiled in the public domain. The inventors also noticed that in many cancers these robustly unmethylated sites become methylated in cancer. Thus, creating a qualitative “categorical difference” between tumor DNA and all other DNA that might be found in blood. Using deep next generation sequencing even few methylated molecules could be easily identified on a background of completely unmethylated copies.


Data Bases; Illumina 450K DNA Methylation Data

We used publicly available data bases of normalized beta values of methylation for ˜450,000 CG across the human genome from a large number of individuals deposited either in the Gene expression Omnibus (GEO) https://www.ncbi.nlm.gov/geo/ or The Cancer Genome Atlas TCGA https://cancergenome.nih.gov/ public data bases. We used the following databases to derive the list of robustly unmethylated CG IDs in many normal tissues and blood DNA: GSE50192, GSE50192, GSE40279.


DNA from white blood cells is one of the main sources of CF DNA in plasma. The inventors first generated a list of 47981 CGIDS that are unmethylated in all individuals in 17 different somatic human tissues using Illumina 450K data in GSE50192 and the logical COUNTIF and IF functions in Excel:


NmCGIDx=COUNTIF (betaCGIDxn1:ni,“>0.1”)


umCGIDx=IF(NmCGIDx=0, TRUE, FALSE)


NmCGIDx=number of normal subjects that have the CGIDx methylated.


umCGIDx=unmethylated CGIDx in all subjects


betaCGIDx=the methylation values for a given CGIDx


x=any CGID on the Illumina 450 k array


n1=the first subject in the array,


ni=the last subject in the array.


The inventors then generated a list of 68260 unmethylated CGIDs (UMCGIDs) in blood DNA from 312 individuals using the same criteria. The inventors then overlapped the list of 47981 and 68260 CG IDS and obtained a list of 33477 CG IDs that are unmethylated in both blood and somatic tissues across all individuals (FIG. 1A). To increase the robustness of this list of unmethylated CG IDs the inventors delineated a list of 60,379 CG IDs unmethylated CGIDs in Illumina 450K arrays of whole blood DNA from 656 individuals males and females aged from 19 to 101 years (GSE40279). These are robustly unmethylated sites in blood that are sex and age independent across hundreds of individuals. This list of 60,379 CG IDs was overlapped with the list of 33,477 CG IDs that are unmethylated both in somatic tissues and blood to generate a final list of 28,754 CG IDs which were used for discovery of categorical methylation markers for cancer. This list includes CG ID positions that are robustly unmethylated across tissues and individuals.


To identify DNA methylation positions that are categorically different between cancer and normal tissues the inventors examined whether any of these 28754 CG IDs are methylated in different cancers. The inventors noticed following examination of tumor DNA methylation data that methylation of a subset of these 28754 CG IDs is common in tumor DNA from individual patients. However, not all individuals have the same position methylated. Thus, a combination of CG IDs is required to detect cancer with high specificity. The inventors therefore discovered a polygenic combination of CG IDs for detection of cancers.


The inventors used 10 to 50 DNA methylation profiles from the public domain from either TCGA or GEO as a “discovery set” to discover a polygenic set of CGIDs whose methylation state is “categorically” different between tumor and normal tissues that could detect cancer with highest sensitivity and specificity. These CGIDs were then tested on hundreds of TCGA and GEO tumor DNA methylation array data as a “validation set” to validate the sensitivity and specificity of the polygenic DNA methylation markers for detecting cancer as disclosed in Embodiment 2.


Embodiment 2: Binary-Categorical Differentiation (BCD)” Method for Detecting Cancer in Cell Free DNA

The following publicly available data bases of normalized beta values of methylation for ˜450,000 CGs (CG IDs) across the human genome were used to derive the list of cancer specific DNA methylation markers:









TABLE 29







liver cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Normal Liver
GSE61258
Discovery
79
Detect


HCC
TCGA
Discovery
50
Detect


HCC
TCGA
Discovery
10
Spec


Non-HCC cancers
TCGA
Discovery
80
Spec


HCC
GSE75041
Validatiotext missing or illegible when filed
66
Detect


HCC
GSE76269
Validatiotext missing or illegible when filed
227
Detect


HCC
TCGA
Validatiotext missing or illegible when filed
430
Detect


Bladder cancer
TCGA
Validatiotext missing or illegible when filed
439
Spec


Brain cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast cancer
TCGA
Validatiotext missing or illegible when filed
891
Spec


Cervix cancer
TCGA
Validatiotext missing or illegible when filed
312
Spec


CRC cancer
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA cancer
TCGA
Validatiotext missing or illegible when filed
202
Spec


Kidney cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec


Ovarian cancer
TCGA
Validatiotext missing or illegible when filed
10
Spec


PANC cancer
TCGA
Validatiotext missing or illegible when filed
195
Spec


PRAD cancer
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach cancer
TCGA
Validatiotext missing or illegible when filed
397
Spec


Testis cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal Liver
GSE76269
Validatiotext missing or illegible when filed
10
Spec


Normal Liver
GSE69852
Validatiotext missing or illegible when filed
6
Spec


Normal Liver
GSE75041
Validatiotext missing or illegible when filed
10
Spec


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 30







lung cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Lung Cancer
GSE63704
Discovery
10
Detect


Normal Lung
GSE66836
Discovery
10
Detect


Lung Cancer
TCGA
Discovery
10
Spec


Non-lung cancers
TCGA
Discovery
80
Spec


Lung Cancer
GSE66836
Validatiotext missing or illegible when filed
164
Detect


Lung Cancer
GSE63704
Validatiotext missing or illegible when filed
17
Detect


Lung Cancer
GSE76269
Validatiotext missing or illegible when filed
56
Detect


Lung Cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec


Bladder Cancer
TCGA
Validatiotext missing or illegible when filed
439
Detect


Brain Cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast Cancer
TCGA
Validatiotext missing or illegible when filed
891
Spec


Cervical Cancer
TCGA
Validatiotext missing or illegible when filed
312
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney Cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec


Ovarian Cancer
TCGA
Validatiotext missing or illegible when filed
10
Spec


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach Cancer
TCGA
Validatiotext missing or illegible when filed
396
Spec


Testis Cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal Lung
GSE63704
Validatiotext missing or illegible when filed
112
Detect






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 31







prostate cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Control
GSE52955
Discovery
5
Detect


PRAD
TCGA
Discovery
10
Detect


PRAD
TCGA
Discovery
10
Spec


Non-PRAD
TCGA
Discovery
80
Spec


PRAD
GSE73549
Validatiotext missing or illegible when filed
77
Detect


PRAD
GSE52955
Validatiotext missing or illegible when filed
25
Detect


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Bladder
TCGA
Validatiotext missing or illegible when filed
439
Spec


Brain
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast
TCGA
Validatiotext missing or illegible when filed
891
Spec


Cervix
TCGA
Validatiotext missing or illegible when filed
312
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung
TCGA
Validatiotext missing or illegible when filed
919
Spec


Ovarian
TCGA
Validatiotext missing or illegible when filed
10
Spec


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec


Stomach
TCGA
Validatiotext missing or illegible when filed
397
Spec


Testis
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal Prostattext missing or illegible when filed
GSE73549
Validatiotext missing or illegible when filed
15
Detect


Normal Prostattext missing or illegible when filed
GSE52955
Validatiotext missing or illegible when filed
5
Detect


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 32







breast cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Healthy
GSE60185
Discovery
17
Detect


Breast Cancer
TCGA
Discovery
10
Detect


Breast Cancer
TCGA
Discovery
10
Spec


Non-breast cancers
TCGA
Discovery
80
Spec


Breast Cancer
GSE60185
Validation
239
Detect


Breast Cancer
GSE75067
Validation
188
Detect


Breast Cancer
TCGA
Validation
891
Detect


Bladder Cancer
TCGA
Validation
439
Spec


brain Cancer
TCGA
Validation
689
Spec


Cervical Cancer
TCGA
Validation
312
Spec


CRC
TCGA
Validation
459
Spec


ESCA
TCGA
Validation
202
Spec


HCC
TCGA
Validation
430
Spec


Kidney Cancer
TCGA
Validation
871
Spec


Lung Cancer
TCGA
Validation
919
Spec


Ovarian Cancer
TCGA
Validation
10
Spec


PANC
TCGA
Validation
195
Spec


PRAD
TCGA
Validation
553
Spec


Stomach Cancer
TCGA
Validation
396
Spec


Testis Cancer
TCGA
Validation
156
Spec


Healthy breast tissutext missing or illegible when filed
GSE10196text missing or illegible when filed
Validation
121
Detect


Healthy breast tissutext missing or illegible when filed
GSE60185
Validation
17
Detect


Normal adjasent tisstext missing or illegible when filed
GSE60185
Validation
29
Detect


Normal Blood
GSE40279
Validation
656
Spec


Normal Blood
GSE61496
Validation
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 33







colorectal cancer CRC











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Control
GSE6550
Discovery
25
Detect


CRC
TCGA
Discovery
50
Detect


CRC
TCGA
Discovery
10
Spec


Non-HCC
TCGA
Discovery
80
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
260
Detect


HCC
TCGA
Validatiotext missing or illegible when filed
459
Detect


Bladder
TCGA
Validatiotext missing or illegible when filed
439
Spec


Brain
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast
TCGA
Validatiotext missing or illegible when filed
891
Spec


Cervix
TCGA
Validatiotext missing or illegible when filed
312
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung
TCGA
Validatiotext missing or illegible when filed
919
Spec


Ovarian
TCGA
Validatiotext missing or illegible when filed
10
Spec


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach
TCGA
Validatiotext missing or illegible when filed
397
Spec


Testis
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal Colorectext missing or illegible when filed
GSE6550
Validatiotext missing or illegible when filed
8
Detect


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 34







Pancreatic cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Healthy
GSE53051
Discovery
12
Detect


Pancreatic Canctext missing or illegible when filed
TCGA
Discovery
20
Detect


Pancreatic Canctext missing or illegible when filed
TCGA
Discovery
20
Spec


Non-pancreatic
TCGA
Discovery
100
Spec


Pancreatic Canctext missing or illegible when filed
E-MTAB-text missing or illegible when filed
Validatiotext missing or illegible when filed
24
Detect


Pancreatic Canctext missing or illegible when filed
TCGA
Validatiotext missing or illegible when filed
195
Detect


Bladder Cancer
TCGA
Validatiotext missing or illegible when filed
439
Spec


brain Cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast Cancer
TCGA
Validatiotext missing or illegible when filed
891
Spec


Cervical Cancer
TCGA
Validatiotext missing or illegible when filed
312
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney Cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung Cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec


Ovarian Cancer
TCGA
Validatiotext missing or illegible when filed
10
Spec


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach Cancer
TCGA
Validatiotext missing or illegible when filed
396
Spec


Testis Cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal Blood
GSE40179
Validatiotext missing or illegible when filed
656
Spec


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 35







Brain cancer











Disease Status
Source
Cohort
N
Detect/Spec














Brain Cancer
TCGA
Discovery
10
Spec-Detect


Non-brain canctext missing or illegible when filed
TCGA
Discovery
158
Spec-Detect


Brain Cancer
GSE36278
Validatiotext missing or illegible when filed
136
Spec-Detect


Brain Cancer
GSE58298
Validatiotext missing or illegible when filed
40
Spec-Detect


Brain Cancer
GSE58218
Validatiotext missing or illegible when filed
228
Spec-Detect


Brain Cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec-Detect


Bladder Cancer
TCGA
Validatiotext missing or illegible when filed
439
Spec-Detect


Breast Cancer
TCGA
Validatiotext missing or illegible when filed
891
Spec-Detect


Cervical Cancer
TCGA
Validatiotext missing or illegible when filed
312
Spec-Detect


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec-Detect


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec-Detect


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec-Detect


Kidney Cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec-Detect


Lung Cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec-Detect


Ovarian Cancer
TCGA
Validatiotext missing or illegible when filed
10
Spec-Detect


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec-Detect


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec-Detect


Stomach Cancer
TCGA
Validatiotext missing or illegible when filed
396
Spec-Detect


Testis Cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec-Detect


Brain Cancer
GSE36278
Validatiotext missing or illegible when filed
6
Spec-Detect


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec-Detect


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec-Detect






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 36







Stomach cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Healthy
GSE99553
Discovery
14
Detect


Stomach Cancer
TCGA
Discovery
20
Detect


Stomach Cancer
TCGA
Discovery
7
Spec


Non-stomach cancer
TCGA
Discovery
100
Spec


Stomach Cancer
TCGA
Validation
396
Detect


Bladder Cancer
TCGA
Validation
439
Spec


brain Cancer
TCGA
Validation
689
Spec


Breast Cancer
TCGA
Validation
891
Spec


Cervical Cancer
TCGA
Validation
312
Spec


CRC
TCGA
Validation
459
Spec


ESCA
TCGA
Validation
202
Spec


HCC
TCGA
Validation
430
Spec


Kidney Cancer
TCGA
Validation
871
Spec


Lung Cancer
TCGA
Validation
919
Spec


Ovarian Cancer
TCGA
Validation
10
Spec


PANC
TCGA
Validation
195
Spec


PRAD
TCGA
Validation
553
Spec


Testis Cancer
TCGA
Validation
156
Spec


Normal Tissue
GSE99553
Validation
42
Detect


Normal Blood
GSE40179
Validation
656
Spec


Normal Blood
GSE61496
Validation
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 37







Ovarian cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Healthy
GSE65821
Discovery
6
Detect


Ovarian Cancer
TCGA
Discovery
10
Detect


Ovarian Cancer
TCGA
Discovery
10
Spec


Non-ovarian cancers
TCGA
Discovery
110
Spec


Ovarian Cancer
GSE65821
Validatiotext missing or illegible when filed
113
Detect


Bladder Cancer
TCGA
Validatiotext missing or illegible when filed
439
Spec


brain Cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast Cancer
TCGA
Validatiotext missing or illegible when filed
891
Detect


Cervical Cancer
TCGA
Validatiotext missing or illegible when filed
312
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney Cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung Cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach Cancer
TCGA
Validatiotext missing or illegible when filed
396
Spec


Testis Cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec


Healthy ovarian tissue
GSE87621
Validatiotext missing or illegible when filed
9
Detect


Healthy ovarian tissue
GSE74845
Validatiotext missing or illegible when filed
216
Detect


Healthy ovarian tissue
GSE81228
Validatiotext missing or illegible when filed
10
Detect


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 38







Cervical cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Healthy
GSE46306
Discovery
20
Detect


Cervical Cancer
TCGA
Discovery
10
Detect


Cervical Cancer
TCGA
Discovery
10
Spec


Non-cervical cancers
TCGA
Discovery
110
Spec


Cervical Cancer
GSE68339
Validatiotext missing or illegible when filed
270
Detect


Cervical Cancer
TCGA
Validatiotext missing or illegible when filed
312
Detect


Bladder Cancer
TCGA
Validatiotext missing or illegible when filed
439
Spec


brain Cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast Cancer
TCGA
Validatiotext missing or illegible when filed
891
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney Cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung Cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec


Ovarian Cancer
TCGA
Validatiotext missing or illegible when filed
10
Spec


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach Cancer
TCGA
Validatiotext missing or illegible when filed
396
Spec


Testis Cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 39







HNSC











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














HNSC
TCGA
Discovery
20
Detect


Control
GSE75537
Discovery
10
Detect


HNSC
TCGA
Discovery
20
Spec


Non-HNSC
TCGA
Discovery
190
Spec


Healthy
GSE40279
Discovery
10
Spec


HNSC
TCGA
Validation
580
Detect


Control
GSE52068
Validation
24
Detect


HNSC
GSE75537
Validation
54
Detect


HNSC
GSE79556
Validation
83
Detect


Control
GSE75537
Validation
54
Detect


HNSC
GSE52068
Validation
24
Detect


Bladder
TCGA
Validation
439
Spec


Brain
TCGA
Validation
689
Spec


Breast
TCGA
Validation
891
Spec


Cervix
TCGA
Validation
312
Spec


HCC
TCGA
Validation
459
Spec


ESCA
TCGA
Validation
202
Spec


HCC
TCGA
Validation
430
Spec


Kidney
TCGA
Validation
871
Spec


Lung
TCGA
Validation
919
Spec


Ovarian
TCGA
Validation
10
Spec


PANC
TCGA
Validation
195
Spec


PRAD
TCGA
Validation
553
Spec


Stomach
TCGA
Validation
396
Spec


Testis
TCGA
Validation
156
Spec


Normal Blood
GSE40279
Validation
656
Spec


Normal Blood
GSE52068
Validation
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 40







Esophageal cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Esophageal Cancer
IGSE52826
Discovery
6
Detect


ESCA
TCGA
Discovery
10
Detect


Non-esophageal canctext missing or illegible when filed
TCGA
Discovery
190
Spec


Healthy blood
GSE40279
Discovery
10
Spec


ESCA
TCGA
Discovery
20
Spec


Esophageal Cancer
TCGA
Validation
202
Detect


Bladder Cancer
TCGA
Validation
439
Spec


Brain Cancer
TCGA
Validation
689
Spec


Breast Cancer
TCGA
Validation
891
Spec


Cervical Cancer
TCGA
Validation
312
Spec


CRC Cancer
TCGA
Validation
459
Spec


Renal Cancer
TCGA
Validation
871
Spec


HNSC
TCGA
Validation
580
Spec


HCC
TCGA
Validation
430
Spec


Lung Cancer
TCGA
Validation
919
Spec


Ovarian Cancer
TCGA
Validation
10
Spec


Pancreatic Cancer
TCGA
Validation
195
Spec


Prostate Cancer
TCGA
Validation
553
Spec


Stomach Cancer
TCGA
Validation
397
Spec


Testis
TCGA
Validation
156
Spec


Normal Blood
GSE40279
Validation
656
Spec


Normal Blood
GSE61496
Validation
312
Spec


Normal tissues
Mix
Validation
115
Spec


Normal tissues
GSE85566
Validation
189
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 41







Bladder cancer











Disease Status
Source
Cohort
N
Detect/Spetext missing or illegible when filed














Bladder Cancer
TCGA
Discovery
10
Detect


Normal bladder
GSE52955
Discovery
5
Detect


Bladder Cancer
TCGA
Discovery
10
Spec


Non-bladder cancers
TCGA
Discovery
180
Spec


Bladder Cancer
TCGA
Validatiotext missing or illegible when filed
439
Detect


Brain Cancer
TCGA
Validatiotext missing or illegible when filed
689
Spec


Breast Cancer
TCGA
Validatiotext missing or illegible when filed
891
Spec


Cervical Cancer
TCGA
Validatiotext missing or illegible when filed
312
Spec


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec


Kidney Cancer
TCGA
Validatiotext missing or illegible when filed
871
Spec


Lung Cancer
TCGA
Validatiotext missing or illegible when filed
919
Spec


Ovarian Cancer
TCGA
Validatiotext missing or illegible when filed
10
Spec


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec


Stomach Cancer
TCGA
Validatiotext missing or illegible when filed
396
Spec


Testis Cancer
TCGA
Validatiotext missing or illegible when filed
156
Spec


Normal tissues
GSE50192
Validatiotext missing or illegible when filed
70
Spec


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 42







Kidney cancer











Disease Status
Source
Cohort
N
Detect/Spec














Control
GSE52955
Discovery
6
Spec-Detect


Kidney Cancer
TCGA
Discovery
30
Spec-Detect


Healthy blood
GSE40279
Discovery
10
Spec-Detect


Non-kidney cantext missing or illegible when filed
TCGA
Discovery
180
Spec-Detect


Kidney
TCGA
Validatiotext missing or illegible when filed
871
Spec-Detect


Bladder
TCGA
Validatiotext missing or illegible when filed
439
Spec-Detect


Brain
TCGA
Validatiotext missing or illegible when filed
689
Spec-Detect


Breast
TCGA
Validatiotext missing or illegible when filed
891
Spec-Detect


Cervix
TCGA
Validatiotext missing or illegible when filed
312
Spec-Detect


CRC
TCGA
Validatiotext missing or illegible when filed
459
Spec-Detect


ESCA
TCGA
Validatiotext missing or illegible when filed
202
Spec-Detect


HNSC
TCGA
Validatiotext missing or illegible when filed
580
Spec-Detect


HCC
TCGA
Validatiotext missing or illegible when filed
430
Spec-Detect


Lung
TCGA
Validatiotext missing or illegible when filed
919
Spec-Detect


Ovarian
TCGA
Validatiotext missing or illegible when filed
10
Spec-Detect


PANC
TCGA
Validatiotext missing or illegible when filed
195
Spec-Detect


PRAD
TCGA
Validatiotext missing or illegible when filed
553
Spec-Detect


Stomach
TCGA
Validatiotext missing or illegible when filed
397
Spec-Detect


Testis
TCGA
Validatiotext missing or illegible when filed
156
Spec-Detect


Normal Blood
GSE40279
Validatiotext missing or illegible when filed
656
Spec-Detect


Normal Blood
GSE61496
Validatiotext missing or illegible when filed
312
Spec-Detect


Normal tissues
Mix
Validatiotext missing or illegible when filed
189
Spec-Detect


Normal tissues
GSE85566
Validatiotext missing or illegible when filed
115
Spec-Detect






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 43







Testicular cancer











Disease Status
Source
Cohort
N
Detect/Spec














Testicular Cancer
TCGA
Discovery
10
Spec-Detect


Healthy blood
GSE40279
Discovery
10
Spec-Detect


Non-testis cancers
TCGA
Discovery
170
Spec-Detect


Testis Cancer
TCGA
Validation
156
Spec-Detect


Bladder Cancer
TCGA
Validation
439
Spec-Detect


Brain Cancer
TCGA
Validation
689
Spec-Detect


Breast Cancer
TCGA
Validation
891
Spec-Detect


Cervical Cancer
TCGA
Validation
312
Spec-Detect


CRC
TCGA
Validation
459
Spec-Detect


ESCA
TCGA
Validation
202
Spec-Detect


HCC
TCGA
Validation
430
Spec-Detect


Kidney Cancer
TCGA
Validation
871
Spec-Detect


Lung Cancer
TCGA
Validation
919
Spec-Detect


Ovarian Cancer
TCGA
Validation
10
Spec-Detect


PANC
TCGA
Validation
195
Spec-Detect


PRAD
TCGA
Validation
553
Spec-Detect


Stomach Cancer
TCGA
Validation
396
Spec-Detect


Normal tissues
GSE50192
Validation
70
Spec-Detect


Normal tissues
Mix
Validation
119
Spec-Detect


Normal Blood
GSE40279
Validation
656
Spec-Detect


Normal Blood
GSE61496
Validation
312
Spec-Detect
















TABLE 44







Pan-cancer











Disease Status
Source
Cohort
N
Detect/Spec














Healthy Blood
GSE40279
Discovery
10
Detect


TCGA cancers
TCGA
Discovery
170
Detect


Bladder Cancer
TCGA
Validation
439
Detect


Brain Cancer
TCGA
Validation
689
Detect


Breast Cancer
TCGA
Validation
891
Detect


Cervical Cancer
TCGA
Validation
312
Detect


CRC Cancer
TCGA
Validation
459
Detect


Esophagus Cancer
TCGA
Validation
202
Detect


HNSC Cancer
TCGA
Validation
580
Detect


HCC Cancer
TCGA
Validation
430
Detect


Lung Cancer
TCGA
Validation
919
Detect


Ovarian Cancer
TCGA
Validation
10
Detect


Pancreatic Cancer
TCGA
Validation
195
Detect


Prostate Cancer
TCGA
Validation
553
Detect


Stomach Cancer
TCGA
Validation
397
Detect


Normal Blood
GSE40279
Validation
656
Detect


Normal Blood
GSE61496
Validation
312
Detect
















TABLE 45







Melanoma











Disease Statutext missing or illegible when filed
Source
Cohort
N
Detect/Spec














Bladder Cantext missing or illegible when filed
TCGA
Validation
439
Detect/Spec


Brain Cancer
TCGA
Validation
689
Detect/Spec


Breast Cancetext missing or illegible when filed
TCGA
Validation
891
Detect/Spec


Cervical Cantext missing or illegible when filed
TCGA
Validation
312
Detect/Spec


CRC Cancer
TCGA
Validation
459
Detect/Spec


Esophagus Ctext missing or illegible when filed
TCGA
Validation
202
Detect/Spec


HNSC Cancer
TCGA
Validation
580
Detect/Spec


HCC Cancer
TCGA
Validation
430
Detect/Spec






text missing or illegible when filed indicates data missing or illegible when filed














TABLE 46







AML











Disease Statutext missing or illegible when filed
Source
Cohort
N
Detect/Spec














Normal Blootext missing or illegible when filed
GSE40279
Validation
656
Detect/Spec


Normal Blootext missing or illegible when filed
GSE61496
Validation
312
Detect/Spec


AML
GSE86409
Validation
79
Detect/Spec


AML
TCGA
Validation
140
Detect/Spec






text missing or illegible when filed indicates data missing or illegible when filed







BCD Method

The following are the steps of the Binary Categorical Differentiation method (BCD) invented by the inventors to discover polygenic DNA methylation markers for early prediction of different cancers.


We filtered the 28,754 CGIDs that are robustly unmethylated in normal tissues.


For the discovery cohort we delineated within the list of 28,754 CG IDs that are robustly unmethylated in normal tissues, CGIDs that are categorically methylated in a particular cancer and are not methylated in unaffected tissue and normal tissues using the COUNTIF and IF functions in Microsoft excel.


NmcCGIDx=COUNTIF (betaCGIDxCancer n1:ni,“>0.2”)


NmnCGIDx=COUNTIF (betaCGIDxNormal n1:ni,“>0.1”)


DMCGIDx=IF((AND(NmcCGIDx>0, NmnCGIDx=0)),“TRUE”,“FALSE”)

DM CGIDx were sorted from highest to lowest number


Up to 20 top TRUE DM CGIDx positions were selected


NmcCGIDx=Number of cancer patients with methylated CGIDx


Nmn=number of normal adjacent or similar tissue samples with methylated CGIDx


betaCGIDx=level of methylation of CGIDx


n=patient from 1 to i


DM=differentially methylated CGIDx


The inventors noticed that testicular and kidney cancer exhibit pervasive lack of methylation at CG IDs that are highly methylated in all tissues. We therefore used a modification of the BCD method to discover categorically differentially methylated CG ID positions for testicular and kidney (renal) cancer which we term “BCDhypo”; categorically unmethylated in cancer and methylated in normal tissue. The following steps were used for discovery of Differentially hypomethylated CGID positions in testicular and kidney cancer.


For the discovery cohort we delineated hypomethylated CGIDs in testis or kidney that are fully methylated in normal tissues using the COUNTIF and IF functions in excel.


NucCGIDx=COUNTIF (betaCGIDxCancer ni:ni,“<0.2”)


NunCGIDx=COUNTIF (betaCGIDxNormal ni:ni,“<0.9”)


DHMCGIDx=IF((AND(NucCGIDx>0, NunCGIDx=0)),“TRUE”,“FALSE”)

DHM CGID positions were sorted from highest to lowest number


20 top TRUE DHM sites were selected and subjected to penalized regression analysis


NucCGIDx=Number of cancer patients with unmethylated CGID X


NunCGIDx=number of normal tissue samples with unmethylated CGID X


n=patient from 1 to i


DHM=differentially hypomethylated CGID


The inventors then performed on the top 20 DM (or DHM) CGIDx a penalized regression using the package penalized in R to delineate the minimal combination of CGIDx that predicts cancer at highest sensitivity and specificity. The polygenic combination of CGIDx was further tested in a multivariate linear regression equation to determine the regression coefficient between methylation levels of these CGIDs in the polygenic combination and cancer. The model was used to calculate a methylation score for each patient for a typical cancer.






Ms=α+Σ
i=1
nβiCGi


Ms=methylation score, α=intercept, βi=coefficient for CG IDi, CGi=methylation level per CG in combination. 1 to i=number of CGs in combination.


Embodiment 3. Discovery of a Polygenic DNA Methylation Marker for Liver Cancer (HCC)

The inventors used normalized Illumina 450K DNA methylation data from GSE61258 (normal liver) and 66 randomly selected samples from the TCGA HCC collection of HCC DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDx that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect HCC with high sensitivity and specificity in the training cohort (FIG. 5B, Table 1) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 8 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between HCC and other tumors (FIG. 5C, Table 2) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 4. Utility of HCC Polygenic DNA Methylation Markers for Detecting HCC

The inventors then demonstrated that the weighted HCC DNA methylation score detected HCC in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from GSE76269 for 227 HCC patients for the CGIDs in Table 1. Using this method, 95% of the HCC samples were detected as HCC (FIG. 6C). A ROC curve presented in FIG. 6A reveals the specificity (1) and sensitivity (0.96) of this methylation score for detecting cancer. The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for detecting HCC and differentiating between HCC and other cancers using a “validation cohort” with methylation data from GSE75041 and TCGA for HCC and 8 other types of cancer. A ROC curve presented in FIG. 7B reveals the specificity (0.97) and sensitivity (0.95) of this methylation score for differentiating HCC from other normal tissue and other cancers. These DNA methylation markers and the calculated methylation score could be used for screening and early detection of cancer in people at risk as well as the general healthy population using different biomaterial from people such as tissue, feces, saliva, plasma and urine.


Embodiment 5. Discovery of a Polygenic DNA Methylation Marker for Lung Cancer

The inventors used normalized Illumina 450K DNA methylation data for 10 people from GSE61258 (normal lung) and 10 randomly selected samples from the TCGA lung cancer collection of lung cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect lung cancer with high sensitivity and specificity (Samples included both adenocarcinoma and squamous cell carcinoma) in the training cohort (FIG. 8B, Table 3) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 8 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between lung cancer and other tumors (FIG. 8C, Table 4) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 6. Utility of Lung Polygenic DNA Methylation Markers for Detecting Lung Cancer

The inventors then demonstrated that the weighted lung cancer DNA methylation score and threshold developed in embodiment 3 (detect) detects lung cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from GSE66836, GSE63704, GSE76269 and 919 lung cancer patients from TCGA. Using this method 96% of the lung cancer samples were detected as lung cancer (FIG. 9A). The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for differentiating between lung cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for lung cancer and 8 other types of cancer (FIG. 9A). A ROC curve presented in FIG. 9B reveals the specificity (0.96) and sensitivity (0.84) of this methylation score for detecting lung cancer from other normal tissue and other cancers (FIG. 9C). These DNA methylation markers and the calculated methylation score could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 7. Discovery of a Polygenic DNA Methylation Marker for Prostate Cancer

The inventors used normalized Illumina 450K DNA methylation data for 5 people from GSE52955 (normal prostate) and 10 randomly selected samples from the TCGA prostate cancer collection of prostate cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect prostate cancer with high sensitivity and specificity in the training cohort (FIG. 10b, Table 5) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 8 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between prostate cancer and other tumors (FIG. 10C, Table 6) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 8. Utility of Prostate Cancer Polygenic DNA Methylation Markers for Detecting Prostate Cancer

The inventors then demonstrated that the weighted prostate cancer DNA methylation score and threshold developed in embodiment 3 (detect) detects prostate cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from GSE73549, GSE2955, and 430 prostate cancer patients from TCGA. Using this method 99% of the prostate cancer samples were detected as prostate cancer (FIG. 11A). The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for differentiating between prostate cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for prostate cancer and 8 other types of cancer (FIG. 11A). A ROC curve presented in FIG. 11B reveals the specificity (0.99) and sensitivity (0.98) of this methylation score for detecting prostate cancer from other normal tissue and other cancers (FIG. 11C). These DNA methylation markers and the calculated methylation score could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 9. Discovery of a Polygenic DNA Methylation Marker for Breast Cancer

The inventors used normalized Illumina 450K DNA methylation data for 17 people from GSE60185(normal breast) and 10 randomly selected samples from the TCGA breast cancer collection of breast cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect breast cancer with high sensitivity and specificity in the training cohort (FIG. 12B, Table 7) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 8 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between breast cancer and other tumors (FIG. 12C, Table 8) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 10. Utility of Breast Cancer Polygenic DNA Methylation Markers for Detecting Breast Cancer

The inventors then demonstrated that the weighted breast cancer DNA methylation score and threshold developed in embodiment 9 (detect) detects breast cancer in a “validation cohort” that included 891 breast cancer patients using normalized Illumina 450K DNA methylation beta values from GSE60185, GSE75067, and from TCGA. Using this method 91% of the breast cancer samples were detected as breast cancer (FIG. 13A) and both DCIS and invasive cancers were detected. The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for differentiating between breast cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for breast cancer and 8 other types of cancer (FIG. 14A). A ROC curve presented in FIG. 14B reveals the specificity (0.89) and sensitivity (0.87) of this methylation score for differentiating breast cancer from other normal tissue and other cancers (FIG. 14C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of breast cancer in women at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 11. Discovery of a Polygenic DNA Methylation Marker for Colorectal Cancer (CRC)

The inventors used normalized Illumina 450K DNA methylation data for 25 people from GSE(32146) (normal) and 50 randomly selected samples from the TCGA colorectal cancer collection of colorectal cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect colorectal cancer with high sensitivity and specificity in the training cohort (FIG. 15B, Table 9) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 8 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between colorectal cancer and other tumors (FIG. 15C, Table 10) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 12. Utility of Colorectal Cancer Polygenic DNA Methylation Markers for Detecting Colorectal Cancer

The inventors then demonstrated that the weighted colorectal cancer DNA methylation score and threshold developed in embodiment 11 (detect) detects colorectal cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from GSE69550 and 459 colorectal cancer patients from TCGA. Using this method 98% of the colorectal cancer samples were detected as colorectal cancer (FIG. 16A). The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for differentiating between colorectal cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for colorectal cancer and 8 other types of cancer (FIG. 16A). A ROC curve presented in FIG. 16B reveals the specificity (0.96) and sensitivity (0.98) of this methylation score for detecting colorectal cancer from other normal tissues and other cancers (FIG. 16C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk for CRC as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 13. Discovery of a Polygenic DNA Methylation Marker for Pancreatic Cancer

The inventors used normalized Illumina 450K DNA methylation data for 12 people from GSE53051 (normal) and 20 randomly selected samples from the TCGA collection of pancreatic cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect pancreatic cancer with high sensitivity and specificity in the training cohort (FIG. 17B, Table 11) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 100 randomly selected DNA methylation samples from TCGA representing 10 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between pancreatic cancer and other tumors (FIG. 17C, Table 12) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 14. Utility of Pancreatic Cancer Polygenic DNA Methylation Markers for Detecting Pancreatic Cancer

The inventors then demonstrated that the weighted pancreatic cancer DNA methylation score and threshold developed in embodiment 13 (detect) detects pancreatic cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 891 pancreatic cancer patients from TCGA. Using this method 86% of the pancreatic cancer samples were detected as pancreatic cancer (FIG. 18A). The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for differentiating between pancreatic cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for pancreatic cancer and 9 other types of cancer (FIG. 18A). A ROC curve presented in FIG. 18B reveals the specificity (0.93) and sensitivity (0.86) of this methylation score for detecting pancreatic cancer and differentiating it from other normal tissue and other cancers (FIG. 18C). These DNA methylation markers and the calculated methylation scores from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 15. Discovery of a Polygenic DNA Methylation Marker for Brain Cancer

The inventors used normalized Illumina 450K DNA methylation data for 10 people from GSE65820 (normal) and 10 randomly selected samples from the TCGA collection of brain cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a set of binary categorical differentially methylated CGIDs that detect brain cancer with high sensitivity and specificity in the training cohort (FIG. 19B, Table 13) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 110 randomly selected DNA methylation samples from TCGA representing 11 different tumor types. The inventors used this training cohort and discovered that the detect CGID also differentiates between brain cancer and other tumors (FIG. 19C, Table 13) (detect-spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 16. Utility of Brain Cancer Polygenic DNA Methylation Markers for Detecting Brain Cancer

The inventors then demonstrated that the weighted brain cancer DNA methylation score and threshold developed in embodiment 15 (detect) detects brain cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 689 brain cancer patients from TCGA, 40 patients from GSE58298 and 136 patients from GSE36278. Using this method 91-97% of the brain cancer samples were detected as brain cancer (FIG. 20A). The inventors then demonstrated the utility of the same CGIDs for differentiating between brain cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for brain cancer and 9 other types of cancer (FIG. 20A). A ROC curve presented in FIG. 22B reveals the specificity (1) and sensitivity (0.97) of this methylation score for detecting brain cancer from other normal tissue and other cancers (FIG. 20C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 17. Discovery of a Polygenic DNA Methylation Marker for Stomach Cancer

The inventors used normalized Illumina 450K DNA methylation data for 18 people from GSE99553 (normal) and 10 randomly selected samples from the TCGA collection of stomach cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect stomach cancer with high sensitivity and specificity in the training cohort (FIG. 21B, Table 14) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 100 randomly selected DNA methylation samples from TCGA representing 11 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between stomach cancer and other tumors (FIG. 21C, Table 15) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 18. Utility of Stomach Cancer Polygenic DNA Methylation Markers for Detecting Stomach Cancer

The inventors then demonstrated that the weighted stomach cancer DNA methylation score and threshold developed in embodiment 17 (detect) detects stomach cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 397 stomach cancer patients from TCGA. Using this method, 88% of the stomach cancer samples were detected as stomach cancer (FIG. 23A). The inventors then demonstrated the utility of the combined spec and detect DNA methylation scores for differentiating between stomach cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for stomach cancer and 10 other types of cancer (FIG. 23A). A ROC curve presented in FIG. 22B reveals the specificity (0.9 for) and sensitivity (0.9) of this methylation score for detecting stomach cancer from other normal tissues and other cancers (FIG. 22C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for screening and early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 19. Discovery of a Polygenic DNA Methylation Marker for Ovarian Cancer

The inventors used normalized Illumina 450K DNA methylation data for 5 people from GSE65820 (normal) and 10 randomly selected samples from the TCGA collection of ovarian cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect ovarian cancer with high sensitivity and specificity in the training cohort (FIG. 23B, Table 16) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 100 randomly selected DNA methylation samples from TCGA representing 10 different tumor types and blood. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between ovarian cancer and other tumors (FIG. 2C, Table 17) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 20. Utility of Ovarian Cancer Polygenic DNA Methylation Markers for Detecting Ovarian Cancer

The inventors then demonstrated that the weighted ovarian cancer DNA methylation score and threshold developed in embodiment 19 (detect) detects ovarian cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 114 ovarian cancer patients from TCGA. Using this method 86% of the ovarian cancer samples were detected as ovarian cancer (FIG. 24A). The inventors then demonstrated the utility of the spec DNA methylation scores for differentiating between ovarian cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for ovarian cancer and 9 other types of cancer (FIG. 24A). A ROC curve presented in FIG. 24B reveals the specificity (0.99) and sensitivity (1) of this methylation score for differentiating ovarian cancer from other normal tissue and other cancers (FIG. 24C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 21. Discovery of a Polygenic DNA Methylation Marker for Cervical Cancer

The inventors used normalized Illumina 450K DNA methylation data for 20 people from GSE46306 (normal) and 10 randomly selected samples from the TCGA collection of cervix cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect cervical cancer with high sensitivity and specificity in the training cohort (FIG. 25B, Table 18) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 8 different tumor types and blood. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between cervical cancer and other tumors (FIG. 25C, Table 19) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 22. Utility of Cervical Cancer Polygenic DNA Methylation Markers for Detecting Cervical Cancer

The inventors then demonstrated that the weighted cervical cancer DNA methylation score and threshold developed in embodiment 21 (detect) detects cervical cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 313 cervical cancer patients from TCGA. Using this method 91% of the cervical cancer samples were detected as cervical cancer (FIG. 26A). The inventors then demonstrated the utility of the spec DNA methylation scores for differentiating between cervical cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for cervical cancer and 9 other types of cancer (FIG. 26A). A ROC curve presented in FIG. 26B reveals the specificity (0.9) and sensitivity (0.9) of this methylation score for detecting cervical cancer and differentiating it from other normal tissue and other cancers (FIG. 26C). These DNA methylation markers and the calculated methylation scores from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 23. Discovery of a Polygenic DNA Methylation Marker for Head and Neck Squamous Carcinoma (HNSC)

The inventors used normalized Illumina 450K DNA methylation data for 10 people from GSE(52068) (normal) and 10 randomly selected samples from the TCGA cancer collection of HNSC DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect HNSC with high sensitivity and specificity in the training cohort (FIG. 27B, Table 20) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 12 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between HNSC and other tumors (FIG. 27C, Table 21) (spec).


Embodiment 24. Utility of Head and Neck Squamous Carcinoma (HNSC) Polygenic DNA Methylation Markers for Detecting HNSC

The inventors then demonstrated that the weighted HNSC DNA methylation score and threshold developed in embodiment 23 (detect) detects HNSC in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from GSE52068 and. Using this method 88%-96% of the HNSC samples were detected (FIG. 28A). The inventors then demonstrated the utility of the DNA methylation detect scores for differentiating between HNSC and other cancers using a “validation cohort” with methylation data from GSE and TCGA for HNSC and 12 other types of cancer (FIG. 28A). A ROC curve presented in FIG. 28B reveals the specificity (0.86) and sensitivity (0.88) of this methylation score for differentiating HNSC from other normal tissue and other cancers (FIG. 28C). The markers also detect several other cancers (at relatively high sensitivity and is thus of limited specificity for these cancers). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 25. Discovery of a Polygenic DNA Methylation Marker for ESOPHAGEAL CANCER

The inventors used normalized Illumina 450K DNA methylation data for 10 people from GSE(52068) (normal) and 10 randomly selected samples from the TCGA cancer collection of esophageal cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect esophageal cancer with high sensitivity and specificity in the training cohort (FIG. 29B, Table 22) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 12 different tumor types. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between esophageal cancer and other tumors (FIG. 29C, Table 23) (spec).


Embodiment 26. Utility of ESOPHAGEAL CANCER Polygenic DNA Methylation Markers for Detecting ESOPHAGEAL CANCER

The inventors then demonstrated that the weighted esophageal cancer DNA methylation score and threshold developed in embodiment 25 (detect) detects esophageal cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from GSE52068 and. Using this method 88%-96% of the esophageal cancer samples were detected (FIG. 30A). The inventors then demonstrated the utility of the detect DNA methylation scores for differentiating between esophageal cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for esophageal cancer and 12 other types of cancer (FIG. 30A). A ROC curve presented in FIG. 30B reveals the specificity (0.86) and sensitivity (0.88) of this methylation score for differentiating esophageal cancer from other normal tissue and other cancers (FIG. 30C). The markers also detect several other cancers (at relatively high sensitivity and are thus of limited specificity for these cancers). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 27. Discovery of a Polygenic DNA Methylation Marker for Bladder Cancer

The inventors used normalized Illumina 450K DNA methylation data for 5 people from GSE52955 (normal) and 10 randomly selected samples from the TCGA collection of bladder cancer DNA methylation data as a “training” cohort. The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detects bladder cancer with high sensitivity and specificity in the training cohort (FIG. 31B, Table 24) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2. The inventors then generated a “training cohort” from 80 randomly selected DNA methylation samples from TCGA representing 13 different tumor types and normal blood. The inventors used this training cohort to discover a polygenic set of differentially methylated CGIDs between bladder cancer and other tumors (FIG. 31C, Table 25) (spec). A weighted DNA methylation score was developed for the CGIDs as described in embodiment 2.


Embodiment 28. Utility of Bladder Cancer Polygenic DNA Methylation Markers for Detecting Bladder Cancer

The inventors then demonstrated that the weighted bladder cancer DNA methylation score and threshold developed in embodiment 27 (detect) detects bladder cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 439 bladder cancer patients from TCGA. Using this method 96% of the bladder cancer samples were detected as bladder cancer (FIG. 32B). The inventors then demonstrated the utility of the spec DNA methylation scores for differentiating between bladder cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for bladder cancer and 13 other types of cancer (FIG. 32B). A ROC curve presented in FIG. 32C reveals the specificity (0.86) and sensitivity (0.88) of this methylation score for detecting bladder cancer from other normal tissue and other cancers (FIG. 32C). There is cross detection at reasonably high rates however of stomach, pancreatic esophageal and colorectal cancer. These DNA methylation markers and the calculated methylation scores from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 29. Discovery of a Polygenic DNA Methylation Marker for Kidney Cancer

The inventors used normalized Illumina 450K DNA methylation data for kidney (renal) cancer from 10 people from GSE52955 (normal) and 10 randomly selected samples per cancer from 13 cancers in the TCGA dataset as a “training” cohort as well as normal tissues and blood (GSE40279, GSE 52955). The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that are robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD hypo method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect kidney cancer with high sensitivity and specificity in the training cohort and are specific for kidney cancer against other cancers “Detect-Spec” (FIG. 33B, Table 26) (detect-spec). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2.


Embodiment 30. Utility of Kidney Cancer Polygenic DNA Methylation Markers for Detecting Kidney Cancer

The inventors demonstrated that the weighted kidney cancer DNA methylation score and threshold developed in embodiment 27 (“Detect-Spec”) detects kidney cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 871 kidney cancer patients from TCGA and differentiate kidney cancer from other cancers. Using this method 90% of the kidney cancer samples were detected as kidney cancer (FIG. 34A). The inventors then demonstrated the utility of the “Detect-Spec” DNA methylation scores for differentiating between kidney cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for kidney cancer and 13 other types of cancer (FIG. 34A). A ROC curve presented in FIG. 34B reveals the specificity (0.87) and sensitivity (0.91) of this methylation score for detecting kidney cancer from other normal tissue and other cancers (FIG. 34C) (high crossover with HCC, brain and testis). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of kidney cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 31. Discovery of a Polygenic DNA Methylation Marker for Testicular Cancer

The inventors used normalized Illumina 450K DNA methylation data for testicular cancer from 10 people from GSE46306 (normal) and 10 randomly selected samples per cancer from 13 cancers in the TCGA dataset as a “training” cohort as well as normal tissues and blood (GSE40279, GSE 61496). The inventors first shortlisted in the “training cohort” dataset 28754 CGIDs that were discovered in embodiment 1 as sites that robustly unmethylated across normal tissues and blood samples. The inventors then used the BCD hypo method described in embodiment 2 to discover a polygenic set of binary categorical differentially methylated CGIDs that detect testicular cancer with high sensitivity and specificity in the training cohort and are specific for testicular cancer against other cancers “Detect-Spec” (FIG. 35B, Table 27) (detect-spec). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2.


Embodiment 32. Utility of Testicular Cancer Polygenic DNA Methylation Markers for Detecting Testis Cancer

The inventors then demonstrated that the weighted testicular cancer DNA methylation score and threshold developed in embodiment 31 (“Detect-Spec”) detects testicular cancer in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 156 testicular cancer patients from TCGA and differentiate testicular cancer from other cancer. Using this method 96% of the testicular cancer samples were detected as testicular cancer (FIG. 36A). The inventors then demonstrated the utility of the “Detect-Spec” DNA methylation scores for differentiating between testicular cancer and other cancers using a “validation cohort” with methylation data from GSE and TCGA for testicular cancer and 13 other types of cancer (FIG. 36A). A ROC curve presented in FIG. 36B reveals the specificity (0.97) and sensitivity (0.96) of this methylation score for detecting testicular cancer from other normal tissue and other cancers (FIG. 36C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 33. Discovery of a Polygenic Pan-Cancer DNA Methylation Marker for 13 Common Solid Tumors

The inventors used normalized Illumina 450K DNA methylation data for 10 randomly selected samples per cancer from 13 cancers (bladder cancer, brain cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, HNSC, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, stomach cancer) in the TCGA dataset as a “training” cohort as well as normal tissues and blood from TCGA an GEO. The inventors then performed a penalized regression on the combined list of CGIDs for detection of 10 different cancer listed in tables x-y and shortlisted CGIDs that detect any of the 10 common cancers at high sensitivity and specificity (FIG. 37B, Table 28) (detect). A weighted DNA methylation score and a threshold value for cancer was developed for the CGIDs as described in embodiment 2.


Embodiment 34. Utility of Pan-Cancer Polygenic DNA Methylation Markers for Detecting Cancer

The inventors then demonstrated that the weighted cancer DNA methylation score and threshold developed in embodiment 33 (“Detect”) detects 13 common cancers (bladder cancer, brain cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, HNSC, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, stomach cancer) in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 3644 cancer patients from TCGA from other normal tissues. Using this method 90-95% of cancer samples were detected (FIG. 38A). A ROC curve presented in FIG. 38B reveals the specificity (0.99) and sensitivity (0.95) of this methylation score for detecting 13 cancers from other normal tissue (FIG. 38C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of cancer in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 35. Discovery of a Polygenic DNA Methylation Marker for Detection of Melanoma

The inventors used normalized Illumina 450K DNA methylation data for 10 randomly selected melanoma samples and 220 samples from other cancers (bladder cancer, brain cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, HNSC, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, stomach cancer) and normal blood in the TCGA and GEO datasets as a “training” cohort. The inventors then performed a penalized regression on the combined list of CGIDs for detection of melanoma and shortlisted CGIDs that detect melanoma at high sensitivity and specificity (FIG. 39, Table 28) (detect-spec). A weighted DNA methylation score and a threshold value for melanoma was developed for the CGIDs as described in embodiment 2.


Embodiment 36. Utility of Melanoma Polygenic DNA Methylation Marker for Detecting Melanoma

The inventors then demonstrated that the weighted melanoma DNA methylation score and threshold developed in embodiment 35 (“Detect-spec”) detects melanoma in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 475 melanoma patients from TCGA from other cancer and normal tissues. Using this method 98% of melanoma samples were detected (FIG. 40A). A ROC curve presented in FIG. 40B reveals the specificity (0.98) and sensitivity (0.95) of this methylation score for detecting melanoma from other normal tissue and other cancers (FIG. 40C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of melanoma in people at risk as well as the general healthy population using different biomaterial from the patient from tissues, feces, saliva, plasma and urine.


Embodiment 37. Discovery of a Polygenic DNA Methylation Marker for Detection of Acute Myeloid Leukemia (AML)

The inventors used normalized Illumina 450K DNA methylation data for 10 randomly selected AML samples and 10 normal blood samples in the GEO datasets as a “training” cohort. The inventors then performed a penalized regression on the combined list of CGIDs for detection of AML and shortlisted CGIDs that detect melanoma at high sensitivity and specificity (FIG. 41, Table 27) (detect-spec). A weighted DNA methylation score and a threshold value for melanoma was developed for the CGIDs as described in embodiment 2.


Embodiment 38. Utility of Acute Myeloid Leukemia (AML) Polygenic DNA Methylation Markers for Detecting AML in Blood DNA

The inventors then demonstrated that the weighted melanoma DNA methylation score and threshold developed in embodiment 37 (“Detect-spec”) detects AML in a “validation cohort” that included normalized Illumina 450K DNA methylation beta values from 79 AML patients from GEO and 140 patients from TGCA and normal blood. Using this method 100% of AIL samples were detected (FIG. 42A). A ROC curve presented in FIG. 42B reveals the specificity (1) and sensitivity (1) of this methylation score for detecting AML from blood (FIG. 42C). These DNA methylation markers and the calculated methylation score from the methylation values could be used for early detection of AML in people at risk as well as the general healthy population using blood DNA.


Embodiment 39. Bisulfite Conversion, Multiplex Amplification and Next Generation Sequencing and Calculation of a Methylation Score for Predicting Prostate Cancer

Blood was collected in 9-ml tubes containing K3-EDTA and processed within 1 h. Fresh blood samples were centrifuged at 1000 g for 10 min at 4° C. The supernatant was carefully transferred to a Falcon tube without disturbing the cellular layer and centrifuged again for 10 min for complete removal of any residual cells and frozen at −80 C. Plasma samples are thawed, and DNA is extracted by several available methods and commercially available kits for plasma DNA extraction such as Qiagen kit for plasma DNA or EZ DNA direct extraction method. DNA is purified using commercially available methods such as on AMPure XP magnetic beads and purified DNA is treated with sodium bisulfite using for example the EZ DNA bisulfite treatment kit. A library of targeted sequences is generated by two step PCR reactions (FIG. 40). The first PCR reaction targets the specific CGIDs from table 5 and 6, note PCR1 primer has complementary sequence to second PCR2 primers (FIG. 40). The inventors used human bisulfite converted genomic DNA from HEK293 cells to amplify concurrently three sequences of DNA that contain CGIDs that detect prostate cancer from HIF3A (232 base pairs region), TPM4 (213 base pairs region), and CTTN (199 base pairs region) in a multiplexed PCR reaction using the following primers in a standard Taq polymerase reaction: For CGID cg02879662;











5′ACACTCTTTCCCTACACgACgCTCTT






CCgATCTNNNNNGGTAGGAGTTTTGGG






forward primer:



AATTGG3′






and



reverse primer:



5′gTgACTggAgTTCAgACgTgTgCTCTT






CCgATCTCCACCCCTACAATCCCTAA3′






For CGID cg16232979;



forward primer:



5′ACACTCTTTCCCTACACgACgCTCTTCC






gATCTNNNNNYGGTTTYGGGTTTYGTATT3′






and reverse primer:



5′gTgACTggAgTTCAgACgTgTgCTCTTCCg






ATCTACRCAAAAATATAAATCRACRATC3′






ForCGID: cg14041701 and



cg14498227; forward primer:



5′ACACTCTTTCCCTACACgACgCTCTTCCgA






TCTNNNNNGTTTTGYGTTTYGGATTTGGGTT3′






and reverse primer:



5’gTgACTggAgTTCAgToACgTgTgCTCTT






CCgATCTCATAAACAACACCTTTAAATAAA






CACTAAA3′.







The amplified fragments were fractionated on an agarose gel


To barcode the samples, we use a second PCR reaction with the following primers:











Forward primer:



5′AATgATACggCgACCACCgAgATCTACACT






CTTTCCCTACACgAC3′









Barcoding primer (reverse):



5′CAAgCAgAAgACggCATACgAgATAGTCAT







CGgTgACTggAgTTCAgACgTg3′








(bold bases are the index; 200 variations of this index are used. The second set of primers introduces the index for each patient as well as the reverse and forward sequencing primers. A multiplex PCR1 reaction for the three markers of prostate cancer HIF3A 232 bp, TPM4 213 bp, and CTTN 199 bp is shown on the right panel using varying primer concentrations as indicated in FIG. 41.


Embodiment 40: Utility of the Method of Bisulfite Conversion, Multiplex Amplification and Next Generation Sequencing and Calculation of a Methylation Score for Predicting Cancer

The inventors demonstrate that embodiment 35 could be used for high throughput prediction of prostate cancer and other cancers using plasma samples from hundreds of patients at the same time. Indexed amplification of highly predictive CG IDs and a streamlined method for calculating a methylation score that is indicative of cancer could be used for early detection of prostate cancer and any other cancer.


Embodiment 41. Demonstration that the Biomarkers Selected Exhibit True BCD Properties are Totally Hypomethylated in Plasma from Healthy People

Plasma DNA was extracted from plasma prepared from 40 healthy individuals and was subjected to targeted amplification with cancer specific primers for the following cancers: liver, prostate, lung (FIG. 43) and stomach, pan cancer and CRC (FIG. 44) followed by barcoding using a second set of amplifications (PCR 2) and next generation sequencing as described in embodiments 39 and 40. All CGs exhibited very low levels of methylation in plasma from healthy people (FIGS. 43 and 44).


Embodiment 42. Bioinformatics Work-Flow for Determining DNA Methylation Levels

PCR2 products are combined quantified and purified and subjected to next generation sequencing on a Miseq Illumina sequencer. Sequence is demultiplexed using Illumina software for indexed sequencing, FASTQ files are generated for each patient. A Perl text editing script https://www.activestate.com/activeperl/downloads is use to count Ts and Cs in the FASTQ files per patient per CG IDS and quantify the fraction of methylated Cs in a CG ID in a patient by dividing the number of C/C+T. (see scheme in FIG. 42). The output CSV file is used for calculating the methylation score (Ms) for each patient using the equation: Ms=α+Σi=1nβiCGi when α=intercept β=coefficient for CG ID i, CG=methylation level per CG in the combination of CG IDs from 1 to n. n=number of CGs in combination. MS=methylation score.


Applications of the Inventive Subject Matter

The applications of the inventive subject matter are in the field of molecular diagnostics and early prediction of cancer in general. Any person skilled in the art could use the present inventive subject matter to derive similar noninvasive biomarkers for early prediction of other cancers and other diseases that are accompanied by cell death and shedding of cell free DNA into the system such as neurological diseases, diabetes, heart disease such as cirrhosis and damage to heart tissue in cardiovascular disease. The present inventive subject matter provides a path to finding exquisite methylation markers of specific cell types and tissues using the BCD and BCDhypo method. Also disclosed are methods and biomarkers for early prediction of a wide range of cancers that could be used by anyone skilled in the art to detect cancer early and dramatically enhance survival rates and cure from cancer. The methods disclosed by the present invention could be used by anyone skilled in the art for routine yearly screening of healthy populations, to identify people who are starting to develop cancer and to treat them immediately and prevent the dire personal social and economic consequences of cancer mortality and morbidity, as well as for monitoring “high risk” people and monitoring response to therapy in patients undergoing treatment to detect recurrence or metastasis. Adoption of the present invention described here for routine health care management by health providers and health check-up facilities will have a vast impact on reducing the burden of cancer as well as health care costs.


The fact that the inventive subject matter includes a number of different dependent claims does not mean that one cannot use a combination of these claims for predicting cancer. The embodiments disclosed herein for measuring and statistically analyzing and predicting cancer should not be considered limiting. Various other modifications will be apparent to those skilled in the art to measure DNA methylation in cancer patients such as Illumina EPIC arrays, capture array sequencing, next generation sequencing, methylation specific PCR, epityper, restriction enzyme-based analyses and other methods found in the public domain. Similarly, there are numerous statistical methods in the public domain in addition to those listed here to use the inventive subject matter for prediction of cancer in patient samples.


Although the inventive subject matter has been explained in relation to its embodiments including one or more preferred embodiments, it is to be understood that many other possible modifications and variations can be made without departing from the spirit and scope of the claimed subject matter.

Claims
  • 1. A method of using a “binary-categorical” DNA methylation signature of cancer to detect cancer comprising the step of deriving said “binary” DNA methylation signature by using a “binary-categorical differentiation (BCD)” method from genome wide DNA methylation maps.
  • 2. The method of claim 1 wherein said genome wide DNA methylation maps are one or more of cancer cells, normal tissues and blood DNA.
  • 3. The method of claim 1 wherein said “binary-categorical differentiation (BCD)” method includes use of one or more of the following: Illumina 27K, 450K or EPIC arrays, genome wide bisulfite sequencing, methylated DNA Immunoprecipitation (MeDIP) sequencing and hybridization with oligonucleotide arrays.
  • 4. The method of claim 1 for detecting hepatocellular carcinoma (HCC) Liver cancer (detect) and for differentiating from other tumors (spec) by measuring DNA methylation levels of one or more of the CG IDs from the following tables and subsets deriving a “cancer methylation score” using either a “binary categorical” or linear regression equation in DNA originating from the tumor in biological material derived from the patient such as saliva, urine, feces and cell free plasma DNA wherein the tables and subsets are the following:
  • 5.-22. (canceled)
  • 5. A kit for detecting at least one of cancer, hepatocellular carcinoma (HCC), lung cancer, prostate cancer, breast cancer, colorectal cancer, pancreatic cancer, brain cancer “glioblastoma”, gastric (stomach) cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma (HNSC), esophageal carcinoma, bladder cancer, renal kidney cancer, testicular cancer, common solid tumors, acute myeloid leukemia (AML), melanoma, comprising equipment and one or more reagents used for deriving DNA methylation measurements of the DNA methylation score according to the method of claim 1.
  • 24.-42. (canceled)
  • 6. The method of claim 4, further comprising the step of using DNA pyrosequencing methylation assays for predicting cancer by using DNA methylation combination.
  • 7. The method of claim 4, further comprising the step of using multiplexed amplification targeted bisulfite sequencing on a next generation sequencer such as the Illumina MiSeq for detecting cancer.
  • 8. The method of claim 4, further comprising the step of using a “binary categorical assay” for calculating a methylation score for predicting cancer.
  • 9. The method of claim 4, further comprising the step of using a multivariate linear regression equation for calculating a methylation score to predict cancer.
  • 10. The method of claim 4, further comprising the step of using a Receiver operating characteristics (ROC) assay to define a “methylation score” threshold differentiating cancer from noncancer and tissue of origin by using measurements of DNA methylation combinations.
  • 11. A method for identifying DNA methylation signature and a “polygenic DNA methylation marker” for detecting tissue of origin of cell free DNA in body fluids such as saliva, urine, plasma, feces or tissue biopsy comprising using the “binary-categorical differentiation (BCD)” method on genome wide DNA methylation maps of a specific tissue and genome wide DNA methylation maps of all other normal tissues and blood DNA, such as Illumina 27K, 450K or EPIC arrays, genome wide bisulfite sequencing, methylated DNA Immunoprecipitation (MeDIP) sequencing or hybridization with oligonucleotide arrays comprising the step of performing statistical analysis on DNA methylation measurements obtained from samples.
  • 12. The method according to claim 11 for further identifying “polygenic DNA methylation markers” of a diseased tissue such as neurons in brain, pancreas in diabetes, heart in ischemia and heart disease wherein an identification such as dying neurons CF DNA in plasma can be used for early detection of disease such as Alzheimer disease.
  • 13. The method according to claim 11, wherein said DNA methylation measurements are obtained by performing Illumina Beadchip 450K or EPIC assay of DNA extracted from one or more samples.
  • 14. The method according to claim 11, wherein said DNA methylation measurements are obtained by performing Illumina next generation sequencing on one or more of the following platforms: iSeq, MiniSeq, MiSeq or NextSeq sequencers, torrent sequencing, DNA pyrosequencing, mass spectrometry based (Epityper™) and PCR based methylation assays of DNA extracted from sample.
  • 15. The method according to claim 11, wherein said statistical analysis includes Pearson correlation.
  • 16. The method according to claim 11, wherein said statistical analysis includes Receiver operating characteristics (ROC) assays.
  • 17. The method according to claim 11, wherein said statistical analysis includes hierarchical clustering analysis assays
PCT Information
Filing Document Filing Date Country Kind
PCT/IB2019/055855 7/9/2019 WO 00
Provisional Applications (1)
Number Date Country
62695429 Jul 2018 US