Non-invasive early detection of cancer four years before conventional diagnosis using a blood test

Xingdong Chen, Jeffrey Gole, Athurva Gore, Qiye He, Ming Lu, Jun Min, Ziyu Yuan, Xiaorong Yang, Yanfeng Jiang, Tiejun Zhang, Chen Suo, Xiaojie Li, Lei Cheng, Zhenhua Zhang, Hongyu Niu, Zhe Li, Zhen Xie, Han Shi, Xiang Zhang, Min Fan, Xiaofeng Wang, Yajun Yang, Justin Dang, Catie McConnell, Juan Zhang, Jiucun Wang, Shunzhang Yu, Weimin Ye, Yuan Gao, Kun Zhang, Rui Liu, Li Jin, Xingdong Chen, Jeffrey Gole, Athurva Gore, Qiye He, Ming Lu, Jun Min, Ziyu Yuan, Xiaorong Yang, Yanfeng Jiang, Tiejun Zhang, Chen Suo, Xiaojie Li, Lei Cheng, Zhenhua Zhang, Hongyu Niu, Zhe Li, Zhen Xie, Han Shi, Xiang Zhang, Min Fan, Xiaofeng Wang, Yajun Yang, Justin Dang, Catie McConnell, Juan Zhang, Jiucun Wang, Shunzhang Yu, Weimin Ye, Yuan Gao, Kun Zhang, Rui Liu, Li Jin

Abstract

Early detection has the potential to reduce cancer mortality, but an effective screening test must demonstrate asymptomatic cancer detection years before conventional diagnosis in a longitudinal study. In the Taizhou Longitudinal Study (TZL), 123,115 healthy subjects provided plasma samples for long-term storage and were then monitored for cancer occurrence. Here we report the preliminary results of PanSeer, a noninvasive blood test based on circulating tumor DNA methylation, on TZL plasma samples from 605 asymptomatic individuals, 191 of whom were later diagnosed with stomach, esophageal, colorectal, lung or liver cancer within four years of blood draw. We also assay plasma samples from an additional 223 cancer patients, plus 200 primary tumor and normal tissues. We show that PanSeer detects five common types of cancer in 88% (95% CI: 80-93%) of post-diagnosis patients with a specificity of 96% (95% CI: 93-98%), We also demonstrate that PanSeer detects cancer in 95% (95% CI: 89-98%) of asymptomatic individuals who were later diagnosed, though future longitudinal studies are required to confirm this result. These results demonstrate that cancer can be non-invasively detected up to four years before current standard of care.

Conflict of interest statement

J.G., A.G., Q.H., J.M., X.L., L.C., Z.Z., H.N., Z.L., Z.X., H.S., J.D., C.M., and R.L. are employees of Singlera Genomics. Y.G. and R.L. are board members of Singlera Genomics. J.G., A.G., and R.L. are inventors on a patent (US62/657,544) held by Singlera Genomics that covers basic aspects of the library preparation method used in this paper. K.Z. is a co-founder, equity holder, and paid consultant of Singlera Genomics. The terms of these arrangements are being managed by the University of California–San Diego in accordance with its conflict of interest policies. X.C., M.L., Z.Y., X.Y., Y.J., T.Z., C.S., X.Z., M.F., X.W., Y.Y., J.Z., J.W., S.Y., W.Y., and L.J. declare no competing interests.

Figures

Fig. 1. Summary of the Taizhou longitudinal…
Fig. 1. Summary of the Taizhou longitudinal study (TZL).
The flowchart shows recruitment, baseline survey, sample collection, and cohort follow-up for TZL. Qualified pre-diagnosis patients and healthy participants were selected from the TZL cohort and qualified post-diagnosis patients were selected from local Taizhou hospital biobanks; 328 samples were processed but later excluded due to not meeting inclusion criteria or failing quality control metrics.
Fig. 2. Performance of PanSeer.
Fig. 2. Performance of PanSeer.
All presented results used only the test set samples. Dots represent the logistic regression (LR) score. a Receiver operator characteristic curves (ROC) and area under the curve (AUC) values for PanSeer. The red star shows the cutoff value derived from the training set. Separate curves are shown for post-diagnosis samples and pre-diagnosis samples (divided by years before diagnosis). b LR scores for PanSeer samples by years before diagnosis. c LR scores for PanSeer samples by cancer stage for post-diagnosis samples. d LR scores for PanSeer samples by tissue of origin for post-diagnosis samples. e LR scores for PanSeer samples by cancer stage at diagnosis for pre-diagnosis samples. f LR scores for PanSeer samples by tissue of origin for pre-diagnosis samples.
Fig. 3. Performance of PanSeer using only…
Fig. 3. Performance of PanSeer using only tissue-concordant genomic regions.
All presented results used only the test set samples, and only utilized target regions showing concordant hyper/hypo-methylation between training set cancer plasma samples and cancer tissue samples. Dots represent the logistic regression (LR) score. a Receiver operator characteristic curves (ROC) and area under the curve (AUC) values. The red star shows the cutoff value derived from the training set. Separate curves are shown for post-diagnosis samples and pre-diagnosis samples (divided by years before diagnosis). b LR scores by years before diagnosis.

References

    1. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65.
    1. Prigerson HG, et al. Chemotherapy use, performance status, and quality of life at the end of life. JAMA Oncol. 2015;1:778.
    1. World Health Organization. Guide to Early Cancer Diagnosis. (2017)
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J. Clin. 2018;68:7–30.
    1. Pickhardt PJ, Hassan C, Halligan S, Marmo R. Colorectal cancer: CT colonography and colonoscopy for detection—systematic review and meta-analysis. Radiology. 2011;259:393–405.
    1. Brawer MK. Prostate-specific antigen. Semin. Surg. Oncol. 2000;18:3–9.
    1. van den Biggelaar FJHM, Nelemans PJ, Flobbe K. Performance of radiographers in mammogram interpretation: a systematic review. Breast. 2008;17:85–90.
    1. Partridge EE, et al. Cervical cancer screening. J. Natl Compr. Canc. Netw. 2010;8:1358–1386.
    1. Pinsky PF, Prorok PC, Kramer BS. Prostate cancer screening—a perspective on the current state of the evidence. N. Engl. J. Med. 2017;376:1285–1289.
    1. Subramanian S, Klosterman M, Amonkar MM, Hunt TL. Adherence with colorectal cancer screening guidelines: a review. Prev. Med. 2004;38:536–550.
    1. PDQ Screening and Prevention Editorial Board. Cancer Screening Overview (PDQ®): Health Professional Version. PDQ Cancer Information Summaries (2002).
    1. Donaldson J, Park BH. Circulating tumor DNA: measurement and clinical utility. Annu. Rev. Med. 2018;69:223–234.
    1. Widschwendter M, et al. Methylation patterns in serum DNA for early identification of disseminated breast cancer. Genome Med. 2017;9:115.
    1. Widschwendter M, et al. The potential of circulating tumor DNA methylation analysis for the early detection and management of ovarian cancer. Genome Med. 2017;9:116.
    1. Cohen JD, et al. Detection and localization of surgically resectable cancers with a multi-analyte blood test. Science. 2018;359:926–930.
    1. Snyder MW, Kircher M, Hill AJ, Daza RM, Shendure J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 2016;164:57–68.
    1. Abelson S, et al. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature. 2018;559:400–404.
    1. Lanman RB, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS ONE. 2015;10:e0140712.
    1. Newman AM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 2016;34:547–555.
    1. Murtaza M, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112.
    1. Etzioni R, et al. Early detection: the case for early detection. Nat. Rev. Cancer. 2003;3:243–252.
    1. Aravanis AM, Lee M, Klausner RD. Next-generation sequencing of circulating tumor DNA for early cancer detection. Cell. 2017;168:571–574.
    1. Ferreira M. Early cancer detection via ctDNA may not add up. Clin. Omi. 2018;5:19–19.
    1. Laird PW. The power and the promise of DNA methylation markers. Nat. Rev. Cancer. 2003;3:253–266.
    1. Lu Y, et al. Methylated DNA/RNA in body fluids as biomarkers for lung cancer. Biol. Proced. Online. 2017;19:2.
    1. Diaz LA, Bardelli A, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J. Clin. Oncol. 2014;32:579–586.
    1. Wang X, et al. Rationales, design and recruitment of the Taizhou longitudinal study. BMC Public Health. 2009;9:223.
    1. Weisenberger DJ. Characterizing DNA methylation alterations from The Cancer Genome Atlas. J. Clin. Invest. 2014;124:17–23.
    1. Liu, L. et al. Targeted methylation sequencing of plasma cell-free DNA for cancer detection and classification. Ann. Oncol. 1–9, 10.1093/annonc/mdy119/4964860 (2018).
    1. Li W, et al. CancerDetector: ultrasensitive and non-invasive cancer detection at the resolution of individual reads using cell-free DNA methylation sequencing data. Nucleic Acids Res. 2018 doi: 10.1093/nar/gky423.
    1. Xu R, et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat. Mater. 2017;16:1155–1161.
    1. Hansen KD, et al. Increased methylation variation in epigenetic domains across cancer types. Nat. Genet. 2011;43:768–775.
    1. Hinoue T, et al. Genome-scale analysis of aberrant DNA methylation in colorectal cancer. Genome Res. 2012;22:271–282.
    1. Guo S, et al. Identification of methylation haplotype blocks aids in deconvolution of heterogeneous tissue samples and tumor tissue-of-origin mapping from plasma DNA. Nat. Genet. 2017;49:635–642.
    1. Gu H, et al. Genome-scale DNA methylation mapping of clinical samples at single-nucleotide resolution. Nat. Methods. 2010;7:133–136.
    1. Aigrain L, Gu Y, Quail MA. Quantitation of next generation sequencing library preparation protocol efficiencies using droplet digital PCR assays - a systematic comparison of DNA library preparation kits for Illumina sequencing. BMC Genomics. 2016;17:458.
    1. Meyer M, et al. A high coverage genome sequence from an archaic denisovan individual. Science. 2012;338:222.
    1. Gansauge M-T, Meyer M. Single-stranded DNA library preparation for the sequencing of ancient or damaged DNA. Nat. Protoc. 2013;8:737–748.
    1. Burnham P, et al. Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cell-free DNA in plasma. Sci. Rep. 2016;6:27859.
    1. Zheng Z, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat. Med. 2014;20:1479–1484.
    1. Warren JD, et al. Septin 9 methylated DNA is a sensitive and specific blood test for colorectal cancer. BMC Med. 2011;9:133.
    1. Klein EA, et al. Development of a comprehensive cell-free DNA (cfDNA) assay for early detection of multiple tumor types: The Circulating Cell-free Genome Atlas (CCGA) study. J. Clin. Oncol. 2018;36:12021–12021.
    1. Deng J, et al. Targeted bisulfite sequencing reveals changes in DNA methylation associated with nuclear reprogramming. Nat. Biotechnol. 2009;27:353–360.
    1. Shen SY, Burgener JM, Bratman SV, De Carvalho DD. Preparation of cfMeDIP-seq libraries for methylome profiling of plasma cell-free DNA. Nat. Protoc. 2019;14:2749–2780.
    1. Liu X, et al. Comprehensive DNA methylation analysis of tissue of origin of plasma cell-free DNA by methylated CpG tandem amplification and sequencing (MCTA-Seq) Clin. Epigenet. 2019;11:93.
    1. van Roon EH, et al. BRAF mutation-specific promoter methylation of FOX genes in colorectal cancer. Clin. Epigenet. 2013;5:2.
    1. Pilato B, et al. HOX gene methylation status analysis in patients with hereditary breast cancer. J. Hum. Genet. 2013;58:51–53.
    1. Homminga I, Pieters R, Meijerink JPP. NKL homeobox genes in leukemia. Leukemia. 2012;26:572–581.
    1. Li CG, Eccles MR. PAX genes in cancer; friends or foes? Front. Genet. 2012;3:6.
    1. Arribas J, et al. NF-κB mediates the expression of TBX15 in cancer cells. PLoS ONE. 2016;11:e0157761.
    1. deVos T, et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin. Chem. 2009;55:1337–1346.
    1. McLean CY, et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 2010;28:495–501.
    1. Chen W, et al. Cancer statistics in China. CA Cancer J. Clin. 2016;66:115–132.
    1. Teschendorff AE. Avoiding common pitfalls in machine learning omic data science. Nat. Mater. 2019;18:422–427.
    1. Shen SY, et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature. 2018;563:579–583.
    1. Lennon, A. M. et al. Feasibility of blood testing combined with PET-CT to screen for cancer and guide intervention. Science eabb9601, 10.1126/science.abb9601 (2020).
    1. Lam K, Pan K, Linnekamp JF, Medema JP, Kandimalla R. DNA methylation based biomarkers in colorectal cancer: A systematic review. Biochim. Biophys. Acta Rev. Cancer. 2016;1866:106–120.
    1. Liu, M. C. et al. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann. Oncol. 31, P745–759 10.1016/j.annonc.2020.04.013 (2020).
    1. Bormann F, et al. Cell-of-origin DNA methylation signatures are maintained during colorectal carcinogenesis. Cell Rep. 2018;23:3407–3418.
    1. Kakushadze Z, Raghubanshi R, Yu W. Estimating cost savings from early cancer diagnosis. Data. 2017;2:30.
    1. Taizhou Center for Disease Control and Prevention. . (Accessed 14 June 2019).
    1. Lu H, Zhao X, Zhang D. Analysis on mortality rate and years of life lost of cancer in Taizhou in 2010. China Cancer. 2010;5:360–364.
    1. Buderer NM. Statistical methodology: I. Incorporating the prevalence of disease into the sample size calculation for sensitivity and specificity. Acad. Emerg. Med. 1996;3:895–900.
    1. Jones SR, Carley S, Harrison M. An introduction to power and sample size estimation. Emerg. Med. J. 2003;20:453–458.

Source: PubMed

3
Subscribe