Validation of a liquid biopsy assay with molecular and clinical profiling of circulating tumor DNA

Justin D Finkle, Hala Boulos, Terri M Driessen, Christine Lo, Richard A Blidner, Ashraf Hafez, Aly A Khan, Ariane Lozac'hmeur, Kelly E McKinnon, Jason Perera, Wei Zhu, Afshin Dowlati, Kevin P White, Robert Tell, Nike Beaubier, Justin D Finkle, Hala Boulos, Terri M Driessen, Christine Lo, Richard A Blidner, Ashraf Hafez, Aly A Khan, Ariane Lozac'hmeur, Kelly E McKinnon, Jason Perera, Wei Zhu, Afshin Dowlati, Kevin P White, Robert Tell, Nike Beaubier

Abstract

Liquid biopsy is a valuable precision oncology tool that is increasingly used as a non-invasive approach to identify biomarkers, detect resistance mutations, monitor disease burden, and identify early recurrence. The Tempus xF liquid biopsy assay is a 105-gene, hybrid-capture, next-generation sequencing (NGS) assay that detects single-nucleotide variants, insertions/deletions, copy number variants, and chromosomal rearrangements. Here, we present extensive validation studies of the xF assay using reference standards, cell lines, and patient samples that establish high sensitivity, specificity, and accuracy in variant detection. The Tempus xF assay is highly concordant with orthogonal methods, including ddPCR, tumor tissue-based NGS assays, and another commercial plasma-based NGS assay. Using matched samples, we developed a dynamic filtering method to account for germline mutations and clonal hematopoiesis, while significantly decreasing the number of false-positive variants reported. Additionally, we calculated accurate circulating tumor fraction estimates (ctFEs) using the Off-Target Tumor Estimation Routine (OTTER) algorithm for targeted-panel sequencing. In a cohort of 1,000 randomly selected cancer patients who underwent xF testing, we found that ctFEs correlated with disease burden and clinical outcomes. These results highlight the potential of serial testing to monitor treatment efficacy and disease course, providing strong support for incorporating liquid biopsy in the management of patients with advanced disease.

Conflict of interest statement

J.D.F., H.B., T.M.D., C.L., R.A.B., A.H., A.A.K., A.L., K.E.M., J.P., W.Z., K.P.W., R.T. and N.B are employees and shareholders of Tempus Labs, Inc. The remaining authors declare no competing interests.

Figures

Fig. 1. Inter-assay comparison between Tempus xF,…
Fig. 1. Inter-assay comparison between Tempus xF, ddPCR, and Tempus xT results.
Patient samples with selected variants (n = 38) were analyzed by ddPCR and compared with xF variant allele fraction (VAF), resulting in high correlation overall (R2 = 0.892) (a), and in individual variants such as KRAS G12D (n = 12, R2 = 0.970) (b). c Results from the Tempus xF liquid biopsy and xT solid tumor assays were compared in patients who received both tests (n = 55) for colon, breast, and non-small cell lung cancers. The ctDNA VAFs for each variant are categorized by assay type in which they were detected and clonal hematopoiesis (CH) or germline status (top). The number of reportable variants for each individual patient are categorized by the assays in which they were detected (bottom). A total of 36 out of 55 xF samples had at least one pathogenic variant not detected in xT. d Among the 65 samples included in the microsatellite instability (MSI) validation cohort, 16 were deemed MSI-high and 49 microsatellite-stable. MSI was detected by xF in 6 out of 16 MSI-high patients, with 100% specificity (blue dots above dotted line).
Fig. 2. Circulating tumor fraction estimate (ctFE)…
Fig. 2. Circulating tumor fraction estimate (ctFE) and variant allele fraction (VAF).
ctFE of xF-sequenced patients (n = 1000) shows a modest correlation with max pathogenic (a) and median (c) VAF (ρ = 0.43 and 0.41, respectively) even after removing likely germline variants and variants that fall within an amplified region of the genome. A variant can be detected at or below the tumor fraction of the sample, so if the detected VAF was less than the OTTER ctFE it was considered consistent (blue). If the maximum or median VAF was within ±20% of the relative OTTER ctFE, or an absolute 0.02 of the fraction, then the sample was considered within tolerance (green). If the detected VAF was greater than the OTTER ctFE it was considered inconsistent (orange). b, d Cumulative fraction of samples with consistent VAF and OTTER estimates of tumor fraction as the ctFE increases. Overall, 84.7% of samples had OTTER ctFEs within tolerance or consistent with the maximal VAF detected in the sample (b). Overall, 94.1% of samples had OTTER ctFEs within tolerance or consistent with the medial VAF detected in the sample (d). e The distribution of ctFE across the cohort (median ctFE = 0.07, mean ctFE = 0.12, SD = 0.15). f In samples that also underwent low-pass whole-genome sequencing (LPWGS, n = 375), ichorCNA detected tumor fraction in just 165 samples (black). Among those samples, there was a strong correlation between LPWGS-predicted tumor fraction and OTTER ctFE (ρ = 0.890). In the majority of samples (60%, orange) with no ctFE detected by IchorCNA, we also detected a variant using the xF assay, indicating that there was detectable tumor DNA and the ichorCNA estimate was a false negative.
Fig. 3. Circulating tumor fraction estimate (ctFE)…
Fig. 3. Circulating tumor fraction estimate (ctFE) and mutational landscape by cancer type.
a Median ctFE among the most common cancer types was 0.07, with the exception of prostate (ctFE = 0.06). b The mutational landscape was evaluated in the xF 1000 cohort, with variants categorized as reportable, pathogenic, or actionable. Across all patients, the most commonly mutated gene was TP53. The heatmap was normalized within rows to depict the most prevalent variants detected for each common cancer type in the cohort (breast n = 254, colorectal n = 98, lung n = 241, prostate n = 96, and pancreatic n = 83).
Fig. 4. Circulating tumor fraction estimate (cfTE)…
Fig. 4. Circulating tumor fraction estimate (cfTE) according to stage and number of distant metastases.
a Among the xF 1000 cohort, there was a significant difference in ctFE between stages (Kruskal-Wallis P=2.97e−5). Overall, patients with stage 4 cancer (n = 879, median ctFE = 0.07) had a higher ctFE than those with stages 1 (n = 20, median ctFE = 0.06), 2 (n = 25, median ctFE = 0.06), or 3 (n = 76, median ctFE = 0.06). ctFE increased with the number of metastatic distant sites (Mann-Whitney U test P = 7.57e−7) (b), and there was a significant difference in ctFEs between patients with no metastatic lesions (n = 104) and those with 1 or more distant sites affected (n = 884, Mann–Whitney U test P = 5.21e−6) (c). d ROC and PR curves for calling metastases at different ctFE thresholds. Samples with and without identified metastases (n = 100) were selected to calculate the curves with balanced classes. AUROC=0.62 and AUPR=0.64. The dashed lines show expectations when selecting samples at random.
Fig. 5. Circulating tumor fraction estimate (cfTE)…
Fig. 5. Circulating tumor fraction estimate (cfTE) and abstracted clinical outcomes in a sub-cohort of the xF 1000 (n = 388).
a Patients with complete response (n = 9, ctFE = 0.05) exhibited lower ctFEs than those with progressive disease (n = 298, ctFE = 0.08), partial response (n = 56, ctFE = 0.06), or stable disease (n = 25, ctFE = 0.06). b ctFE was also assessed temporally among a few randomly selected patients with multiple xF tests throughout the course of treatment (n = 26), with most patients showing large differences in ctFEs between test dates.

References

    1. Leighl NB, et al. Clinical utility of comprehensive cell-free DNA analysis to identify genomic biomarkers in patients with newly diagnosed metastatic non-small cell lung cancer. Clin. Cancer Res. 2019;25:4691–4700. doi: 10.1158/1078-0432.CCR-19-0624.
    1. Aggarwal C, et al. Clinical implications of plasma-based genotyping with the delivery of personalized therapy in metastatic non-small cell lung cancer. JAMA Oncol. 2019;5:173–180. doi: 10.1001/jamaoncol.2018.4305.
    1. Kato, S. et al. Revisiting epidermal growth factor receptor (EGFR) amplification as a target for anti-EGFR therapy: analysis of cell-free circulating tumor DNA in patients with advanced malignancies. JCO Precis Oncol.10.1200/PO.18.00180 (2019).
    1. Lee J, et al. Detection of ERBB2 (HER2) gene amplification events in cell-free DNA and response to Anti-HER2 agents in a large asian cancer patient cohort. Front Oncol. 2019;9:212. doi: 10.3389/fonc.2019.00212.
    1. van Helden, E. J. et al. RAS and BRAF mutations in cell-free DNA are predictive for outcome of cetuximab monotherapy in patients with tissue-tested RAS wild-type advanced colorectal cancer. Mol. Oncol.10.1002/1878-0261.12550 (2019).
    1. Hyun MH, et al. Clinical implications of circulating cell-free DNA quantification and metabolic tumor burden in advanced non-small cell lung cancer. Lung Cancer. 2019;134:158–166. doi: 10.1016/j.lungcan.2019.06.014.
    1. Parikh AR, et al. Liquid versus tissue biopsy for detecting acquired resistance and tumor heterogeneity in gastrointestinal cancers. Nat. Med. 2019;25:1415–1421. doi: 10.1038/s41591-019-0561-9.
    1. Pailler E, et al. Acquired resistance mutations to ALK-inhibitors identified by single circulating tumor cell sequencing in ALK-rearranged non-small-cell lung cancer. Clin. Cancer Res. 2019;1176:2019.
    1. Jensen TJ, et al. Genome-wide sequencing of cell-free DNA identifies copy-number alterations that can be used for monitoring response to immunotherapy in cancer patients. Mol. Cancer Ther. 2019;18:448–458. doi: 10.1158/1535-7163.MCT-18-0535.
    1. Dal Maso, A. et al. Clinical features and progression pattern of acquired T790M-positive compared with T790M-negative EGFR mutant non-small-cell lung cancer: catching tumor and clinical heterogeneity over time through liquid biopsy. Clin. Lung Cancer10.1016/j.cllc.2019.07.009 (2019).
    1. Mayrhofer M, et al. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med. 2018;10:85. doi: 10.1186/s13073-018-0595-5.
    1. Hu Y, et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin. Cancer Res. 2018;24:4437–4443. doi: 10.1158/1078-0432.CCR-18-0143.
    1. Razavi P, et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat. Med. 2019;25:1928–1937. doi: 10.1038/s41591-019-0652-7.
    1. Adalsteinsson VA, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat. Commun. 2017;8:1324. doi: 10.1038/s41467-017-00965-y.
    1. Chen M, Zhao H. Next-generation sequencing in liquid biopsy: cancer screening and early detection. Hum. Genomics. 2019;13:34. doi: 10.1186/s40246-019-0220-8.
    1. Savli H, et al. TP53, EGFR and PIK3CA gene variations observed as prominent biomarkers in breast and lung cancer by plasma cell-free DNA genomic testing. J. Biotechnol. 2019;300:87–93. doi: 10.1016/j.jbiotec.2019.05.005.
    1. Cheng FT, et al. Liquid biopsy detects relapse five months earlier than regular clinical follow-up and guides targeted treatment in breast cancer. Case Rep. Oncol. Med. 2019;2019:6545298.
    1. Keup, C. et al. Targeted deep sequencing revealed variants in cell-free DNA of hormone receptor-positive metastatic breast cancer patients. Cell Mol. Life Sci.10.1007/s00018-019-03189-z (2019).
    1. Li G, et al. Genomic profiling of cell-free circulating tumor DNA in patients with colorectal cancer and its fidelity to the genomics of the tumor biopsy. J. Gastrointest. Oncol. 2019;10:831–840. doi: 10.21037/jgo.2019.05.05.
    1. Colleoni M, et al. Annual hazard rates of recurrence for breast cancer during 24 years of follow-up: results from the international breast cancer study group trials I to V. J. Clin. Oncol. 2016;34:927–935. doi: 10.1200/JCO.2015.62.3504.
    1. Yates LR, et al. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell. 2017;32:169–184 e167. doi: 10.1016/j.ccell.2017.07.005.
    1. Uramoto H, Tanaka F. Recurrence after surgery in patients with NSCLC. Transl. Lung Cancer Res. 2014;3:242–249.
    1. Taunk NK, et al. Immunotherapy and radiation therapy for operable early stage and locally advanced non-small cell lung cancer. Transl. Lung Cancer Res. 2017;6:178–185. doi: 10.21037/tlcr.2017.03.05.
    1. Coombes RC, et al. Personalized detection of circulating tumor DNA antedates breast cancer metastatic recurrence. Clin. Cancer Res. 2019;25:4255–4263. doi: 10.1158/1078-0432.CCR-18-3663.
    1. Tie, J. et al. Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage III colon cancer. JAMA Oncol.10.1001/jamaoncol.2019.3616 (2019).
    1. McEvoy AC, et al. Monitoring melanoma recurrence with circulating tumor DNA: a proof of concept from three case studies. Oncotarget. 2019;10:113–122. doi: 10.18632/oncotarget.26451.
    1. Christensen E, et al. Early detection of metastatic relapse and monitoring of therapeutic efficacy by ultra-deep sequencing of plasma cell-free DNA in patients with urothelial bladder carcinoma. J. Clin. Oncol. 2019;37:1547–1557. doi: 10.1200/JCO.18.02052.
    1. Isaksson S, et al. Pre-operative plasma cell-free circulating tumor DNA and serum protein tumor markers as predictors of lung adenocarcinoma recurrence. Acta Oncol. 2019;58:1079–1086. doi: 10.1080/0284186X.2019.1610573.
    1. Bettegowda C, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014;6:224ra224. doi: 10.1126/scitranslmed.3007094.
    1. Xue VW, Wong CSC, Cho WCS. Early detection and monitoring of cancer in liquid biopsy: advances and challenges. Expert Rev. Mol. Diagn. 2019;19:273–276. doi: 10.1080/14737159.2019.1583104.
    1. Hennigan, S. T. et al. Low Abundance of circulating tumor DNA in localized prostate cancer. JCO Precis. Oncol.10.1200/PO.19.00176 (2019).
    1. Abbosh C, Birkbak NJ, Swanton C. Early stage NSCLC - challenges to implementing ctDNA-based screening and MRD detection. Nat. Rev. Clin. Oncol. 2018;15:577–586. doi: 10.1038/s41571-018-0058-3.
    1. Cheng J, et al. Clinical validation of a cell-free DNA gene panel. J. Mol. Diagn. 2019;21:632–645. doi: 10.1016/j.jmoldx.2019.02.008.
    1. Hanibuchi M, et al. Development, validation, and comparison of gene analysis methods for detecting EGFR mutation from non-small cell lung cancer patients-derived circulating free DNA. Oncotarget. 2019;10:3654–3666. doi: 10.18632/oncotarget.26951.
    1. Van Laar R, Lincoln M, Van Laar B. Development and validation of a plasma-based melanoma biomarker suitable for clinical use. Br. J. Cancer. 2018;118:857–866. doi: 10.1038/bjc.2017.477.
    1. Odegaard JI, et al. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin. Cancer Res. 2018;24:3539–3549. doi: 10.1158/1078-0432.CCR-17-3831.
    1. Clark TA, et al. Analytical validation of a hybrid capture-based next-generation sequencing clinical assay for genomic profiling of cell-free circulating tumor DNA. J. Mol. Diagn. 2018;20:686–702. doi: 10.1016/j.jmoldx.2018.05.004.
    1. Plagnol V, et al. Analytical validation of a next generation sequencing liquid biopsy assay for high sensitivity broad molecular profiling. PLoS One. 2018;13:e0193802. doi: 10.1371/journal.pone.0193802.
    1. Kuderer NM, et al. Comparison of 2 commercially available next-generation sequencing platforms in oncology. JAMA Oncol. 2017;3:996–998. doi: 10.1001/jamaoncol.2016.4983.
    1. Beaubier N, et al. Integrated genomic profiling expands clinical options for patients with cancer. Nat. Biotechnol. 2019;37:1351–1360. doi: 10.1038/s41587-019-0259-z.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Lai Z, et al. VarDict: a novel and versatile variant caller for next-generation sequencing in cancer research. Nucleic Acids Res. 2016;44:e108–e108. doi: 10.1093/nar/gkw227.
    1. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 2016;12:e1004873–e1004873. doi: 10.1371/journal.pcbi.1004873.
    1. Chiang C, et al. SpeedSeq: ultra-fast personal genome analysis and interpretation. Nat. Methods. 2015;12:966–968. doi: 10.1038/nmeth.3505.
    1. Layer RM, Chiang C, Quinlan AR, Hall IM. LUMPY: a probabilistic framework for structural variant discovery. Genome Biol. 2014;15:R84–R84. doi: 10.1186/gb-2014-15-6-r84.
    1. Li MM, et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a joint consensus recommendation of the association for molecular pathology, American society of clinical oncology, and college of American pathologists. J. Mol. diagnostics: JMD. 2017;19:4–23. doi: 10.1016/j.jmoldx.2016.10.002.
    1. Kalia SS, et al. Recommendations for reporting of secondary findings in clinical exome and genome sequencing, 2016 update (ACMG SF v2.0): a policy statement of the American College of Medical Genetics and Genomics. Genet. Med. 2017;19:249–255. doi: 10.1038/gim.2016.190.
    1. Shen R, Seshan VE. FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016;44:e131. doi: 10.1093/nar/gkw520.
    1. Olshen AB, Venkatraman ES, Lucito R, Wigler M. Circular binary segmentation for the analysis of array‐based DNA copy number data. Biostatistics. 2004;5:557–572. doi: 10.1093/biostatistics/kxh008.
    1. Finkle, J. D. et al. Metadata record for the manuscript: Analytical validation of a targeted liquid biopsy assay with comprehensive molecular and clinical profiling of circulating tumor DNA from 1000 patients. figshare 10.6084/m9.figshare.14672673 (2021).
    1. Vivli (2021).

Source: PubMed

3
購読する