Longitudinal Circulating Tumor DNA Analysis in Blood and Saliva for Prediction of Response to Osimertinib and Disease Progression in EGFR-Mutant Lung Adenocarcinoma

Chul Kim, Liqiang Xi, Constance M Cultraro, Fang Wei, Gregory Jones, Jordan Cheng, Ahmad Shafiei, Trinh Hoc-Tran Pham, Nitin Roper, Elizabeth Akoth, Azam Ghafoor, Vikram Misra, Nina Monkash, Charles Strom, Michael Tu, Wei Liao, David Chia, Clive Morris, Seth M Steinberg, Hadi Bagheri, David T W Wong, Mark Raffeld, Udayan Guha, Chul Kim, Liqiang Xi, Constance M Cultraro, Fang Wei, Gregory Jones, Jordan Cheng, Ahmad Shafiei, Trinh Hoc-Tran Pham, Nitin Roper, Elizabeth Akoth, Azam Ghafoor, Vikram Misra, Nina Monkash, Charles Strom, Michael Tu, Wei Liao, David Chia, Clive Morris, Seth M Steinberg, Hadi Bagheri, David T W Wong, Mark Raffeld, Udayan Guha

Abstract

Background: We assessed whether serial ctDNA monitoring of plasma and saliva predicts response and resistance to osimertinib in EGFR-mutant lung adenocarcinoma. Three ctDNA technologies-blood-based droplet-digital PCR (ddPCR), next-generation sequencing (NGS), and saliva-based EFIRM liquid biopsy (eLB)-were employed to investigate their complementary roles. Methods: Plasma and saliva samples were collected from patients enrolled in a prospective clinical trial of osimertinib and local ablative therapy upon progression (NCT02759835). Plasma was analyzed by ddPCR and NGS. Saliva was analyzed by eLB. Results: A total of 25 patients were included. We analyzed 534 samples by ddPCR (n = 25), 256 samples by NGS (n = 24) and 371 samples by eLB (n = 22). Among 20 patients who progressed, ctDNA progression predated RECIST 1.1 progression by a median of 118 days (range: 61-272 days) in 11 (55%) patients. Of nine patients without ctDNA progression by ddPCR, two patients had an increase in mutant EGFR by eLB and two patients were found to have ctDNA progression by NGS. Levels of ctDNA measured by ddPCR and NGS at early time points, but not volumetric tumor burden, were associated with PFS. EGFR/ERBB2/MET/KRAS amplifications, EGFR C797S, PIK3CA E545K, PTEN V9del, and CTNNB1 S45P were key resistance mechanisms identified by NGS. Conclusion: Serial assessment of ctDNA in plasma and saliva predicts response and resistance to osimertinib, with each assay having supplementary roles.

Keywords: EGFR; NSCLC; ctDNA; osimertinib.

Conflict of interest statement

Chul Kim receives research funding from his current institution (Georgetown University), AstraZeneca, Novartis, Debiopharm, Genentech, Karyopharm, Regeneron, Janssen, BMS, and Mirati. Chul Kim also served as a consultant for Novartis, Janssen, and PierianDx Oncology. Clive Morris and Greg Jones are employees of Inivata. Charles Strom, Michael Tu, and Wei Liao are employees of EZLife Bio Inc. David T.W. Wong receives research grant funding from NIH, Colgate-Palmolive, and Hirshberg Foundation. Udayan Guha has a clinical trial agreement (CTA) with AstraZeneca for the current study and receives research funding from AstraZeneca, Esanex, and Aurigene. David T.W. Wong has equity in Liquid Diagnostics. The other authors have no conflicts of interest to report.

Figures

Figure 1
Figure 1
Clinical protocol schema.
Figure 2
Figure 2
(A,B) Spearman correlation between EGFR mutation AF by ddPCR and tumor volume (A). Spearman correlation between EGFR mutation AF by NGS and tumor volume (B).
Figure 3
Figure 3
(A–C) Correlations between ddPCR, NGS, and EFIRM liquid biopsy (eLB) assays.
Figure 4
Figure 4
(A–O) Serial monitoring of ctDNA by ddPCR, NGS, and eLB for prediction of response and the detection of emergence of resistance. Solid lines represent genomic alterations and red dotted lines represent calculated tumor volume measured by volumetric CT measurement. AFs for ddPCR and NGS and current values for eLB are plotted on the left y-axis. Calculated tumor volume measured by volumetric CT measurement is plotted on the right T-axis. Treatment duration is plotted on the x-axis. The shaded gray areas indicate the duration of RECIST 1.1 response (complete or partial response). ctDNA progression, defined as increases in mutant EGFR AF by ddPCR, preceded RECIST 1.1 progression by a median of 118 days (range: 61–272 days) in 11 patients (LAT001, 002, 003, 005, 010, 013, 014, 015, 017, 023, 028). Of the 10 patients without ctDNA progression by ddPCR, 2 patients had an increase in EGFR mutation-level by eLB (LAT006, LAT026), 1 patient had an increase in the AF of PTEN Y88* by NGS (LAT007), and another patient had an increase in TP53 V157F by NGS (LAT016). The arrowhead (in black) indicates the beginning of ctDNA progression.
Figure 4
Figure 4
(A–O) Serial monitoring of ctDNA by ddPCR, NGS, and eLB for prediction of response and the detection of emergence of resistance. Solid lines represent genomic alterations and red dotted lines represent calculated tumor volume measured by volumetric CT measurement. AFs for ddPCR and NGS and current values for eLB are plotted on the left y-axis. Calculated tumor volume measured by volumetric CT measurement is plotted on the right T-axis. Treatment duration is plotted on the x-axis. The shaded gray areas indicate the duration of RECIST 1.1 response (complete or partial response). ctDNA progression, defined as increases in mutant EGFR AF by ddPCR, preceded RECIST 1.1 progression by a median of 118 days (range: 61–272 days) in 11 patients (LAT001, 002, 003, 005, 010, 013, 014, 015, 017, 023, 028). Of the 10 patients without ctDNA progression by ddPCR, 2 patients had an increase in EGFR mutation-level by eLB (LAT006, LAT026), 1 patient had an increase in the AF of PTEN Y88* by NGS (LAT007), and another patient had an increase in TP53 V157F by NGS (LAT016). The arrowhead (in black) indicates the beginning of ctDNA progression.
Figure 4
Figure 4
(A–O) Serial monitoring of ctDNA by ddPCR, NGS, and eLB for prediction of response and the detection of emergence of resistance. Solid lines represent genomic alterations and red dotted lines represent calculated tumor volume measured by volumetric CT measurement. AFs for ddPCR and NGS and current values for eLB are plotted on the left y-axis. Calculated tumor volume measured by volumetric CT measurement is plotted on the right T-axis. Treatment duration is plotted on the x-axis. The shaded gray areas indicate the duration of RECIST 1.1 response (complete or partial response). ctDNA progression, defined as increases in mutant EGFR AF by ddPCR, preceded RECIST 1.1 progression by a median of 118 days (range: 61–272 days) in 11 patients (LAT001, 002, 003, 005, 010, 013, 014, 015, 017, 023, 028). Of the 10 patients without ctDNA progression by ddPCR, 2 patients had an increase in EGFR mutation-level by eLB (LAT006, LAT026), 1 patient had an increase in the AF of PTEN Y88* by NGS (LAT007), and another patient had an increase in TP53 V157F by NGS (LAT016). The arrowhead (in black) indicates the beginning of ctDNA progression.
Figure 5
Figure 5
(A–E) Progression free survival (PFS) based on the baseline mutant EGFR ctDNA copy number, its clearance on therapy at early time periods on treatment, and baseline tumor volume assessed by volumetric CT. PFS based on mutant EGFR copy number at baseline and on day 21/28 by ddPCR (A,B), EGFR AF at baseline and day 7 by NGS (C,D), calculated tumor volume at baseline (E), and difference in tumor volume between day 42/46 vs. baseline (F).
Figure 6
Figure 6
Detection of co-occurring genomic alterations and resistance mechanisms by NGS. Each row represents genomic alterations and columns indicate each patient. Co-occurring mutations at baseline prior to exposure to osimertinib are color-coded in green and genomic alterations seen at progression are in dark purple. Germline mutations are shown in gray. Patients whose disease did not progress at the time of analysis are color-coded in blue (LAT004, LAT008, LAT019, LAT024).

References

    1. Kim C., Giaccone G. Precision oncology in non-small-cell lung cancer: Opportunities and challenges. Nat. Rev. Clin. Oncol. 2018;15:348–349. doi: 10.1038/s41571-018-0008-0.
    1. Heerink W.J., de Bock G.H., de Jonge G.J., Groen H.J., Vliegenthart R., Oudkerk M. Complication rates of CT-guided transthoracic lung biopsy: Meta-Analysis. Eur. Radiol. 2017;27:138–148. doi: 10.1007/s00330-016-4357-8.
    1. Gerlinger M., Rowan A.J., Horswell S., Math M., Larkin J., Endesfelder D., Gronroos E., Martinez P., Matthews N., Stewart A., et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012;366:883–892. doi: 10.1056/NEJMoa1113205.
    1. Roper N., Brown A.L., Wei J.S., Pack S., Trindade C., Kim C., Restifo O., Gao S., Sindiri S., Mehrabadi F., et al. Clonal Evolution and Heterogeneity of Osimertinib Acquired Resistance Mechanisms in EGFR Mutant Lung Cancer. Cell. Rep. Med. 2020;1 doi: 10.1016/j.xcrm.2020.100007.
    1. Diaz L.A., Jr., Bardelli A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011.
    1. Bettegowda C., Sausen M., Leary R.J., Kinde I., Wang Y., Agrawal N., Bartlett B.R., Wang H., Luber B., Alani R.M., et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014;6:224ra24. doi: 10.1126/scitranslmed.3007094.
    1. Murtaza M., Dawson S.J., Tsui D.W., Gale D., Forshew T., Piskorz A.M., Parkinson C., Chin S.F., Kingsbury Z., Wong A.S., et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112. doi: 10.1038/nature12065.
    1. Oxnard G.R., Paweletz C.P., Kuang Y., Mach S.L., O’Connell A., Messineo M.M., Luke J.J., Butaney M., Kirschmeier P., Jackman D.M., et al. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin. Cancer. Res. 2014;20:1698–1705. doi: 10.1158/1078-0432.CCR-13-2482.
    1. Forshew T., Murtaza M., Parkinson C., Gale D., Tsui D.W., Kaper F., Dawson S.J., Piskorz A.M., Jimenez-Linan M., Bentley D., et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 2012;4:136ra168. doi: 10.1126/scitranslmed.3003726.
    1. Pritchett M.A., Camidge D.R., Patel M., Khatri J., Boniol S., Friedman E.K., Khomani A., Dalia S., Baker-Neblett K., Plagnol V. Prospective clinical validation of the InVisionFirst-Lung circulating tumor DNA assay for molecular profiling of patients with advanced nonsquamous non–small-cell lung cancer. JCO Precis. Oncol. 2019;3:1–15. doi: 10.1200/PO.18.00299.
    1. Thress K.S., Markovets A., Barrett J.C., Chmielecki J., Goldberg S.B., Shepherd F.A., Vowler S., Oxnard G.R. Complete clearance of plasma EGFR mutations as a predictor of outcome on osimertinib in the AURA trial. Am. Soc. Clin. Oncol. 2017;35:9018. doi: 10.1200/JCO.2017.35.15_suppl.9018.
    1. Sacher A.G., Paweletz C., Dahlberg S.E., Alden R.S., O’Connell A., Feeney N., Mach S.L., Janne P.A., Oxnard G.R. Prospective Validation of Rapid Plasma Genotyping for the Detection of EGFR and KRAS Mutations in Advanced Lung Cancer. JAMA Oncol. 2016;2:1014–1022. doi: 10.1001/jamaoncol.2016.0173.
    1. He J., Tan W., Tang X., Ma J. Variations in EGFR ctDNA Correlates to the Clinical Efficacy of Afatinib in Non Small Cell Lung Cancer with Acquired Resistance. Pathol. Oncol. Res. 2017;23:307–315. doi: 10.1007/s12253-016-0097-y.
    1. Mok T., Wu Y.L., Lee J.S., Yu C.J., Sriuranpong V., Sandoval-Tan J., Ladrera G., Thongprasert S., Srimuninnimit V., Liao M., et al. Detection and Dynamic Changes of EGFR Mutations from Circulating Tumor DNA as a Predictor of Survival Outcomes in NSCLC Patients Treated with First-line Intercalated Erlotinib and Chemotherapy. Clin. Cancer. Res. 2015;21:3196–3203. doi: 10.1158/1078-0432.CCR-14-2594.
    1. Riediger A.L., Dietz S., Schirmer U., Meister M., Heinzmann-Groth I., Schneider M., Muley T., Thomas M., Sultmann H. Mutation analysis of circulating plasma DNA to determine response to EGFR tyrosine kinase inhibitor therapy of lung adenocarcinoma patients. Sci. Rep. 2016;6:33505. doi: 10.1038/srep33505.
    1. Kato K., Uchida J., Kukita Y., Kumagai T., Nishino K., Inoue T., Kimura M., Oba S., Imamura F. Numerical indices based on circulating tumor DNA for the evaluation of therapeutic response and disease progression in lung cancer patients. Sci. Rep. 2016;6:29093. doi: 10.1038/srep29093.
    1. Moran T., Felip E., Bosch-Barrera J., de Aguirre I., Ramirez J.L., Mesia C., Carcereny E., Roa D., Sais E., Garcia Y., et al. Monitoring EGFR-T790M mutation in serum/plasma for prediction of response to third-generation EGFR inhibitors in patients with lung cancer. Oncotarget. 2018;9:27074–27086. doi: 10.18632/oncotarget.25478.
    1. Xiong L., Cui S., Ding J., Sun Y., Zhang L., Zhao Y., Gu A., Chu T., Wang H., Zhong H., et al. Dynamics of EGFR mutations in plasma recapitulates the clinical response to EGFR-TKIs in NSCLC patients. Oncotarget. 2017;8:63846–63856. doi: 10.18632/oncotarget.19139.
    1. Wei F., Lin C.C., Joon A., Feng Z., Troche G., Lira M.E., Chia D., Mao M., Ho C.L., Su W.C., et al. Noninvasive saliva-based EGFR gene mutation detection in patients with lung cancer. Am. J. Respir. Crit. Care. Med. 2014;190:1117–1126. doi: 10.1164/rccm.201406-1003OC.
    1. Wei F., Strom C.M., Cheng J., Lin C.C., Hsu C.Y., Soo Hoo G.W., Chia D., Kim Y., Li F., Elashoff D., et al. Electric Field-Induced Release and Measurement Liquid Biopsy for Noninvasive Early Lung Cancer Assessment. J. Mol. Diagn. 2018;20:738–742. doi: 10.1016/j.jmoldx.2018.06.008.
    1. Li F., Wei F., Huang W.L., Lin C.C., Li L., Shen M.M., Yan Q., Liao W., Chia D., Tu M., et al. Ultra-Short Circulating Tumor DNA (usctDNA) in Plasma and Saliva of Non-Small Cell Lung Cancer (NSCLC) Patients. Cancers. 2020;12:2041. doi: 10.3390/cancers12082041.
    1. Gale D., Lawson A.R.J., Howarth K., Madi M., Durham B., Smalley S., Calaway J., Blais S., Jones G., Clark J., et al. Development of a highly sensitive liquid biopsy platform to detect clinically-relevant cancer mutations at low allele fractions in cell-free DNA. PLoS ONE. 2018;13:e0194630. doi: 10.1371/journal.pone.0194630.
    1. Plagnol V., Woodhouse S., Howarth K., Lensing S., Smith M., Epstein M., Madi M., Smalley S., Leroy C., Hinton J., 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. Ramalingam S.S., Vansteenkiste J., Planchard D., Cho B.C., Gray J.E., Ohe Y., Zhou C., Reungwetwattana T., Cheng Y., Chewaskulyong B., et al. Overall Survival with Osimertinib in Untreated, EGFR-Mutated Advanced NSCLC. N. Engl. J. Med. 2020;382:41–50. doi: 10.1056/NEJMoa1913662.
    1. Shepherd F.A., Papadimitrakopoulou V., Mok T., Wu Y.-L., Han J.-Y., Ahn M.-J., Ramalingam S.S., John T., Sebastian M., Theelen W., et al. Early clearance of plasma EGFR mutations as a predictor of response to osimertinib in the AURA3 trial. J. Clin. Oncol. 2018;36:9027. doi: 10.1200/JCO.2018.36.15_suppl.9027.
    1. Zhou C., Imamura F., Cheng Y., Okamoto I., Cho B.C., Lin M.C., Majem M., Gautschi O., Gray J.E., Boyer M.J., et al. Early clearance of plasma EGFR mutations as a predictor of response to osimertinib and comparator EGFR-TKIs in the FLAURA trial. J. Clin. Oncol. 2019;37:9020. doi: 10.1200/JCO.2019.37.15_suppl.9020.
    1. Hartmaier R., Han J.-Y., Ahn M.-J., Cho B.C., Cantarini M., Frewer P., Frigault M.M., Oxnard G. Abstract CT303: The Effect of Savolitinib Plus Osimertinib on ctDNA Clearance in Patients with EGFR Mutation Positive (EGFRm) MET-Amplified NSCLC in the TATTON Study. AACR; Philadelphia, PA, USA: 2020.
    1. Gray J.E., Okamoto I., Sriuranpong V., Vansteenkiste J., Imamura F., Lee J.S., Pang Y.K., Cobo M., Kasahara K., Cheng Y., et al. Tissue and Plasma EGFR Mutation Analysis in the FLAURA Trial: Osimertinib versus Comparator EGFR Tyrosine Kinase Inhibitor as First-Line Treatment in Patients with EGFR-Mutated Advanced Non-Small Cell Lung Cancer. Clin. Cancer Res. 2019;25:6644–6652. doi: 10.1158/1078-0432.CCR-19-1126.
    1. Nishino M., Dahlberg S.E., Cardarella S., Jackman D.M., Rabin M.S., Hatabu H., Janne P.A., Johnson B.E. Tumor volume decrease at 8 weeks is associated with longer survival in EGFR-mutant advanced non-small-cell lung cancer patients treated with EGFR TKI. J. Thorac. Oncol. 2013;8:1059–1068. doi: 10.1097/JTO.0b013e318294c909.
    1. Nishino M., Dahlberg S.E., Fulton L.E., Digumarthy S.R., Hatabu H., Johnson B.E., Sequist L.V. Volumetric Tumor Response and Progression in EGFR-mutant NSCLC Patients Treated with Erlotinib or Gefitinib. Acad. Radiol. 2016;23:329–336. doi: 10.1016/j.acra.2015.11.005.
    1. Bardelli A., Pantel K. Liquid biopsies, what we do not know (yet) Cancer Cell. 2017;31:172–179. doi: 10.1016/j.ccell.2017.01.002.
    1. Postel M., Roosen A., Laurent-Puig P., Taly V., Wang-Renault S.F. Droplet-based digital PCR and next generation sequencing for monitoring circulating tumor DNA: A cancer diagnostic perspective. Expert. Rev. Mol. Diagn. 2018;18:7–17. doi: 10.1080/14737159.2018.1400384.
    1. Blakely C.M., Watkins T.B.K., Wu W., Gini B., Chabon J.J., McCoach C.E., McGranahan N., Wilson G.A., Birkbak N.J., Olivas V.R., et al. Evolution and clinical impact of co-occurring genetic alterations in advanced-stage EGFR-mutant lung cancers. Nat. Genet. 2017;49:1693–1704. doi: 10.1038/ng.3990.
    1. Lim S., Yang S., Lim S., Heo S., Daniel S., Markovets A., Rafati M., Park C., Yun J., Pyo K. P76. 18 Tissue-and Plasma-Based Landscape of Resistance to Osimertinib. J. Thorac. Oncol. 2021;16:S593–S594. doi: 10.1016/j.jtho.2021.01.1075.
    1. Arasada R.R., Shilo K., Yamada T., Zhang J., Yano S., Ghanem R., Wang W., Takeuchi S., Fukuda K., Katakami N., et al. Notch3-dependent beta-catenin signaling mediates EGFR TKI drug persistence in EGFR mutant NSCLC. Nat. Commun. 2018;9:3198. doi: 10.1038/s41467-018-05626-2.
    1. Zhao J., Lin G., Zhuo M., Fan Z., Miao L., Chen L., Zeng A., Yin R., Ou Y., Shi Z., et al. Next-generation sequencing based mutation profiling reveals heterogeneity of clinical response and resistance to osimertinib. Lung Cancer. 2020;141:114–118. doi: 10.1016/j.lungcan.2019.10.021.

Source: PubMed

3
Subscribe