Intra-patient stability of tumor mutational burden from tissue biopsies at different time points in advanced cancers

Timothy V Pham, Aaron M Goodman, Smruthy Sivakumar, Garrett Frampton, Razelle Kurzrock, Timothy V Pham, Aaron M Goodman, Smruthy Sivakumar, Garrett Frampton, Razelle Kurzrock

Abstract

Background: Tumor mutational burden (TMB) may be a predictive biomarker of immune checkpoint inhibitor (ICI) responsiveness. Genomic landscape heterogeneity is a well-established cancer feature. Molecular characteristics may differ even within the same tumor specimen and undoubtedly evolve with time. However, the degree to which TMB differs between tumor biopsies within the same patient has not been established.

Methods: We curated data on 202 patients enrolled in the PREDICT study (NCT02478931), seen at the University of California San Diego (UCSD), who had 404 tissue biopsies for TMB (two per patient, mean of 722 days between biopsies) to assess difference in TMB before and after treatment in a pan-cancer cohort. We also performed an orthogonal analysis of 2872 paired pan-solid tumor biopsies in the Foundation Medicine database to examine difference in TMB between first and last biopsies.

Results: The mean (95% CI) TMB difference between samples was 0.583 [- 0.900-2.064] (p = 0.15). Pearson correlation showed a flat line for time elapsed between biopsies versus TMB change indicating no correlation (R2 = 0.0001; p = 0.8778). However, in 55 patients who received ICIs, there was an increase in TMB (before versus after mean mutations/megabase [range] 12.50 [range, 0.00-98.31] versus 14.14 [range, 0.00-100.0], p = 0.025). Analysis of 2872 paired pan-solid tumor biopsies in the Foundation Medicine database also indicated largely stable TMB patterns; TMB increases were only observed in specific tumors (e.g., breast, colorectal, glioma) within certain time intervals.

Conclusions: Overall, our results suggest that tissue TMB remains stable with time, though specific therapies such as immunotherapy may correlate with an increase in TMB.

Trial registration: NCT02478931 , registered June 23, 2015.

Keywords: Immunotherapy; Immunotherapy effect on TMB; TMB over time; TMB over treatment; Tumor mutational burden.

Conflict of interest statement

RK has the following disclosure information: Stock and Other Equity Interests (IDbyDNA, CureMatch, Inc., and Soluventis); consulting or advisory role (Gaido, LOXO, X-Biotech, Actuate Therapeutics, Roche, NeoMed, Soluventis, and Pfizer); speaker’s fee (Roche); research funding (Incyte, Genentech, Merck Serono, Pfizer, Sequenom, Foundation Medicine, Guardant Health, Grifols, Konica Minolta, DeBiopharm, Boerhringer Ingelheim, and OmniSeq [all institutional]); board member (CureMatch, Inc and CureMetrix Inc.).

AG receives consulting fees from Seattle Genetics and EUSA Pharma.

GF and SS are employees at Foundation Medicine, Inc., a wholly owned subsidiary of Roche Holdings, Inc. and Roche Finance Ltd, and have equity interest in an affiliate of these Roche entities.

The remaining author, TP, declares that he has no competing interests.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
UCSD TMB differences. All patients with valid UCSD TMB data (n = 202) were considered for this figure. Exposure or treatment is defined as having received agent between the initial and final biopsies, regardless of dose or duration, as per electronic medical records. A The TMB of the earlier and later biopsies are somewhat correlated with a significant, almost unity, slope (p < 0.0001), and a Pearson R2 = 0.459. This indicates there is no difference between early and later TMBs. B TMB difference does not correlate with time elapsed between biopsies (Pearson correlation R2 = 0.0001). The slope of the line of best fit is also not significantly greater than zero (p = 0.8778). C There is no significant different between TMBs measured at different times as determined by the Wilcoxon matched pairs signed rank test. Red lines are mean (center) ± 95% confidence interval (CI) for each group. The average TMB of the earlier biopsy was 6.181 [4.405–7.957] mutations/Mb versus 6.764 [4.861–8.666] mutations/Mb for the later biopsy (p = 0.1467). D TMB difference (increase) with time in immunotherapy-treated patients is greater than in those who did not receive immunotherapy (immunotherapy response not considered). All patients with valid TMB data considered. Red lines are mean (center) ± 95% CI for each group. Patients who were treated with immunotherapy between biopsies had a mean ± 95% CI TMB change of 1.641 [− 3.492–6.775] mutations/Mb whereas those who were not had a mean ± 95% CI TMB change of 0.1857 [− 0.5974–0.9688] mutations/Mb (p = 0.0365). Drugs received included the following: ipilimumab, nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, and cemiplimab
Fig. 2
Fig. 2
Foundation Medicine TMB differences in categorical elapsed time. A total of 2872 paired tissue biopsy samples from the Foundation Medicine database were studied for patterns of TMB change based on time between tests. Tumor types with at least 50 pairs were considered for this figure. Each panel represents a specific tumor type with the total number of assessed samples denoted by “N.” The collection time differences were binned into three categories: ≤ 365 days, 366 to 1095 days, and > 1095 days. Boxplots of the TMB change were plotted for each bin of collection time difference, with the total number of pairs in each bin denoted by “n.” In each boxplot: the horizontal line represents the median, the box represents the interquartile range (IQR) and the whiskers represent extremes of the data (capped at 1.5xIQR). Wilcoxon test for statistical difference for each pairwise comparison was performed; the p value is presented based on different thresholds (*0.05, **0.01, ***0.001, NS.: not significant). The TMB difference (y-axis) is capped to be between − 15 and 15 for better visualization; however, statistical analysis was performed on all available samples. Individual data points for TMB change, colored by the categorical time between biopsy, are available in Additional file 1: Fig. S4. See Additional file 2: Table S1 for detailed data and for false discovery rate adjusted p value for multiple comparisons

References

    1. Goodman AM, Kato S, Bazhenova L, Patel SP, Frampton GM, Miller V, Stephens PJ, Daniels GA, Kurzrock R. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol Cancer Ther. 2017;16(11):2598–2608. doi: 10.1158/1535-7163.MCT-17-0386.
    1. Wu Y, Xu J, Du C, Wu Y, Xia D, Lv W, et al. The predictive value of tumor mutation burden on efficacy of immune checkpoint inhibitors in cancers: a systematic review and meta-analysis. Front Oncol. 2019;9:1161. doi: 10.3389/fonc.2019.01161.
    1. Steuer CE, Ramalingam SS. Tumor mutation burden: leading immunotherapy to the era of precision medicine? JCO. 2018;36(7):631–632. doi: 10.1200/JCO.2017.76.8770.
    1. Schrock AB, Ouyang C, Sandhu J, Sokol E, Jin D, Ross JS, Miller VA, Lim D, Amanam I, Chao J, Catenacci D, Cho M, Braiteh F, Klempner SJ, Ali SM, Fakih M. Tumor mutational burden is predictive of response to immune checkpoint inhibitors in MSI-high metastatic colorectal cancer. Ann Oncol. 2019;30(7):1096–1103. doi: 10.1093/annonc/mdz134.
    1. Fernandez EM, Eng K, Beg S, Beltran H, Faltas BM, Mosquera JM, Nanus DM, Pisapia DJ, Rao RA, Robinson BD, Rubin MA, Elemento O, Sboner A, Shah MA, Song W. Cancer-specific thresholds adjust for whole exome sequencing-based tumor mutational burden distribution. JCO Precis Oncol. 2019;3(3):1–12. doi: 10.1200/PO.18.00400.
    1. Heeke S, Hofman P. Tumor mutational burden assessment as a predictive biomarker for immunotherapy in lung cancer patients: getting ready for prime-time or not? Transl Lung Cancer Res. 2018;7(6):631–638. doi: 10.21037/tlcr.2018.08.04.
    1. Garassino M, Rodriguez-Abreu D, Gadgeel S, Esteban E, Felip E, Speranza G, Reck M, Hui R, Boyer M, Cristescu R, Aurora-Garg D, Albright A, Loboda A, Kobie J, Lunceford J, Ayers M, Lubiniecki G, Piperdi B, Pietanza MC, Garon E. OA04.06 Evaluation of TMB in KEYNOTE-189: pembrolizumab plus chemotherapy vs placebo plus chemotherapy for nonsquamous NSCLC. J Thorac Oncol. 2019;14(10):S216–S217. doi: 10.1016/j.jtho.2019.08.427.
    1. Langer C, Gadgeel S, Borghaei H, Patnaik A, Powell S, Gentzler R, et al. OA04.05 KEYNOTE-021: TMB and outcomes for carboplatin and pemetrexed with or without pembrolizumab for nonsquamous NSCLC. J Thorac Oncol. 2019;14(10):S216. doi: 10.1016/j.jtho.2019.08.426.
    1. Fancello L, Gandini S, Pelicci PG, Mazzarella L. Tumor mutational burden quantification from targeted gene panels: major advancements and challenges. J Immunother Cancer. 2019;7(1):183. doi: 10.1186/s40425-019-0647-4.
    1. Patel SP, Kurzrock R. PD-L1 Expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther. 2015;14(4):847–856. doi: 10.1158/1535-7163.MCT-14-0983.
    1. Schlam I, Dyehouse KM, Ward PJ. Application of NCCN guidelines in PD-L1 expression for patients with stage IV non-small cell lung cancer. J Clin Oncol. 2018;36(15_suppl):e15030. doi: 10.1200/JCO.2018.36.15_suppl.e15030.
    1. Alexandrov LB, Ju YS, Haase K, Van Loo P, Martincorena I, Nik-Zainal S, et al. Mutational signatures associated with tobacco smoking in human cancer. Science. 2016;354(6312):618–622. doi: 10.1126/science.aag0299.
    1. Boichard A, Pham TV, Yeerna H, Goodman A, Tamayo P, Lippman S, Frampton GM, Tsigelny IF, Kurzrock R. APOBEC-related mutagenesis and neo-peptide hydrophobicity: implications for response to immunotherapy. Oncol Immunol. 2018;24(3):1–12. doi: 10.1080/2162402X.2018.1550341.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SAJR, Behjati S, Biankin AV, et al. Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421. doi: 10.1038/nature12477.
    1. Boichard A, Tsigelny IF, Kurzrock R. High expression of PD-1 ligands is associated with kataegis mutational signature and APOBEC3 alterations. Oncoimmunology. 2017;6(3):e1284719. doi: 10.1080/2162402X.2017.1284719.
    1. Gerlinger M, Rowan AJ, Horswell S, Math M, Larkin J, Endesfelder D, Gronroos E, Martinez P, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald N, Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA, Swanton C. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–892. doi: 10.1056/NEJMoa1113205.
    1. Riaz N, Havel JJ, Makarov V, Desrichard A, Urba WJ, Sims JS, et al. Tumor and microenvironment evolution during immunotherapy with nivolumab. Cell. 2017;171(4):934–949.e16. doi: 10.1016/j.cell.2017.09.028.
    1. Frampton GM, Fichtenholtz A, Otto GA, Wang K, Downing SR, He J, Schnall-Levin M, White J, Sanford EM, An P, Sun J, Juhn F, Brennan K, Iwanik K, Maillet A, Buell J, White E, Zhao M, Balasubramanian S, Terzic S, Richards T, Banning V, Garcia L, Mahoney K, Zwirko Z, Donahue A, Beltran H, Mosquera JM, Rubin MA, Dogan S, Hedvat CV, Berger MF, Pusztai L, Lechner M, Boshoff C, Jarosz M, Vietz C, Parker A, Miller VA, Ross JS, Curran J, Cronin MT, Stephens PJ, Lipson D, Yelensky R. Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol. 2013;31(11):1023–1031. doi: 10.1038/nbt.2696.
    1. Johnson DB, Frampton GM, Rioth MJ, Yusko E, Xu Y, Guo X, Ennis RC, Fabrizio D, Chalmers ZR, Greenbowe J, Ali SM, Balasubramanian S, Sun JX, He Y, Frederick DT, Puzanov I, Balko JM, Cates JM, Ross JS, Sanders C, Robins H, Shyr Y, Miller VA, Stephens PJ, Sullivan RJ, Sosman JA, Lovly CM. Targeted next generation sequencing identifies markers of response to PD-1 blockade. Cancer Immunol Res. 2016;4(11):959–967. doi: 10.1158/2326-6066.CIR-16-0143.
    1. Chalmers ZR, Connelly CF, Fabrizio D, Gay L, Ali SM, Ennis R, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017;9(1):34. doi: 10.1186/s13073-017-0424-2.
    1. Bonta I, Isac JF, Meiri E, Bonta D, Rich P. Correlation between tumor mutation burden and response to immunotherapy. JCO. 2017;35(15_suppl):e14579. doi: 10.1200/JCO.2017.35.15_suppl.e14579.
    1. Devarakonda S, Rotolo F, Tsao M-S, Lanc I, Brambilla E, Masood A, et al. Tumor mutation burden as a biomarker in resected non-small-cell lung cancer. J Clin Oncol. 2018;14:JCO2018781963.
    1. Johnson A, Severson E, Gay L, Vergilio J-A, Elvin J, Suh J, Daniel S, Covert M, Frampton GM, Hsu S, Lesser GJ, Stogner-Underwood K, Mott RT, Rush SZ, Stanke JJ, Dahiya S, Sun J, Reddy P, Chalmers ZR, Erlich R, Chudnovsky Y, Fabrizio D, Schrock AB, Ali S, Miller V, Stephens PJ, Ross J, Crawford JR, Ramkissoon SH. Comprehensive genomic profiling of 282 pediatric low- and high-grade gliomas reveals genomic drivers, tumor mutational burden, and hypermutation signatures. Oncologist. 2017;22(12):1478–1490. doi: 10.1634/theoncologist.2017-0242.
    1. Goodman AM, Sokol ES, Frampton GM, Lippman SM, Kurzrock R. Microsatellite-stable tumors with high mutational burden benefit from immunotherapy. Cancer Immunol Res. 2019;7(10):1570–1573. doi: 10.1158/2326-6066.CIR-19-0149.
    1. Chang L, Chang M, Chang HM, Chang F. Microsatellite instability: a predictive biomarker for cancer immunotherapy. Appl Immunohistochem Mol Morphol. 2018;26(2):e15–e21. doi: 10.1097/PAI.0000000000000575.
    1. Choi S, Yu Y, Grimmer MR, Wahl M, Chang SM, Costello JF. Temozolomide-associated hypermutation in gliomas. Neuro Oncol. 2018;20(10):1300–1309. doi: 10.1093/neuonc/noy016.

Source: PubMed

3
구독하다