Evolution of RAS Mutational Status in Liquid Biopsies During First-Line Chemotherapy for Metastatic Colorectal Cancer

Susanne Klein-Scory, Ingo Wahner, Marina Maslova, Yosef Al-Sewaidi, Michael Pohl, Thomas Mika, Swetlana Ladigan, Roland Schroers, Alexander Baraniskin, Susanne Klein-Scory, Ingo Wahner, Marina Maslova, Yosef Al-Sewaidi, Michael Pohl, Thomas Mika, Swetlana Ladigan, Roland Schroers, Alexander Baraniskin

Abstract

Treatment options for patients with metastatic colorectal cancer (mCRC) are limited. This particularly affects the largest group of patients with RAS mutations, who are considered ineligible for therapy with antiEGFR antibodies. In this liquid biopsy-based study, we performed the first in-depth analysis of the RAS mutational status in initially RAS-mutated patients during first-line therapy. RAS status of twelve patients with initially RAS-mutated mCRC was monitored longitudinally in 69 liquid biopsy samples. We focused on patients with stable disease (SD) or partial remission (PR) during first-line therapy (11 patients). Detection of fragmented RAS-mutated circulating cell-free tumor DNA (ctDNA) in plasma was performed by digital-droplet PCR (ddPCR) and BEAMing. Patients' total tumor masses were determined by measuring the tumor volumes using CT scan data. All patients with PR or SD at first follow-up showed a significant decrease of RAS mutational load. In ten patients (91%), the ctDNA-based RAS mutational status converted to wild-type in ddPCR and BEAMing. Remarkably, conversions were observed early after the first cycle of chemotherapy. Plasma concentration of ctDNA was controlled by determination of methylated WIF1-promotor ctDNA burden as a second tumor marker for mCRC. Persistent presence of methylated WIF1-promotor fragments confirmed the ongoing release of ctDNA during treatment. In patients with initially RAS-mutated mCRC, RAS mutations rapidly disappeared during first-line therapy in liquid biopsy, independent of type and intensity of chemotherapy and irrespective of anti-VEGF treatments. Following our results demonstrating conversion of RAS-mutational status, potential effectiveness of anti-EGFR antibodies in selected patients becomes an attractive hypothesis for future studies.

Keywords: BEAMing; circulating tumor DNA; clonal selection; colorectal cancer; digital droplet PCR; liquid biopsy; methylation WIF1 promotor; neoRAS wild-type.

Copyright © 2020 Klein-Scory, Wahner, Maslova, Al-Sewaidi, Pohl, Mika, Ladigan, Schroers and Baraniskin.

Figures

Figure 1
Figure 1
Dynamics of KRAS mutant clones measured in plasma samples. (A)Patient 3: RAS mutations disappeared already after 3 cycles FOLFOXIRI and remained not detectable for 9 months. PR after 6 cycles FOLFOXIRI made the patient eligible for tumor resection (arrow). After the following chemotherapy break PD occurred simultaneously with renewed rise of RAS mutation load. Already after 1 cycle FOLFIRI and bevacizumab the RAS mutations disappeared again and rose only after the next chemotherapy break due to TACE treatment. This case conclusively demonstrated that even multiple conversions of RAS status are possible with appropriate chemotherapy. (B)Patient 4: Already after 2 cycles FOLFIRI + bevacizumab the RAS mutations disappeared and remained not detectable for 6 months. Due to PR after 8 further cycles, the treatment was deescalated to 5-FU and bevacizumab. During this period, RAS mutation load increased again. Next, PD was diagnosed and the subsequent treatment change to FOLFIRI and aflibercept failed to achieve tumor response. However, RAS mutation load did not increase. This case points out that a renewed rise of RAS mutation load may be an early indicator for a lack of response. Furthermore, it can decrease during PD indicating that PD is caused by a RAS wild-type clone. (C)Patient 12: Neither FOLFOX nor FOLFIRI or addition of anti-VEGF antibody therapy led to a decrease of RAS mutation load or tumor response.
Figure 2
Figure 2
The dramatic decrease of RAS mutational load was detectable after 2 cycles (A,B,D) and the disappearance of RAS mutations occurs earliest after 2 cycles of therapy (B,D). In other cases (A,C,E–H) the disappearance is obvious after 4–12 cycles of therapy. The tumor loads, measured by CT scans, were strongly reduced by 75–90% in two cases (F–H), and more moderately reduced by 20–60% in other cases (A–E). In case (I), despite the dramatic decrease of RAS mutational load the Ras mutations remained detectable. (A) Patient 7, (B) patient 8, (C) patient 2, (D) patient 6, (E) patient 10, (F) patient 9, (G) patient 1, (H) patient 5, and (I) patient 11.
Figure 3
Figure 3
Change of WIF1 promotor methylation proportion vs. change of RAS mutational allele frequency. Samples of patients at the diagnosis time points vs. samples of the time point of disappearance of RAS mutations were measured by methylation specific ddPCR (see Supplementary File). WIF1 promotor methylation proportion remained detectable in samples with massive RAS MAF% reduction. (A–F) (solid line with circle, change of RAS mutant allele frequency; dashed line with cross, change of WIF1 promotor methylation proportion).

References

    1. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H, et al. . Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. (2013) 49:1374–403. 10.1016/j.ejca.2012.12.027
    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2018) 68:394–424. 10.3322/caac.21492
    1. Schmoll HJ, van Cutsem E, Stein A, Valentini V, Glimelius B, Haustermans K, et al. . ESMO consensus guidelines for management of patients with colon and rectal cancer. A personalized approach to clinical decision making. Ann Oncol. (2012) 23:2479–516. 10.1093/annonc/mds236
    1. van Cutsem E, Cervantes A, Adam R, Sobrero A, van Krieken JH, Aderka D, et al. . ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol. (2016) 27:1386–422. 10.1093/annonc/mdw235
    1. Boleij A, Tack V, Taylor A, Kafatos G, Jenkins-Anderson S, Tembuyser L, et al. . RAS testing practices and RAS mutation prevalence among patients with metastatic colorectal cancer: results from a Europe-wide survey of pathology centres. BMC Cancer. (2016) 16:825. 10.1186/s12885-016-2810-3
    1. Klein-Scory S, Maslova M, Pohl M, Eilert-Micus C, Schroers R, Schmiegel W, et al. . Significance of liquid biopsy for monitoring and therapy decision of colorectal cancer. Transl Oncol. (2018) 11:213–20. 10.1016/j.tranon.2017.12.010
    1. Findlay JM, Castro-Giner F, Makino S, Rayner E, Kartsonaki C, Cross W, et al. . Differential clonal evolution in oesophageal cancers in response to neo-adjuvant chemotherapy. Nat Commun. (2016) 7:11111. 10.1038/ncomms11111
    1. Li W, Qiu T, Guo L, Ying J. Major challenges related to tumor biological characteristics in accurate mutation detection of colorectal cancer by next-generation sequencing. Cancer Lett. (2017) 410:92–9. 10.1016/j.canlet.2017.09.014
    1. Morelli MP, Overman MJ, Dasari A, Kazmi SMA, Mazard T, Vilar E, et al. . Characterizing the patterns of clonal selection in circulating tumor DNA from patients with colorectal cancer refractory to anti-EGFR treatment. Ann Oncol. (2015) 26:731–6. 10.1093/annonc/mdv005
    1. Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, et al. . Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. (2015) 21:795–801. 10.1038/nm.3870
    1. Yamada T, Iwai T, Takahashi G, Kan H, Koizumi M, Matsuda A, et al. . Utility of KRAS mutation detection using circulating cell-free DNA from patients with colorectal cancer. Cancer Sci. (2016) 107:936–43. 10.1111/cas.12959
    1. Toledo RA, Cubillo A, Vega E, Garralda E, Alvarez R, La Varga LU, et al. . Clinical validation of prospective liquid biopsy monitoring in patients with wild-type RAS metastatic colorectal cancer treated with FOLFIRI-cetuximab. Oncotarget. (2017) 8:35289–300. 10.18632/oncotarget.13311
    1. Khan KH, Cunningham D, Werner B, Vlachogiannis G, Spiteri I, Heide T, et al. . Longitudinal liquid biopsy and mathematical modeling of clonal evolution forecast time to treatment failure in the PROSPECT-C phase II colorectal cancer clinical trial. Cancer Discov. (2018) 8:1270–85. 10.1158/-17-0891
    1. Cremolini C, Rossini D, Dell'Aquila E, Lonardi S, Conca E, Del Re M, et al. . Rechallenge for patients with RAS and BRAF wild-type metastatic colorectal cancer with acquired resistance to first-line cetuximab and irinotecan: a phase 2 single-arm clinical trial. JAMA Oncol. (2019) 5:343–350. 10.1001/jamaoncol.2018.5080
    1. Diehl F, Li M, He Y, Kinzler KW, Vogelstein B, Dressman D. BEAMing: single-molecule PCR on microparticles in water-in-oil emulsions. Nat Methods. (2006) 3:551–9. 10.1038/nmeth898
    1. Schmiegel W, Scott RJ, Dooley S, Lewis W, Meldrum CJ, Pockney P, et al. . Blood-based detection of RAS mutations to guide anti-EGFR therapy in colorectal cancer patients: concordance of results from circulating tumor DNA and tissue-based RAS testing. Mol Oncol. (2017) 11:208–19. 10.1002/1878-0261.12023
    1. Garcia J, Forestier J, Dusserre E, Wozny A-S, Geiguer F, Merle P, et al. . Cross-platform comparison for the detection of RAS mutations in cfDNA (ddPCR Biorad detection assay, BEAMing assay, and NGS strategy). Oncotarget. (2018) 9:21122–31. 10.18632/oncotarget.24950
    1. van Ginkel JH, Huibers MMH, van Es RJJ, de Bree R., Willems SM. Droplet digital PCR for detection and quantification of circulating tumor DNA in plasma of head and neck cancer patients. BMC Cancer. (2017) 17:428. 10.1186/s12885-017-3424-0
    1. Garrigou S, Perkins G, Garlan F, Normand C, Didelot A, Le Corre D, et al. . A study of hypermethylated circulating tumor DNA as a universal colorectal cancer biomarker. Clin Chem. (2016) 62:1129–39. 10.1373/clinchem.2015.253609
    1. Yu M, Carter KT, Makar KW, Vickers K, Ulrich CM, Schoen RE, et al. . MethyLight droplet digital PCR for detection and absolute quantification of infrequently methylated alleles. Epigenetics. (2015) 10:803–9. 10.1080/15592294.2015.1068490
    1. Moati E, Blons H, Taly V, Garlan F, Wang-Renault SF, Pietrasz D, et al. . Plasma clearance of RAS mutation under therapeutic pressure is a rare event in metastatic colorectal cancer. Int J Cancer. (2020) 147:1185–89. 10.1002/ijc.32657
    1. Roperch J-P, Incitti R, Forbin S, Bard F, Mansour H, Mesli F, et al. . Aberrant methylation of NPY, PENK, and WIF1 as a promising marker for blood-based diagnosis of colorectal cancer. BMC Cancer. (2013) 13:566. 10.1186/1471-2407-13-566
    1. Weisenberger DJ, Trinh BN, Campan M, Sharma S, Long TI, Ananthnarayan S, et al. . DNA methylation analysis by digital bisulfite genomic sequencing and digital MethyLight. Nucleic Acids Res. (2008) 36:4689–98. 10.1093/nar/gkn455
    1. Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. (2003) 3:459–65. 10.1038/nrc1193
    1. Tsuchida N, Murugan AK, Grieco M. Kirsten Ras* oncogene: significance of its discovery in human cancer research. Oncotarget. (2016) 7:46717–33. 10.18632/oncotarget.8773
    1. Fakih MG. Metastatic colorectal cancer: current state and future directions. J Clin Oncol. (2015) 33:1809–24. 10.1200/JCO.2014.59.7633
    1. García-Foncillas J, Tabernero J, Élez E, Aranda E, Benavides M, Camps C, et al. . Prospective multicenter real-world RAS mutation comparison between OncoBEAM-based liquid biopsy and tissue analysis in metastatic colorectal cancer. Br J Cancer. (2018) 119:1464–70. 10.1038/s41416-018-0293-5
    1. Gazzaniga P, Raimondi C, Nicolazzo C, Gradilone A, Cortesi E. 1672PctDNA might expand therapeutic options for second line treatment of KRAS mutant mCRC. Ann Oncol. (2017) 28(suppl_5):586 10.1093/annonc/mdx390.044
    1. Gazzaniga P, Raimondi C, Urbano F, Cortesi E. EGFR inhibitor as second-line therapy in a patient with mutant RAS metastatic colorectal cancer: circulating tumor DNA to personalize treatment. JCO Precis Oncol. (2018) 2:1–6. 10.1200/PO.17.00277
    1. Raimondi C, Nicolazzo C, Belardinilli F, Loreni F, Gradilone A, Mahdavian Y, et al. . Transient disappearance of RAS mutant clones in plasma: a counterintuitive clinical use of EGFR inhibitors in RAS mutant metastatic colorectal cancer. Cancers. (2019) 11:42. 10.3390/cancers11010042
    1. Vivancos A, Aranda E, Benavides M, Elez E, Gomez A, Toledano M, et al. . Evaluation of the sensitivity of RAS mutation detection of the Idylla platform in comparison to the OncoBEAM RAS CRC assay. JCO. (2018) 36:592. 10.1200/JCO.2018.36.4_suppl.592
    1. Sunakawa Y, Usher J, Satake H, Jaimes Y, Miyamoto Y, Nakamura M, et al. 543PGene mutation status in circulating tumor DNA (ctDNA) and first-line FOLFOXIRI plus bevacizumab (bev) in metastatic colorectal cancer (mCRC) harboring RAS mutation. Ann Oncol. (2018) 29 (suppl. 8):mdy281–089. 10.1093/annonc/mdy281.089
    1. Spindler K-LG, Pallisgaard N, Andersen RF, Jakobsen A. Changes in mutational status during third-line treatment for metastatic colorectal cancer–results of consecutive measurement of cell free DNA, KRAS and BRAF in the plasma. Int J Cancer. (2014) 135:2215–22. 10.1002/ijc.28863
    1. Vidal J, Muinelo L, Dalmases A, Jones F, Edelstein D, Iglesias M, et al. . Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann Oncol. (2017) 28:1325–32. 10.1093/annonc/mdx125
    1. Haigis KM, Kendall KR, Wang Y, Cheung A, Haigis MC, Glickman JN, et al. . Differential effects of oncogenic K-Ras and N-Ras on proliferation, differentiation and tumor progression in the colon. Nat Genet. (2008) 40:600–8. 10.1038/ng.115
    1. Prior IA, Lewis PD, Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer Res. (2012) 72:2457–67. 10.1158/0008-5472.CAN-11-2612
    1. Ling S, Hu Z, Yang Z, Yang F, Li Y, Lin P, et al. . Extremely high genetic diversity in a single tumor points to prevalence of non-darwinian cell evolution. Proc Natl Acad Sci USA. (2015) 112:E6496–E6505. 10.1073/pnas.1519556112
    1. Baker A-M, Wang M, Ma X-J, Anderson C, Huang W, Domingo E, et al. Abstract 3953: visualization of treatment resistant subclones in colorectal cancer by mutation specific RNA in situ hybridization. Cancer Res. (2017) 77:3953 10.1158/1538-7445.AM2017-3953
    1. McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. (2017) 168:613–28. 10.1016/j.cell.2017.01.018
    1. Parsons BL. Multiclonal tumor origin: evidence and implications. Mutat Res. (2018) 777:1–18. 10.1016/j.mrrev.2018.05.001
    1. Holch JW, Ricard I, Stintzing S, Modest DP, Heinemann V. The relevance of primary tumour location in patients with metastatic colorectal cancer: a meta-analysis of first-line clinical trials. Eur J Cancer. (2017) 70:87–98. 10.1016/j.ejca.2016.10.007
    1. van Cutsem E, Lenz H-J, Köhne C-H, Heinemann V, Tejpar S, Melezínek I, et al. . Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol. (2015) 33:692–700. 10.1200/JCO.2014.59.4812
    1. Laurent-Puig P, Pekin D, Normand C, Kotsopoulos SK, Nizard P, Perez-Toralla K, et al. . Clinical relevance of KRAS-mutated subclones detected with picodroplet digital PCR in advanced colorectal cancer treated with anti-EGFR therapy. Clin Cancer Res. (2015) 21:1087–97. 10.1158/1078-0432.CCR-14-0983
    1. Santos C, Azuara D, Garcia-Carbonero R, Alfonso PG, Carrato A, Elez ME, et al. . Optimization of RAS/BRAF mutational analysis confirms improvement in patient selection for clinical benefit to Anti-EGFR treatment in metastatic colorectal cancer. Mol Cancer Ther. (2017) 16:1999–2007. 10.1158/1535-7163.MCT-17-0153
    1. Fernández Montes A, Martinez Lago N, De la Cámara Gómez J, Covela Rúa M, Cousillas Castiñeiras A, Gonzalez Villarroel P, et al. P-089FOLFIRI plus panitumumab as second-line treatment in mutated RAS metastatic colorectal cancer patients who converted to wild type RAS after receiving first-line FOLFOX/CAPOX plus bevacizumab-based treatment: phase II CONVERTIX trial. Ann Oncol. (2019) 30 (Suppl. 4): mdz155–088. 10.1093/annonc/mdz155.088

Source: PubMed

3
Suscribir