ctDNA applications and integration in colorectal cancer: an NCI Colon and Rectal-Anal Task Forces whitepaper

Arvind Dasari, Van K Morris, Carmen J Allegra, Chloe Atreya, Al B Benson 3rd, Patrick Boland, Ki Chung, Mehmet S Copur, Ryan B Corcoran, Dustin A Deming, Andrea Dwyer, Maximilian Diehn, Cathy Eng, Thomas J George, Marc J Gollub, Rachel A Goodwin, Stanley R Hamilton, Jaclyn F Hechtman, Howard Hochster, Theodore S Hong, Federico Innocenti, Atif Iqbal, Samuel A Jacobs, Hagen F Kennecke, James J Lee, Christopher H Lieu, Heinz-Josef Lenz, O Wolf Lindwasser, Clara Montagut, Bruno Odisio, Fang-Shu Ou, Laura Porter, Kanwal Raghav, Deborah Schrag, Aaron J Scott, Qian Shi, John H Strickler, Alan Venook, Rona Yaeger, Greg Yothers, Y Nancy You, Jason A Zell, Scott Kopetz, Arvind Dasari, Van K Morris, Carmen J Allegra, Chloe Atreya, Al B Benson 3rd, Patrick Boland, Ki Chung, Mehmet S Copur, Ryan B Corcoran, Dustin A Deming, Andrea Dwyer, Maximilian Diehn, Cathy Eng, Thomas J George, Marc J Gollub, Rachel A Goodwin, Stanley R Hamilton, Jaclyn F Hechtman, Howard Hochster, Theodore S Hong, Federico Innocenti, Atif Iqbal, Samuel A Jacobs, Hagen F Kennecke, James J Lee, Christopher H Lieu, Heinz-Josef Lenz, O Wolf Lindwasser, Clara Montagut, Bruno Odisio, Fang-Shu Ou, Laura Porter, Kanwal Raghav, Deborah Schrag, Aaron J Scott, Qian Shi, John H Strickler, Alan Venook, Rona Yaeger, Greg Yothers, Y Nancy You, Jason A Zell, Scott Kopetz

Abstract

An increasing number of studies are describing potential uses of circulating tumour DNA (ctDNA) in the care of patients with colorectal cancer. Owing to this rapidly developing area of research, the Colon and Rectal-Anal Task Forces of the United States National Cancer Institute convened a panel of multidisciplinary experts to summarize current data on the utility of ctDNA in the management of colorectal cancer and to provide guidance in promoting the efficient development and integration of this technology into clinical care. The panel focused on four key areas in which ctDNA has the potential to change clinical practice, including the detection of minimal residual disease, the management of patients with rectal cancer, monitoring responses to therapy, and tracking clonal dynamics in response to targeted therapies and other systemic treatments. The panel also provides general guidelines with relevance for ctDNA-related research efforts, irrespective of indication.

Conflict of interest statement

C.A. has received research funding from Guardant Health. R.C. is an advisory board member of Guardant Health and holds equity interests in nRichDx. M.D. has acted as a consultant of Quanticell. C.L. has acted as a consultant of Foundation Medicine. C.M. has acted as a consultant of Guardant Health. The other authors declare no competing interests.

Figures

Fig. 1. Clinical applications of ctDNA.
Fig. 1. Clinical applications of ctDNA.
Circulating tumour DNA (ctDNA) provides a more sensitive method of detecting malignancies than imaging or other conventional approaches. This sensitivity can be exploited in several ways: early diagnosis of colorectal cancer prior to the emergence of clinical or radiological manifestations and in the detection of minimal residual disease (MRD), defined as the detection of ctDNA with no other clinical evidence of disease recurrence in patients who have completed all potentially curative therapies. In patients with radiographically evident disease, ctDNA also seems to be more sensitive to changes in tumour burden and might assist in tailoring the intensity of therapy in the neoadjuvant setting and in monitoring for tumour response in patients requiring palliative treatment. Furthermore, qualitative assessments of the types of aberrations and their subsequent alterations might assist in assessments of tumour evolution and heterogeneity that lead to the emergence of resistance as well as in selection of the most appropriate therapies.
Fig. 2. ctDNA isolation and analysis.
Fig. 2. ctDNA isolation and analysis.
a | Circulating cell-free DNA, of which circulating tumour DNA (ctDNA) is a part of, is isolated from plasma samples after serial centrifugation of blood collected in either K2EDTA or cell-stabilizing tubes. ctDNA is subsequently isolated from cell-free DNA using library preparations and analysed for the presence of various genetic aberrations, including mutations, copy-number variations, fusions and/or other alterations such as changes in DNA methylation. b | Molecular barcoding: prior to PCR and sequencing, each DNA fragment can be labelled with unique DNA barcodes; subsequently, reads that share the same barcode (typically in thousands) can be grouped together because they all originate from the same ctDNA fragment. This approach enables sequencing errors (orange pentagon, seen in the minority) to be distinguished from true mutations (red pentagon, seen in the majority). Molecular barcoding also helps correct potential biases in amplification and thus assists in the precise quantification of mutations or amplification frequencies. NGS, next-generation sequencing; RBC, red blood cells.
Fig. 3. Applications of ctDNA in tailoring…
Fig. 3. Applications of ctDNA in tailoring the aggressiveness of adjuvant therapy.
Owing to the high specificity of circulating tumour DNA (ctDNA) for the prediction of disease recurrence, patients with detectable ctDNA might be considered candidates for the escalation of adjuvant therapy over the standard-of-care approach in order to reduce the risk of disease recurrence. Conversely, patients who lack detectable ctDNA, as determined using a sufficiently sensitive assay, and who have a low risk of disease recurrence might benefit from de-escalation to less-intense adjuvant therapies that reduce the risk of toxicities.
Fig. 4. Tumour dynamics of patients receiving…
Fig. 4. Tumour dynamics of patients receiving biomarker-selected targeted therapies.
Patients with RAS/RAF-wild-type, HER2-negative colorectal cancer receiving anti-EGFR antibodies demonstrate growth of clones of resistant cells over time, as reflected by increases in the variant allele frequency (VAF) of the resistance mutations present in these clones in circulating tumour DNA (ctDNA) (part a). Of note, the emergence of such aberrations in ctDNA typically predates radiographic or clinical disease progression. Such patients can be managed in several ways, including a ‘holiday’ from the targeted agent during the next line of therapy followed by the subsequent re-introduction of the original targeted agent based on a reduction in the VAF of resistance mutations on ctDNA-based monitoring (part b); by introducing a different biomarker-selected targeted therapy or an agent targeting treatment-emergent mechanisms of resistance, as determined by ctDNA findings (part c); or the introduction of one or more agents with activity against other downstream targets (part d).

References

    1. Holdhoff M, Schmidt K, Donehower R, et al. Analysis of circulating tumor DNA to confirm somatic KRAS mutations. J. Natl Cancer. Inst. 2009;101:1284–1285. doi: 10.1093/jnci/djp240.
    1. Diehl F, Li M, Dressman D, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc. Natl Acad. Sci. USA. 2005;102:16368–16373. doi: 10.1073/pnas.0507904102.
    1. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008;14:985–990. doi: 10.1038/nm.1789.
    1. Diehl F, Schmidt K, Durkee KH, et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology. 2008;135:489–498. doi: 10.1053/j.gastro.2008.05.039.
    1. De Mattos-Arruda L, Mayor R, Ng CKY, et al. Cerebrospinal fluid-derived circulating tumour DNA better represents the genomic alterations of brain tumours than plasma. Nat. Commun. 2015;6:8839. doi: 10.1038/ncomms9839.
    1. Kimura H, Fujiwara Y, Sone T, et al. EGFR mutation status in tumour-derived DNA from pleural effusion fluid is a practical basis for predicting the response to gefitinib. Br. J. Cancer. 2006;95:1390–1395. doi: 10.1038/sj.bjc.6603428.
    1. Wang Y, Springer S, Mulvey CL, et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci. Transl Med. 2015;7:293ra104. doi: 10.1126/scitranslmed.aaa8507.
    1. Reckamp KL, Melnikova VO, Karlovich C, et al. A highly sensitive and Quantitative test platform for detection of NSCLC EGFR mutations in urine and plasma. J. Thorac. Oncol. 2016;11:1690–1700. doi: 10.1016/j.jtho.2016.05.035.
    1. Bettegowda C, Sausen M, Leary RJ, 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. Diehn M, Alizadeh AA, Adams H-P, et al. Early prediction of clinical outcomes in resected stage II and III colorectal cancer (CRC) through deep sequencing of circulating tumor DNA (ctDNA) J. Clin. Oncol. 2017;35:3591–3591. doi: 10.1200/JCO.2017.35.15_suppl.3591.
    1. Overman MJ, Vauthey J-N, Aloia TA, et al. Circulating tumor DNA (ctDNA) utilizing a high-sensitivity panel to detect minimal residual disease post liver hepatectomy and predict disease recurrence. J. Clin. Oncol. 2017;35:3522–3522. doi: 10.1200/JCO.2017.35.15_suppl.3522.
    1. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci. Transl Med. 2016;8:346ra92. doi: 10.1126/scitranslmed.aaf6219.
    1. Tie JC, Cohen JD, Wang Y, et al. Serial circulating tumor DNA analysis as a prognostic marker and a real-time indicator of adjuvant chemotherapy efficacy in stage III colon cancer. J. Clin. Oncol. 2018;36(Suppl. 15):3516. doi: 10.1200/JCO.2018.36.15_suppl.3516.
    1. Wang JY, Hsieh JS, Chang MY, et al. Molecular detection of APC, K- ras, and p53 mutations in the serum of colorectal cancer patients as circulating biomarkers. World J. Surg. 2004;28:721–726.
    1. Sun X, Huang T, Cheng F, et al. Monitoring colorectal cancer following surgery using plasma circulating tumor DNA. Oncol. Lett. 2018;15:4365–4375.
    1. Bazan V, Bruno L, Augello C, et al. Molecular detection of TP53, Ki-Ras and p16INK4A promoter methylation in plasma of patients with colorectal cancer and its association with prognosis. Results of a 3-year GOIM (Gruppo Oncologico dell’Italia Meridionale) prospective study. Ann. Oncol. 2006;17:vii84–vii90. doi: 10.1093/annonc/mdl958.
    1. Petit J, Carroll G, Gould T, et al. Cell-free DNA as a diagnostic blood-based biomarker for colorectal cancer: a systematic review. J. Surg. Res. 2019;236:184–197. doi: 10.1016/j.jss.2018.11.029.
    1. Cree IA, Uttley L, Buckley Woods H, et al. The evidence base for circulating tumour DNA blood-based biomarkers for the early detection of cancer: a systematic mapping review. BMC Cancer. 2017;17:697. doi: 10.1186/s12885-017-3693-7.
    1. Siravegna G, Marsoni S, Siena S, et al. Integrating liquid biopsies into the management of cancer. Nat. Rev. Clin. Oncol. 2017;14:531–548. doi: 10.1038/nrclinonc.2017.14.
    1. Meddeb R, Pisareva E, Thierry AR. Guidelines for the preanalytical conditions for analyzing circulating cell-free DNA. Clin. Chem. 2019;65:623–633. doi: 10.1373/clinchem.2018.298323.
    1. Merker JD, Oxnard GR, Compton C, et al. Circulating tumor DNA analysis in patients with cancer: American Society of clinical oncology and college of American pathologists joint review. Arch. Pathol. Lab. Med. 2018;142:1242–1253. doi: 10.5858/arpa.2018-0901-SA.
    1. Merker JD, Oxnard GR, Compton C, et al. Circulating tumor DNA Analysis in Patients with cancer: American Society of Clinical Oncology and College of American Pathologists joint review. J. Clin. Oncol. 2018;36:1631–1641. doi: 10.1200/JCO.2017.76.8671.
    1. van Dessel LF, Beije N, Helmijr JC, et al. Application of circulating tumor DNA in prospective clinical oncology trials - standardization of preanalytical conditions. Mol. Oncol. 2017;11:295–304. doi: 10.1002/1878-0261.12037.
    1. Medina Diaz I, Nocon A, Mehnert DH, et al. Performance of streck cfDNA blood collection tubes for liquid biopsy testing. PLoS One. 2016;11:e0166354. doi: 10.1371/journal.pone.0166354.
    1. Kang Q, Henry NL, Paoletti C, et al. Comparative analysis of circulating tumor DNA stability In K3EDTA, streck, and cellsave blood collection tubes. Clin. Biochem. 2016;49:1354–1360. doi: 10.1016/j.clinbiochem.2016.03.012.
    1. Page K, Powles T, Slade MJ, et al. The importance of careful blood processing in isolation of cell-free DNA. Ann. N. Y. Acad. Sci. 2006;1075:313–317. doi: 10.1196/annals.1368.042.
    1. Corcoran RB, Chabner BA. Application of cell-free DNA analysis to cancer treatment. N. Engl. J. Med. 2018;379:1754–1765. doi: 10.1056/NEJMra1706174.
    1. Oxnard GR, Paweletz CP, Sholl LM. Genomic analysis of plasma cell-free DNA in patients with cancer. JAMA Oncol. 2017;3:740–741. doi: 10.1001/jamaoncol.2016.2835.
    1. Saiyed ZM, Bochiwal C, Gorasia H, et al. Application of magnetic particles (Fe3O4) for isolation of genomic DNA from mammalian cells. Anal. Biochem. 2006;356:306–308. doi: 10.1016/j.ab.2006.06.027.
    1. Benson AB, Venook AP, Al-Hawary MM, et al. NCCN guidelines insights: colon cancer, version 2.2018. J. Natl Compr. Canc. Netw. 2018;16:359–369. doi: 10.6004/jnccn.2018.0021.
    1. Ellervik C, Vaught J. Preanalytical variables affecting the integrity of human biospecimens in biobanking. Clin. Chem. 2015;61:914–934. doi: 10.1373/clinchem.2014.228783.
    1. Koessler T, Addeo A, Nouspikel T. Implementing circulating tumor DNA analysis in a clinical laboratory: a user manual. Adv. Clin. Chem. 2019;89:131–188. doi: 10.1016/bs.acc.2018.12.004.
    1. Hojbjerg JA, Madsen AT, Schmidt HH, et al. Intra-individual variation of circulating tumour DNA in lung cancer patients. Mol. Oncol. 2019;13:2098–2106. doi: 10.1002/1878-0261.12546.
    1. Compton CC, Fielding LP, Burgart LJ, et al. Prognostic factors in colorectal cancer. College of American pathologists consensus statement 1999. Arch. Pathol. Lab. Med. 2000;124:979–994.
    1. Li L, Zhou W, Li C, et al. Analysis of circulating tumor DNA to monitor disease status in colorectal cancer after surgery. J. Clin. Oncol. 2018;36:e15583. doi: 10.1200/JCO.2018.36.15_suppl.e15583.
    1. Overman MJ, Vauthey J-N, Aloia TA, et al. Circulating tumor DNA (ctDNA) utilizing a high-sensitivity panel to detect minimal residual disease post liver hepatectomy and predict disease recurrence. J. Clin. Oncol. 2017;35(Suppl. 15):3522. doi: 10.1200/JCO.2017.35.15_suppl.3522.
    1. Tie J, Cohen JD, Wang Y, et al. Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage III colon cancer. JAMA Oncol. 2019;5:1710–1717. doi: 10.1001/jamaoncol.2019.3616.
    1. Wang Y, Li L, Cohen JD, et al. Prognostic potential of circulating tumor DNA measurement in postoperative surveillance of nonmetastatic colorectal cancer. JAMA Oncol. 2019;5:1118–1123. doi: 10.1001/jamaoncol.2019.0512.
    1. Reinert T, Henriksen TV, Christensen E, et al. Analysis of plasma cell-free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer. JAMA Oncol. 2019;5:1124–1131. doi: 10.1001/jamaoncol.2019.0528.
    1. Wan JCM, Massie C, Garcia-Corbacho J, et al. Liquid biopsies come of age: towards implementation of circulating tumour DNA. Nat. Rev. Cancer. 2017;17:223–238. doi: 10.1038/nrc.2017.7.
    1. Kopetz S, McDonough SL, Morris VK, et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG 1406) J. Clin. Oncol. 2017;35:520. doi: 10.1200/JCO.2017.35.4_suppl.520.
    1. Kopetz S, Grothey A, Yaeger R, et al. Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer. N. Engl. J. Med. 2019;381:1632–1643. doi: 10.1056/NEJMoa1908075.
    1. Dasari A, Grothey A, Kopetz S. Circulating tumor DNA-defined minimal residual disease in solid tumors: opportunities to accelerate the development of adjuvant therapies. J. Clin. Oncol. 2018;36:JCO2018789032. doi: 10.1200/JCO.2018.78.9032.
    1. Hong DS, Morris VK, El Osta B, et al. Phase IB study of vemurafenib in combination with irinotecan and cetuximab in patients with metastatic colorectal cancer with BRAFV600E mutation. Cancer Discov. 2016;6:1352–1365. doi: 10.1158/-16-0050.
    1. Grothey A, Sobrero AF, Shields AF, et al. Duration of adjuvant chemotherapy for stage III colon cancer. N. Engl. J. Med. 2018;378:1177–1188. doi: 10.1056/NEJMoa1713709.
    1. Sargent D, Sobrero A, Grothey A, et al. Evidence for cure by adjuvant therapy in colon cancer: observations based on individual patient data from 20,898 patients on 18 randomized trials. J. Clin. Oncol. 2009;27:872–877. doi: 10.1200/JCO.2008.19.5362.
    1. Sawai CM, Babovic S, Upadhaya S, et al. Hematopoietic stem cells are the major source of multilineage hematopoiesis in adult animals. Immunity. 2016;45:597–609. doi: 10.1016/j.immuni.2016.08.007.
    1. Pang WW, Price EA, Sahoo D, et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc. Natl Acad. Sci. USA. 2011;108:20012–20007. doi: 10.1073/pnas.1116110108.
    1. Genovese G, Kahler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 2014;371:2477–2487. doi: 10.1056/NEJMoa1409405.
    1. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 2014;371:2488–2498. doi: 10.1056/NEJMoa1408617.
    1. Young AL, Challen GA, Birmann BM, et al. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat. Commun. 2016;7:12484. doi: 10.1038/ncomms12484.
    1. Chen S, Wang Q, Yu H, et al. Mutant p53 drives clonal hematopoiesis through modulating epigenetic pathway. Nat. Commun. 2019;10:5649. doi: 10.1038/s41467-019-13542-2.
    1. Gibson CJ, Steensma DP. New insights from studies of clonal hematopoiesis. Clin. Cancer Res. 2018;24:4633–4642. doi: 10.1158/1078-0432.CCR-17-3044.
    1. Severson EA, Riedlinger GM, Connelly CF, et al. Detection of clonal hematopoiesis of indeterminate potential in clinical sequencing of solid tumor specimens. Blood. 2018;131:2501–2505. doi: 10.1182/blood-2018-03-840629.
    1. Schou JV, Larsen FO, Sorensen BS, et al. Circulating cell-free DNA as predictor of treatment failure after neoadjuvant chemo-radiotherapy before surgery in patients with locally advanced rectal cancer. Ann. Oncol. 2018;29:610–615. doi: 10.1093/annonc/mdx778.
    1. Tie J, Cohen JD, Wang Y, et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: a prospective biomarker study. Gut. 2019;68:663–671. doi: 10.1136/gutjnl-2017-315852.
    1. Khakoo S, Carter PD, Brown G, et al. MRI tumor regression grade and circulating tumor DNA as complementary tools to assess response and guide therapy adaptation in rectal cancer. Clin. Cancer Res. 2020;26:183–192. doi: 10.1158/1078-0432.CCR-19-1996.
    1. Boysen AK, Schou JV, Spindler KG. Cell-free DNA and preoperative chemoradiotherapy for rectal cancer: a systematic review. Clin. Transl Oncol. 2019;21:874–880. doi: 10.1007/s12094-018-1997-y.
    1. Massihnia D, Pizzutilo EG, Amatu A, et al. Liquid biopsy for rectal cancer: a systematic review. Cancer Treat. Rev. 2019;79:101893. doi: 10.1016/j.ctrv.2019.101893.
    1. van der Valk MJM, Hilling DE, Bastiaannet E, et al. Long-term outcomes of clinical complete responders after neoadjuvant treatment for rectal cancer in the International Watch & Wait Database (IWWD): an international multicentre registry study. Lancet. 2018;391:2537–2545. doi: 10.1016/S0140-6736(18)31078-X.
    1. Diaz LA, Jr, Bardelli A. Liquid biopsies: genotyping circulating tumor DNA. J. Clin. Oncol. 2014;32:579–586. doi: 10.1200/JCO.2012.45.2011.
    1. Tie J, Kinde I, Wang Y, et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann. Oncol. 2015;26:1715–1722. doi: 10.1093/annonc/mdv177.
    1. Vidal J, Muinelo L, Dalmases A, et al. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann. Oncol. 2017;28:1325–1332. doi: 10.1093/annonc/mdx125.
    1. Garlan F, Laurent-Puig P, Sefrioui D, et al. Early evaluation of circulating tumor DNA as marker of therapeutic efficacy in metastatic colorectal cancer patients (PLACOL Study) Clin. Cancer Res. 2017;23:5416–5425. doi: 10.1158/1078-0432.CCR-16-3155.
    1. Wong AL, Lim JS, Sinha A, et al. Tumour pharmacodynamics and circulating cell free DNA in patients with refractory colorectal carcinoma treated with regorafenib. J. Transl Med. 2015;13:57. doi: 10.1186/s12967-015-0405-4.
    1. Vandeputte C, Kehagias P, El Housni H, et al. Circulating tumor DNA in early response assessment and monitoring of advanced colorectal cancer treated with a multi-kinase inhibitor. Oncotarget. 2018;9:17756–17769. doi: 10.18632/oncotarget.24879.
    1. Lima Pereira A, Lam M, Marie PK, Raghav K, Morris VK, et al. Circulating tumor DNA (ctDNA) as an early marker to monitor clinical benefit of regorafenib and TAS-102 in patients with metastatic colorectal cancer (mCRC) J. Clin. Oncol. 2018;36:3533. doi: 10.1200/JCO.2018.36.15_suppl.3533.
    1. Snyder A, Morrissey MP, Hellmann MD. Use of circulating tumor DNA for cancer immunotherapy. Clin. Cancer Res. 2019;25:6909–6915. doi: 10.1158/1078-0432.CCR-18-2688.
    1. Cabel L, Proudhon C, Romano E, et al. Clinical potential of circulating tumour DNA in patients receiving anticancer immunotherapy. Nat. Rev. Clin. Oncol. 2018;15:639–650. doi: 10.1038/s41571-018-0074-3.
    1. Lee JH, Long GV, Menzies AM, et al. Association between circulating tumor DNA and pseudoprogression in patients with metastatic melanoma treated with anti-programmed cell death 1 antibodies. JAMA Oncol. 2018;4:717–721. doi: 10.1001/jamaoncol.2017.5332.
    1. Cabel L, Riva F, Servois V, et al. Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study. Ann. Oncol. 2017;28:1996–2001. doi: 10.1093/annonc/mdx212.
    1. Bachet JB, Bouche O, Taieb J, et al. RAS mutation analysis in circulating tumor DNA from patients with metastatic colorectal cancer: the AGEO RASANC prospective multicenter study. Ann. Oncol. 2018;29:1211–1219. doi: 10.1093/annonc/mdy061.
    1. Luo H, Zhao Q, Wei W, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci. Transl Med. 2020;12:eaax7533. doi: 10.1126/scitranslmed.aax7533.
    1. Symonds EL, Pedersen SK, Murray D, et al. Circulating epigenetic biomarkers for detection of recurrent colorectal cancer. Cancer. 2020;126:1460–1469. doi: 10.1002/cncr.32695.
    1. Gai W, Ji L, Lam WKJ, et al. Liver- and colon-specific DNA methylation markers in plasma for investigation of colorectal cancers with or without liver metastases. Clin. Chem. 2018;64:1239–1249. doi: 10.1373/clinchem.2018.290304.
    1. Mouliere F, Chandrananda D, Piskorz AM, et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl Med. 2018;10:eaat4921. doi: 10.1126/scitranslmed.aat4921.
    1. Wang X, Shi XQ, Zeng PW, et al. Circulating cell free DNA as the diagnostic marker for colorectal cancer: a systematic review and meta-analysis. Oncotarget. 2018;9:24514–24524. doi: 10.18632/oncotarget.25314.
    1. Thomsen CB, Hansen TF, Andersen RF, et al. Early identification of treatment effect by methylated circulating tumour DNA in metastatic colorectal cancer. Ann. Oncol. 2019;30(Suppl. 5):V201. doi: 10.1093/annonc/mdz246.007.
    1. Strickler JH, Zemla T, Ou F-S, et al. Trastuzumab and tucatinib for the treatment of HER2 amplified metastatic colorectal cancer (mCRC): initial results from the MOUNTAINEER trial. Ann. Oncol. 2019;30(Suppl. 5):V200. doi: 10.1093/annonc/mdz246.005.
    1. Batzer MA, Deininger PL. Alu repeats and human genomic diversity. Nat. Rev. Genet. 2002;3:370–379. doi: 10.1038/nrg798.
    1. Paz N, Levanon EY, Amariglio N, et al. Altered adenosine-to-inosine RNA editing in human cancer. Genome Res. 2007;17:1586–1595. doi: 10.1101/gr.6493107.
    1. Di Ruocco F, Basso V, Rivoire M, et al. Alu RNA accumulation induces epithelial-to-mesenchymal transition by modulating miR-566 and is associated with cancer progression. Oncogene. 2018;37:627–637. doi: 10.1038/onc.2017.369.
    1. Lazzari E, Mondala PK, Santos ND, et al. Alu-dependent RNA editing of GLI1 promotes malignant regeneration in multiple myeloma. Nat. Commun. 2017;8:1922. doi: 10.1038/s41467-017-01890-w.
    1. Hao TB, Shi W, Shen XJ, et al. Circulating cell-free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer. Br. J. Cancer. 2014;111:1482–1489. doi: 10.1038/bjc.2014.470.
    1. El-Gayar D, El-Abd N, Hassan N, et al. Increased free circulating DNA integrity Index as a serum biomarker in patients with colorectal carcinoma. Asian Pac. J. Cancer Prev. 2016;17:939–944. doi: 10.7314/APJCP.2016.17.3.939.
    1. Lanman RB, Mortimer SA, Zill OA, 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. doi: 10.1371/journal.pone.0140712.
    1. Sacher AG, Paweletz C, Dahlberg SE, et al. 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. Kim ST, Lee WS, Lanman RB, et al. Prospective blinded study of somatic mutation detection in cell-free DNA utilizing a targeted 54-gene next generation sequencing panel in metastatic solid tumor patients. Oncotarget. 2015;6:40360–40369. doi: 10.18632/oncotarget.5465.
    1. Sepulveda AR, Hamilton SR, Allegra CJ, et al. Molecular biomarkers for the evaluation of colorectal cancer: guideline from the American society for clinical pathology, college of American pathologists, association for molecular pathology and American society of clinical oncology. J. Mol. Diagn. 2017;19:187–225. doi: 10.1016/j.jmoldx.2016.11.001.
    1. Meric-Bernstam F, Hurwitz H, Raghav KPS, et al. Pertuzumab plus trastuzumab for HER2-amplified metastatic colorectal cancer (MyPathway): an updated report from a multicentre, open-label, phase 2a, multiple basket study. Lancet Oncol. 2019;20:518–530. doi: 10.1016/S1470-2045(18)30904-5.
    1. Nakamura Y, Okamoto W, Kato T, et al. 526PD - TRIUMPH: primary efficacy of a phase II trial of trastuzumab (T) and pertuzumab (P) in patients (pts) with metastatic colorectal cancer (mCRC) with HER2 (ERBB2) amplification (amp) in tumour tissue or circulating tumour DNA (ctDNA): a GOZILA sub-study. Ann. Oncol. 2019;30:v199–v200. doi: 10.1093/annonc/mdz246.004.
    1. Montagut C, Tsui DW, Diaz LA., Jr Detection of somatic RAS mutations in circulating tumor DNA from metastatic colorectal cancer patients: are we ready for clinical use? Ann. Oncol. 2018;29:1083–1084. doi: 10.1093/annonc/mdy091.
    1. Russo M, Siravegna G, Blaszkowsky LS, et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 2016;6:147–153. doi: 10.1158/-15-1283.
    1. Montagut C, Argiles G, Ciardiello F, et al. Efficacy of Sym004 in patients with metastatic colorectal cancer with acquired resistance to anti-EGFR therapy and molecularly selected by circulating tumor DNA Analyses: a phase 2 randomized clinical trial. JAMA Oncol. 2018;4:e175245. doi: 10.1001/jamaoncol.2017.5245.
    1. Strickler JH, Loree JM, Ahronian LG, et al. Genomic landscape of cell-free DNA in patients with colorectal cancer. Cancer Discov. 2018;8:164–173. doi: 10.1158/-17-1009.
    1. Morelli MP, Overman MJ, Dasari A, 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;26:731–736. doi: 10.1093/annonc/mdv005.
    1. Arena S, Bellosillo B, Siravegna G, et al. Emergence of multiple EGFR extracellular mutations during cetuximab treatment in colorectal cancer. Clin. Cancer Res. 2015;21:2157–2166. doi: 10.1158/1078-0432.CCR-14-2821.
    1. Misale S, Yaeger R, Hobor S, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486:532–536. doi: 10.1038/nature11156.
    1. Diaz LA, Jr., Williams RT, Wu J, et al. The molecular evolution of acquired resistance to targeted EGFR blockade in colorectal cancers. Nature. 2012;486:537–540. doi: 10.1038/nature11219.
    1. Van Emburgh BO, Arena S, Siravegna G, et al. Acquired RAS or EGFR mutations and duration of response to EGFR blockade in colorectal cancer. Nat. Commun. 2016;7:13665. doi: 10.1038/ncomms13665.
    1. Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat. Med. 2015;21:795–801. doi: 10.1038/nm.3870.
    1. Montagut C, Dalmases A, Bellosillo B, et al. Identification of a mutation in the extracellular domain of the Epidermal Growth Factor Receptor conferring cetuximab resistance in colorectal cancer. Nat. Med. 2012;18:221–223. doi: 10.1038/nm.2609.
    1. Santini D, Vincenzi B, Addeo R, et al. Cetuximab rechallenge in metastatic colorectal cancer patients: how to come away from acquired resistance? Ann. Oncol. 2012;23:2313–2318. doi: 10.1093/annonc/mdr623.
    1. Parseghian CM, Loree JM, Morris VK, et al. Anti-EGFR-resistant clones decay exponentially after progression: implications for anti-EGFR re-challenge. Ann. Oncol. 2019;30:243–249. doi: 10.1093/annonc/mdy509.
    1. Loree JM, Bailey AM, Johnson AM, et al. Molecular landscape of ERBB2/ERBB3 mutated colorectal cancer. J. Natl Cancer. Inst. 2018;110:1409–1417. doi: 10.1093/jnci/djy067.
    1. Sartore-Bianchi A, Trusolino L, Martino C, et al. Dual-targeted therapy with trastuzumab and lapatinib in treatment-refractory, KRAS codon 12/13 wild-type, HER2-positive metastatic colorectal cancer (HERACLES): a proof-of-concept, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016;17:738–746. doi: 10.1016/S1470-2045(16)00150-9.
    1. Siravegna G, Lazzari L, Crisafulli G, et al. Radiologic and genomic evolution of individual metastases during HER2 blockade in colorectal cancer. Cancer Cell. 2018;34:148–162.e7. doi: 10.1016/j.ccell.2018.06.004.
    1. Corcoran RB, Andre T, Atreya CE, et al. Combined BRAF, EGFR, and MEK inhibition in patients with BRAFV600E-mutant colorectal cancer. Cancer Discov. 2018;8:428–443. doi: 10.1158/-17-1226.
    1. Van Cutsem E, Huijberts S, Grothey A, et al. Binimetinib, encorafenib, and cetuximab triplet therapy for patients with BRAF V600E-mutant metastatic colorectal cancer: safety lead-in results from the phase III BEACON colorectal cancer study. J. Clin. Oncol. 2019;37:1460–1469. doi: 10.1200/JCO.18.02459.
    1. Pabla S, Andreas J, Lenzo FL, et al. Development and analytical validation of a next-generation sequencing based microsatellite instability (MSI) assay. Oncotarget. 2019;10:5181–5193. doi: 10.18632/oncotarget.27142.
    1. Georgiadis A, Durham JN, Keefer LA, et al. Noninvasive detection of microsatellite instability and high tumor mutation burden in cancer patients treated with PD-1 blockade. Clin. Cancer Res. 2019;25:7024–7034. doi: 10.1158/1078-0432.CCR-19-1372.
    1. Willis J, Lefterova MI, Artyomenko A, et al. Validation of microsatellite instability detection using a comprehensive plasma-based genotyping panel. Clin. Cancer Res. 2019;25:7035–7045. doi: 10.1158/1078-0432.CCR-19-1324.

Source: PubMed

3
구독하다