Zie ook de literatuurlijst niet-toxische middelen en behandelingen specifiek bij darmkanker van arts-bioloog drs. Engelbert Valstar 

25 juli 2023: Bron: Front. Oncol., 30 January 2023 Sec. Surgical Oncology Volume 13 - 2023

Het regelmatig meten van in bloed circulerend tumor DNA (ctDNA) bij patiënten die behandeld worden voor endeldarmkanker - rectumkanker blijkt een uitstekende biomarker voor een wel of niet optredend recidief na een operatie of tijdens en na de behandeling met meestal chemotherapie. Dat blijkt uit een meta-analyse uitgevoerd bij 19 studies waarvan er 7 overbleven om geanalyseerd te worden wat betreft de relatie tussen het gemeten circulerend tumor DNA (ctDNA) en een optredend recidief. 

Resultaten uit het abstract vertaald :

  • In totaal werden 291 unieke studies gescreend, waarvan 261 originele publicaties en 30 lopende studies. Negentien originele publicaties werden beoordeeld en besproken, waarvan er zeven voldoende gegevens leverden voor meta-analyses over het verband tussen de aanwezigheid van ctDNA na de behandeling en optredend recidief (RFS).
  • De resultaten van de meta-analyses toonden aan dat ctDNA-analyse kan worden gebruikt om patiënten te stratificeren in zeer hoge en lage risicogroepen voor recidief, vooral wanneer gedetecteerd na neoadjuvante behandeling (HR voor RFS: 9,3 [4,6 – 18,8]) en na chirurgie (HR voor RFS: 15,5 [8,2 – 29,3]).
  • Studies onderzochten verschillende soorten assays en gebruikten verschillende technieken voor de detectie en kwantificering van ctDNA.
In Nederland wordt bij vormen van darmkanker waaronder endeldarmkanker en ook slokdarmkanker al enkele jaren bij geselecteerde patiënten circulerend tumor DNA (ctDNA) toegepast. Maar uit verhalen van patiënten blijkt het heel lastig om daarvoor in aanmerking te komen.

De MIRACLE studie in Erasmus MC gebruikt circulerend tumor DNA (ctDNA) om leveruiitzaaiingen bij vormen van darmkanker te traceren en daarmee wellicht te voorkomen met aangepaste behandeling: 

Colorectal cancer patients often develop surgically removable liver metastases. We aim to develop a test to identify patients in need of additional treatment.

Een andere studie die ctDNA metingen gebruikt bij vormen van darmkanker is de DOLPHIN studie: 

DOLPHIN (DNA-testing Of Liquid biopsies for Patient care close to Home In the Netherlands)  

Het abstract van de meta-analyse is dit. Als u op de titel klikt krijgt u het volledige studierapport:

SYSTEMATIC REVIEW article

Front. Oncol., 30 January 2023
Sec. Surgical Oncology
Volume 13 - 2023 | https://doi.org/10.3389/fonc.2023.1083285
This article is part of the Research Topic

Innovations in Surgical Oncology

Circulating tumour DNA as biomarker for rectal cancer: A systematic review and meta-analyses

Jan M. van Rees1* Lissa Wullaert1 Alexander A. J. Grüter2 Yassmina Derraze2 Pieter J. Tanis1 Henk M. W. Verheul3 John W. M. Martens3 Saskia M. Wilting3 Geraldine Vink4,5 Jeroen L. A. van Vugt1 Nick Beije3 Cornelis Verhoef1
  • 1Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, Netherlands
  • 2Department of Surgery, Amsterdam University Medical Centres (UMC), Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
  • 3Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, Netherlands
  • 4Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
  • 5Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, Netherlands

Background: Circulating tumour DNA (ctDNA) has been established as a promising (prognostic) biomarker with the potential to personalise treatment in cancer patients. The objective of this systematic review is to provide an overview of the current literature and the future perspectives of ctDNA in non-metastatic rectal cancer.

Methods: A comprehensive search for studies published prior to the 4th of October 2022 was conducted in Embase, Medline, Cochrane, Google scholar, and Web of Science. Only peer-reviewed original articles and ongoing clinical trials investigating the association between ctDNA and oncological outcomes in non-metastatic rectal cancer patients were included. Meta-analyses were performed to pool hazard ratios (HR) for recurrence-free survival (RFS).

Results: A total of 291 unique records were screened, of which 261 were original publications and 30 ongoing trials. Nineteen original publications were reviewed and discussed, of which seven provided sufficient data for meta-analyses on the association between the presence of post-treatment ctDNA and RFS. Results of the meta-analyses demonstrated that ctDNA analysis can be used to stratify patients into very high and low risk groups for recurrence, especially when detected after neoadjuvant treatment (HR for RFS: 9.3 [4.6 – 18.8]) and after surgery (HR for RFS: 15.5 [8.2 – 29.3]). Studies investigated different types of assays and used various techniques for the detection and quantification of ctDNA.

Conclusions: This literature overview and meta-analyses provide evidence for the strong association between ctDNA and recurrent disease. Future research should focus on the feasibility of ctDNA-guided treatment and follow-up strategies in rectal cancer. A blueprint for agreed-upon timing, preprocessing, and assay techniques is needed to empower adaptation of ctDNA into daily practice.

Discussion

The aim of this literature review was to provide an overview of the current evidence and ongoing trials in the field of ctDNA in non-metastatic rectal cancer. Studies have consistently shown the strong association between detectable ctDNA after treatment and unfavourable prognosis. It can be concluded from these results that ctDNA analysis from peripheral blood samples, especially detected after surgery with curative intent, stratifies patients into two groups: one with a very high risk for recurrence, another with a low risk for recurrence. Thus far, there are no rectal cancer trials published, that have investigated ctDNA-guided adjuvant treatment in a randomised setting.

Based on our systematic search, this systematic review is the first to pool long-term oncological survival outcomes in a meta-analysis. A systematic review by Boyson et al. included nine single arm studies with a total of 615 patients undergoing chemoradiation for rectal cancer and investigated the relation between ctDNA and clinical outcomes (15). Eight of the nine studies showed some degree of correlation between ctDNA and either response to chemoradiation, risk of recurrence or disease-free survival. A second systematic review also included nine studies and investigated the association between clinical outcomes and ctDNA at different time points (at diagnosis, after chemoradiation, and after surgery) (38). No association was found between treatment response and ctDNA status at baseline. Studies reporting the prognostic impact of ctDNA after chemoradiation and before surgery showed varying results. All five studies reporting outcomes of detectable ctDNA postoperative and clinical outcomes, found an association between ctDNA positivity after surgery and worse survival. This review demonstrated that post-operative ctDNA is the most predictive prognostic factor of all investigated time points. A third systematic review investigating different ctDNA measurement techniques on predictive and prognostic outcomes in LARC patients, concluded that detection of ctDNA at different time points of treatment was consistently associated with worse prognosis, but that the ideal method and timing for the liquid biopsy still needed to be defined (39).

Although all studies found a positive correlation between ctDNA and treatment and oncological outcomes, various methods to analyse ctDNA were used, including those with quantitative (e.g. absolute cfDNA concentration) and qualitative (tumour-specific somatic mutations) measurements. Articles that utilized quantitative analyses were generally published between 2008-2018, and were considered relatively inferior because quantitative tests do not have the ability to discriminate tumour DNA from physiological circulating DNA from non-cancerous cells. More recent studies often used qualitative techniques that are able to specifically detect tumour-specific cfDNA. These mutation-specific analyses are nowadays considered as technique of choice, and are acceptable in terms of costs (40). Differences in qualitative analyses exist as well, as was shown as shown by Liu et al. (22) This study revealed that minor differences in the sensitivity of ctDNA are observed when different gene panels and techniques for ctDNA quantification are used, in which a personalised assay targeting tumour-informed mutations was suggested to yield the best performance. However, tumour-informed assays are more expensive and labour-intensive as they require sequencing of the tumour and subsequent design of tumour-specific assays. This can be challenging, especially in a setting where the turnaround time for clinical decision-making needs to be short and will be accompanied by higher costs. A tumour-agnostic method is likely to have a faster turnaround time, as it is easier to conduct, and is accompanied by lower costs. Currently, well-powered studies in a real-world setting comparing all assays with regard to its sensitivity, specificity and turnaround time are lacking.

Another controversy in ctDNA analysis is the optimal timing of measurement to detect MRD after surgery, as it has been suggested that an abundance of surgery‐induced cfDNA fragments could hamper the detection of ctDNA from the tumour (41). In a study by Hendriksen et al., it was shown that cfDNA levels in patients with colorectal cancer were increased by threefold during the first week after surgery (median 3.6‐fold increase, mean: 4.0, 95% CI 2.90–5.37, P = 0.0005), and slowly decreased over the next 3 weeks. Notably, it was assumed that in five of the eight patients, ctDNA was falsely measured as being negative, as these patients were ctDNA positive in all other measurements in which ctDNA surgery‐induced cfDNA fragments were not increased. Therefore, to maximize sensitivity of the measurement, one could argue to only measure ctDNA at least four weeks after surgery. On the other hand, when the results of the ctDNA analyses have clinical consequences, e.g. ctDNA-based adjuvant therapy, results ought to be known within the timeframe that consolidation treatment will still be sufficient. Typically, most ctDNA assays are accompanied by an additional four weeks turnover time from blood withdrawal to definite results (42), so the typical timeframe of a maximum of 8 or 12 weeks from surgery to start with adjuvant treatment could be endangered when delaying the ctDNA result too long (4345). A balance between test sensitivity, and considerations regarding turnaround times inherent to different methods, should be considered for each clinical implication and setting.

Precision biomarkers to predict postoperative outcomes, such as ctDNA, could contribute to the ongoing debate whether additional treatment should be considered after rectal cancer surgery. The role of adjuvant systemic treatment in rectal cancer has not been established globally; practice differs between Europe and the USA, and between European countries as well. In the Netherlands, adjuvant chemotherapy is not recommended for any stage (46). There are only a few randomised controlled trials on adjuvant chemotherapy for rectal cancer available, which yielded conflicting results (47). The fact that the benefit of adjuvant chemotherapy has not yet been demonstrated, is likely related to a dilution effect, and it might very well be true that a subgroup of patients will benefit from additional treatment. Therefore, it would certainly be of interest to explore whether high-risk patients based on ctDNA detected in postoperative peripheral blood samples might benefit from adjuvant treatment. A trial randomising patients with detectable ctDNA into an adjuvant treatment group and a follow-up group is warranted. Such a trial should be able to answer the important question whether ctDNA-guided adjuvant treatment is beneficial in rectal cancer.

Another potential opportunity of ctDNA-guided treatment is the ability to tailor follow-up strategies based on patients’ individual risk of recurrence. As intensive follow-up does not appear to improve overall and cancer-specific survival and quality of life in colorectal cancer, there seems to be an incentive to reduce surveillance after curative surgery (464849). Studies have demonstrated that ctDNA outperforms CEA in (colo)rectal cancer patients to detect relapsing disease (5253150). Therefore, ctDNA-based risk prediction for recurrence may very well be an excellent biomarker to stratify patients without detectable DNA into a less intensive and decentralised surveillance programme in the home environment or even earlier discharge of standard follow-up. This could eventually improve health-related quality of life, cause a reduction in health-related and societal costs as well as anxiety in cancer patients, without compromising oncological outcomes. Further research would be needed to investigate whether this ctDNA-guided follow-up approach is feasible in rectal cancer.

Finally, novel technical advances highlight the promise of several tumour-agnostic ways to detect ctDNA (i.e. without prior tissue-based information) in the future. For example, recent results highlight the merit of circulating cell free (cf)DNA methylation analyses for both detection and classification of many cancer types, including colorectal cancer (5154). Next to methylation profiling, recently discovered “fragmentomics” also shows great promise for the sensitive detection of cancer using cfDNA (5557). Both cfDNA methylation profiling and fragmentomics capture information from a much broader spectrum of the circulating tumour genome, theoretically enabling a higher analytical sensitivity for the detection of minute traces of ctDNA in case of MRD. Supporting this notion, combining features from different molecular levels was shown to have complementary value for MRD detection in colorectal cancer (58).

In conclusion, in rectal cancer patients treated with neoadjuvant treatment and surgery, a very strong association was found between post-treatment detectable ctDNA and recurrent disease as well as overall survival. Randomised controlled trials are needed to investigate whether this ctDNA-informed risk classification could be used during clinical decision making for the purpose of patient-tailored treatment.

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding author.

Author contributions

All authors have significantly contributed to this work. JR, LW, AG, YD, CV, NB were involved in writing the introduction, the systematic search, and the article selection process. JR, LW, NB, JV, SW conducted the quality assessment of the included articles and methodology. GV, PT, HV, JM, CV were part of the writing committee. The manuscript was drafted by JM, LW, AG, YD, and corrected by NB, JV, SW, GV, PT, HV, JM, CV. Supervision was provided by SW, NB and CV. All authors contributed to the article and approved the submitted version.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fonc.2023.1083285/full#supplementary-material

References

1. Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor A, et al. Cancer statistics for the year 2020: An overview. Int J Cancer (2021) 149(4):778–89. doi: 10.1002/ijc.33588

CrossRef Full Text | Google Scholar

2. van Gijn W, Marijnen CA, Nagtegaal ID, Kranenbarg EM, Putter H, Wiggers T, et al. Preoperative radiotherapy combined with total mesorectal excision for resectable rectal cancer: 12-year follow-up of the multicentre, randomised controlled TME trial. Lancet Oncol (2011) 12(6):575–82. doi: 10.1016/S1470-2045(11)70097-3

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Badia-Ramentol J, Linares J, Gómez-Llonin A, Calon A. Minimal residual disease, metastasis and immunity. Biomolecules (2021) 11(2):130. doi: 10.3390/biom11020130

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Litvak A, Cercek A, Segal N, Reidy-Lagunes D, Stadler ZK, Yaeger RD, et al. False-positive elevations of carcinoembryonic antigen in patients with a history of resected colorectal cancer. J Natl Compr Canc Netw (2014) 12(6):907–13. doi: 10.6004/jnccn.2014.0085

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Reinert T, Henriksen TV, Christensen E, Sharma S, Salari R, Sethi H, et al. Analysis of plasma cell-free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer. JAMA Oncol (2019) 5(8):1124–31. doi: 10.1001/jamaoncol.2019.0528

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Diaz LA Jr., Bardelli A. Liquid biopsies: Genotyping circulating tumor DNA. J Clin Oncol (2014) 32(6):579–86. doi: 10.1200/JCO.2012.45.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Trans Med (2014) 6(224):224ra24–ra24. doi: 10.1126/scitranslmed.3007094

CrossRef Full Text | Google Scholar

8. Schøler LV, Reinert T, Ørntoft MW, Kassentoft CG, Árnadóttir SS, Vang S, et al. Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin Cancer Res (2017) 23(18):5437–45. doi: 10.1158/1078-0432.CCR-17-0510

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Tie J, Cohen JD, Lahouel K, Lo SN, Wang Y, Kosmider S, et al. Circulating tumor DNA analysis guiding adjuvant therapy in stage II colon cancer. New Engl J Med (2022) 386(24):2261–27. doi: 10.1056/NEJMoa2200075

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Bujko K, Glynne-Jones R, Bujko M. Does adjuvant fluoropyrimidine-based chemotherapy provide a benefit for patients with resected rectal cancer who have already received neoadjuvant radiochemotherapy? a systematic review of randomised trials. Ann Oncol (2010) 21(9):1743–50. doi: 10.1093/annonc/mdq054

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Breugom AJ, van Gijn W, Muller EW, Berglund Å, van den Broek CBM, Fokstuen T, et al. Adjuvant chemotherapy for rectal cancer patients treated with preoperative (chemo)radiotherapy and total mesorectal excision: A Dutch colorectal cancer group (DCCG) randomized phase III trial. Ann Oncol (2015) 26(4):696–701. doi: 10.1093/annonc/mdu560

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Dasari A, Morris VK, Allegra CJ, Atreya C, Benson AB 3rd, et al. ctDNA applications and integration in colorectal cancer: An NCI colon and rectal-anal task forces whitepaper. Nat Rev Clin Oncol (2020) 17(12):757–70. doi: 10.1038/s41571-020-0392-0

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Connors D, Allen J, Alvarez JD, Boyle J, Cristofanilli M, Hiller C, et al. International liquid biopsy standardization alliance white paper. Crit Rev Oncology/Hematology (2020) 156:103112. doi: 10.1016/j.critrevonc.2020.103112

CrossRef Full Text | Google Scholar

14. van Dessel LF, Beije N, Helmijr JC, Vitale SR, Kraan J, Look MP, et al. Application of circulating tumor DNA in prospective clinical oncology trials - standardization of preanalytical conditions. Mol Oncol (2017) 11(3):295–304. doi: 10.1002/1878-0261.12037

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Boysen AK, Schou JV, Spindler KLG. Cell-free DNA and preoperative chemoradiotherapy for rectal cancer: A systematic review. Clin Transl Oncol (2019) 21(7):874–80. doi: 10.1007/s12094-018-1997-y

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Hayden JA, van der Windt DA, Cartwright JL, Côté P, Bombardier C. Assessing bias in studies of prognostic factors. Ann Intern Med (2013) 158(4):280–6. doi: 10.7326/0003-4819-158-4-201302190-00009

PubMed Abstract | CrossRef Full Text | Google Scholar

17. McGuinness LA, Higgins JPT. Risk-of-bias VISualization (robvis): An r package and shiny web app for visualizing risk-of-bias assessments. Res Synthesis Methods (2020) 12(1):55–61. doi: 10.1002/jrsm.1411

CrossRef Full Text | Google Scholar

18. Zitt M, Müller HM, Rochel M, Schwendinger V, Zitt M, Goebel G, et al. Circulating cell-free DNA in plasma of locally advanced rectal cancer patients undergoing preoperative chemoradiation: A potential diagnostic tool for therapy monitoring. Dis Markers (2008) 25(3):159–65. doi: 10.1155/2008/598071

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Agostini M, Pucciarelli S, Enzo MV, Del Bianco P, Briarava M, Bedin C, et al. Circulating cell-free DNA: A promising marker of pathologic tumor response in rectal cancer patients receiving preoperative chemoradiotherapy. Ann Surg Oncol (2011) 18(9):2461–8. doi: 10.1245/s10434-011-1638-y

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Sun W, Sun Y, Zhu M, Wang Z, Zhang H, Xin Y, et al. The role of plasma cell-free DNA detection in predicting preoperative chemoradiotherapy response in rectal cancer patients. Oncol Rep (2014) 31(3):1466–72. doi: 10.3892/or.2013.2949

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Boysen AK, Wettergren Y, Sorensen BS, Taflin H, Gustavson B, Spindler KLG. Cell-free DNA levels and correlation to stage and outcome following treatment of locally advanced rectal cancer. Tumor Biol (2017) 39(11). doi: 10.1177/1010428317730976

CrossRef Full Text | Google Scholar

22. Liu W, Li Y, Tang Y, Song Q, Wang J, Li N, et al. Response prediction and risk stratification of patients with rectal cancer after neoadjuvant therapy through an analysis of circulating tumour DNA. eBioMedicine (2022) 78:103945. doi: 10.1016/j.ebiom.2022.103945

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Sclafani F, Chau I, Cunningham D, Hahne JC, Vlachogiannis G, Eltahir Z, et al. KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Sci Rep (2018) 8(1):1445. doi: 10.1038/s41598-018-19212-5

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Schou JV, Larsen FO, Sørensen BS, Abrantes R, Boysen AK, Johansen JS, 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(3):610–5. doi: 10.1093/annonc/mdx778

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Tie J, Cohen JD, Wang Y, Li L, Christie M, Simons K, et al. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: A prospective biomarker study. Gut (2019) 68(4):663–71. doi: 10.1136/gutjnl-2017-315852

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Appelt AL, Andersen RF, Lindebjerg J, Jakobsen A. Prognostic value of serum NPY hypermethylation in neoadjuvant chemoradiotherapy for rectal cancer: Secondary analysis of a randomized trial. Am J Clin Oncol Cancer Clin Trials (2020) 43(1):9–13. doi: 10.1097/COC.0000000000000609

CrossRef Full Text | Google Scholar

27. Guo ZW, Xiao WW, Yang XX, Yang X, Cai GX, Wang XJ, et al. Noninvasive prediction of response to cancer therapy using promoter profiling of circulating cell-free DNA. Clin Transl Med (2020) 10(5):e174. doi: 10.1002/ctm2.174

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Khakoo S, Carter PD, Brown G, Valeri N, Picchia S, Bali MA, 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(1):183–92. doi: 10.1158/1078-0432.CCR-19-1996

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Murahashi S, Akiyoshi T, Sano T, Fukunaga Y, Noda T, Ueno M, et al. Serial circulating tumour DNA analysis for locally advanced rectal cancer treated with preoperative therapy: Prediction of pathological response and postoperative recurrence. Br J Cancer (2020) 123(5):803–10. doi: 10.1038/s41416-020-0941-4

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Pazdirek F, Minarik M, Benesova L, Halkova T, Belsanova B, Macek M, et al. Monitoring of early changes of circulating tumor DNA in the plasma of rectal cancer patients receiving neoadjuvant concomitant chemoradiotherapy: Evaluation for prognosis and prediction of therapeutic response. Front Oncol (2020) 10. doi: 10.3389/fonc.2020.01028

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Zhou J, Wang C, Lin G, Xiao Y, Jia W, Xiao G, et al. Serial circulating tumor DNA in predicting and monitoring the effect of neoadjuvant chemoradiotherapy in patients with rectal cancer: A prospective multicenter study. Clin Cancer Res (2021) 27(1):301–10. doi: 10.1158/1078-0432.CCR-20-2299

PubMed Abstract | CrossRef Full Text | Google Scholar

32. McDuff SGR, Hardiman KM, Ulintz PJ, Parikh AR, Zheng H, Kim DW, et al. Circulating tumor dna predicts pathologic and clinical outcomes following neoadjuvant chemoradiation and surgery for patients with locally advanced rectal cancer. JCO Precis Oncol (2021) 5:123–32. doi: 10.1200/PO.20.00220

CrossRef Full Text | Google Scholar

33. Vidal J, Casadevall D, Bellosillo B, Pericay C, Garcia-Carbonero R, Losa F, et al. Clinical impact of presurgery circulating tumor DNA after total neoadjuvant treatment in locally advanced rectal cancer: A biomarker study from the GEMCAD 1402 trial. Clin Cancer Res (2021) 27(10):2890–8. doi: 10.1158/1078-0432.CCR-20-4769

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Wang Y, Yang L, Bao H, Fan X, Xia F, Wan J, et al. Utility of ctDNA in predicting response to neoadjuvant chemoradiotherapy and prognosis assessment in locally advanced rectal cancer: A prospective cohort study. PLoS Med (2021) 18(8):e1003741. doi: 10.1371/journal.pmed.1003741

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Roesel R, Epistolio S, Molinari F, Saletti P, De Dosso S, Valli M, et al. A pilot, prospective, observational study to investigate the value of NGS in liquid biopsies to predict tumor response after neoadjuvant chemo-radiotherapy in patients with locally advanced rectal cancer: The LiBReCa study. Front Oncol (2022) 12:900945. doi: 10.3389/fonc.2022.900945

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Truelsen CG, Kronborg CS, Sorensen BS, Callesen LB, Spindler KG. Circulating cell-free DNA as predictor of pathological complete response in locally advanced rectal cancer patients undergoing preoperative chemoradiotherapy. Clin Transl Radiat Oncol (2022) 36:9–15. doi: 10.1016/j.ctro.2022.06.002

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Tie J. Use of circulating tumour DNA (ctDNA) results to inform the decision for adjuvant chemotherapy in patients with locally advanced rectal cancer who have been treated with pre-operative chemo-radiation and surgery. (2017).

Google Scholar

38. Dizdarevic E, Hansen TF, Jakobsen A. The prognostic importance of ctDNA in rectal cancer: A critical reappraisal. Cancers (2022) 14(9):2252. doi: 10.3390/cancers14092252

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Morais M, Pinto DM, Machado JC, Carneiro S. ctDNA on liquid biopsy for predicting response and prognosis in locally advanced rectal cancer: A systematic review. Eur J Surg Oncol (2022) 48(1):218–27. doi: 10.1016/j.ejso.2021.08.034

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Pascual J, Attard G, Bidard FC, Curigliano G, De Mattos-Arruda L, Diehn M, et al. ESMO recommendations on the use of circulating tumour DNA assays for patients with cancer: A report from the ESMO precision medicine working group. Ann Oncol (2022) 33(8):750–68. doi: 10.1016/j.annonc.2022.05.520

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Henriksen TV, Reinert T, Christensen E, Sethi H, Birkenkamp-Demtröder K, Gögenur M, et al. The effect of surgical trauma on circulating free DNA levels in cancer patients-implications for studies of circulating tumor DNA. Mol Oncol (2020) 14(8):1670–9. doi: 10.1002/1878-0261.12729

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Chakrabarti S, Kasi AK, Parikh AR, Mahipal A. Finding Waldo: The evolving paradigm of circulating tumor DNA (ctDNA)-guided minimal residual disease (MRD) assessment in colorectal cancer (CRC). Cancers (Basel) (2022) 14(13):3078. doi: 10.3390/cancers14133078

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Grothey A, Sobrero AF, Shields AF, Yoshino T, Paul J, Taieb J, et al. Duration of adjuvant chemotherapy for stage III colon cancer. New Engl J Med (2018) 378(13):1177–88. doi: 10.1056/NEJMoa1713709

PubMed Abstract | CrossRef Full Text | Google Scholar

44. André T, Meyerhardt J, Iveson T, Sobrero A, Yoshino T, Souglakos I, et al. Effect of duration of adjuvant chemotherapy for patients with stage III colon cancer (IDEA collaboration): Final results from a prospective, pooled analysis of six randomised, phase 3 trials. Lancet Oncol (2020) 21(12):1620–9. doi: 10.1016/S1470-2045(20)30527-1

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Iveson TJ, Kerr RS, Saunders MP, Cassidy J, Hollander NH, Tabernero J, et al. 3 versus 6 months of adjuvant oxaliplatin-fluoropyrimidine combination therapy for colorectal cancer (SCOT): an international, randomised, phase 3, non-inferiority trial. Lancet Oncol (2018) 19(4):562–78. doi: 10.1016/S1470-2045(18)30093-7

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Dutch Colorectal Cancer Guideline, version 3.0 (2014). Available at: https://richtlijnen.nhg.org/files/2020-05/colorectaalcarcinoom.pdf. (accessed at 01-12-2022)

Google Scholar

47. Breugom AJ, Swets M, Bosset JF, Collette L, Sainato A, Cionini L, et al. Adjuvant chemotherapy after preoperative (chemo)radiotherapy and surgery for patients with rectal cancer: A systematic review and meta-analysis of individual patient data. Lancet Oncol (2015) 16(2):200–7. doi: 10.1016/S1470-2045(14)71199-4

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Galjart B, Höppener DJ, Aerts J, Bangma CH, Verhoef C, Grünhagen DJ. Follow-up strategy and survival for five common cancers: A meta-analysis. Eur J Cancer (2022) 174:185–99. doi: 10.1016/j.ejca.2022.07.025

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Jeffery M, Hickey BE, Hider PN, See AM. Follow-up strategies for patients treated for non-metastatic colorectal cancer. Cochrane Database Syst Rev (2016) 11(11):CD002200. doi: 10.1002/14651858.CD002200.pub3

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Tie J, Cohen JD, Lo SN, Wang Y, Li L, Christie M, et al. Prognostic significance of postsurgery circulating tumor DNA in nonmetastatic colorectal cancer: Individual patient pooled analysis of three cohort studies. Int J Cancer (2021) 148(4):1014–26. doi: 10.1002/ijc.33312

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Liu MC, Oxnard GR, Klein EA, Swanton C, Seiden MV, Consortium C. Sensitive and specific multi-cancer detection and localization using methylation signatures in cell-free DNA. Ann Oncol (2020) 31(6):745–59. doi: 10.1016/j.annonc.2020.02.011

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Shen SY, Singhania R, Fehringer G, Chakravarthy A, Roehrl MHA, Chadwick D, et al. Sensitive tumour detection and classification using plasma cell-free DNA methylomes. Nature (2018) 563(7732):579–83. doi: 10.1038/s41586-018-0703-0

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Luo H, Zhao Q, Wei W, Zheng L, Yi S, Li G, et al. Circulating tumor DNA methylation profiles enable early diagnosis, prognosis prediction, and screening for colorectal cancer. Sci Transl Med (2020) 12(524):eaax7533. doi: 10.1126/scitranslmed.aax7533

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Wu X, Zhang Y, Hu T, He X, Zou Y, Deng Q, et al. A novel cell-free DNA methylation-based model improves the early detection of colorectal cancer. Mol Oncol (2021) 15(10):2702–14. doi: 10.1002/1878-0261.12942

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Cristiano S, Leal A, Phallen J, Fiksel J, Adleff V, Bruhm DC, et al. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature (2019) 570(7761):385–9. doi: 10.1038/s41586-019-1272-6

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Lo YMD, Han DSC, Jiang P, Chiu RWK. Epigenetics, fragmentomics, and topology of cell-free DNA in liquid biopsies. Science (2021) 372(6538):aaw3616. doi: 10.1126/science.aaw3616

CrossRef Full Text | Google Scholar

57. Mouliere F, Chandrananda D, Piskorz AM, Moore EK, Morris J, Ahlborn LB, et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci Transl Med (2018) 10(466):eaat4921. doi: 10.1126/scitranslmed.aat4921

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Parikh AR, Van Seventer EE, Siravegna G, Hartwig AV, Jaimovich A, He Y, et al. Minimal residual disease detection using a plasma-only circulating tumor DNA assay in patients with colorectal cancer. Clin Cancer Res (2021) 27(20):5586–94. doi: 10.1158/1078-0432.CCR-21-0410

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: Ctdna (circulating tumour DNA), cfDNA (circulating free DNA), rectal cancer, minimal residual disease (MRD), liquid biopsy

Citation: van Rees JM, Wullaert L, Grüter AAJ, Derraze Y, Tanis PJ, Verheul HMW, Martens JWM, Wilting SM, Vink G, van Vugt JLA, Beije N and Verhoef C (2023) Circulating tumour DNA as biomarker for rectal cancer: A systematic review and meta-analyses. Front. Oncol. 13:1083285. doi: 10.3389/fonc.2023.1083285

Received: 28 October 2022; Accepted: 09 January 2023;
Published: 30 January 2023.

Edited by:

Marek Minarik, Elphogene, s.r.o, Czechia

Reviewed by:

Manuela Gariboldi, Fondazione IRCCS Istituto Nazionale Tumori, Italy
Dalong Pang, Georgetown University, United States

Copyright © 2023 van Rees, Wullaert, Grüter, Derraze, Tanis, Verheul, Martens, Wilting, Vink, van Vugt, Beije and Verhoef. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Jan M. van Rees, j.vanrees@erasmusmc.nl

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.





Plaats een reactie ...

Reageer op "ctDNA - circulerend tumor DNA is uitstekende biomarker voor wel of geen toekomstig recidief na operatie en of chemotherapie bij endeldarmkanker - rectumkanker blijkt uit meta-analyse"


Gerelateerde artikelen
 

Gerelateerde artikelen

Personalised medicin en gerichte >> Studiepublicaties van niet-toxische >> UEG oktober 2020: Abstracten >> ESMO 2023: aanbevolen abstracten >> ASCO 2023: aanbevolen abstracten >> Aanvullende niet toxische >> Adagrasib met of zonder cetuximab >> Aflibercept (Zaltrap) toegevoegd >> Avastin - bevacizumab de >> Bestraling - radiotherapie >>