Skip to main content

Advertisement

Log in

Prognostic Utility of Pre- and Postoperative Circulating Tumor DNA Liquid Biopsies in Patients with Peritoneal Metastases

  • Translational Research and Biomarkers
  • Published:
Annals of Surgical Oncology Aims and scope Submit manuscript

Abstract

Background

Circulating tumor DNA (ctDNA) is a promising technology for treatment selection, prognostication, and surveillance after definitive therapy. Its use in the perioperative setting for patients with metastatic disease has not been well studied. We characterize perioperative plasma ctDNA and its association with progression-free survival (PFS) in patients undergoing surgery for peritoneal metastases.

Patients and Methods

We recruited 71 patients undergoing surgery for peritoneal metastases and evaluated their plasma with a targeted 73-gene ctDNA next-generation sequencing test before and after surgery. The association between perioperative ctDNA, as well as other patient factors, and PFS was evaluated by Cox regression.

Results

ctDNA was detectable in 28 patients (39.4%) preoperatively and in 37 patients (52.1%) postoperatively. Patients with high ctDNA [maximum somatic variant allele fraction (MSVAF) > 0.25%] had worse PFS than those with low MSVAF (< 0.25%) in both the pre- and postoperative settings (median 4.8 vs. 19.3 months, p < 0.001, and 9.2 vs.15.0 months, p = 0.049, respectively; log-rank test). On multivariate analysis, high-grade histology [hazard ratio (HR) 3.42, p = 0.001], incomplete resection (HR 2.35, p = 0.010), and high preoperative MSVAF (HR 3.04, p = 0.001) were associated with worse PFS. Patients with new postoperative alterations in the context of preoperative alteration(s) also had a significantly shorter PFS compared with other groups (HR 4.28, p < 0.001).

Conclusions

High levels of perioperative ctDNA and new postoperative ctDNA alterations in the context of preoperative alterations predict worse outcomes in patients undergoing resection for peritoneal metastases. This may highlight a role for longitudinal ctDNA surveillance in this population.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Goodall J, Mateo J, Yuan W, et al. Circulating cell-free DNA to guide prostate cancer treatment with PARP inhibition. Cancer Discov. 2017;7(9):1006–1017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ikeda S, Schwaederle M, Mohindra M, Fontes Jardim DL, Kurzrock R. MET alterations detected in blood-derived circulating tumor DNA correlate with bone metastases and poor prognosis. J Hematol Oncol. 2018;11(1):76.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Kato S, Okamura R, Baumgartner JM, et al. Analysis of circulating tumor DNA and clinical correlates in patients with esophageal, gastroesophageal junction, and gastric adenocarcinoma. Clin Cancer Res. 2018;24(24):6248–6256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kato S, Okamura R, Mareboina M, et al. Revisiting epidermal growth factor feceptor (EGFR) amplification as a target for anti-EGFR therapy: analysis of cell-free circulating tumor DNA in patients with advanced malignancies. JCO Precis Oncol. 2019. https://doi.org/10.1200/PO.18.00180.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Kato S, Schwaederle MC, Fanta PT, et al. Genomic assessment of blood-derived circulating tumor DNA in patients with colorectal cancers: correlation with tissue sequencing, therapeutic response, and survival. JCO Precis Oncol. 2019. https://doi.org/10.1200/PO.18.00158.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Riviere P, Fanta PT, Ikeda S, Baumgartner J, Heestand GM, Kurzrock R. The mutational landscape of gastrointestinal malignancies as reflected by circulating tumor DNA. Mol Cancer Ther. 2018;17(1):297–305.

    Article  CAS  PubMed  Google Scholar 

  7. Shatsky R, Parker BA, Bui NQ, et al. Next-Generation sequencing of tissue and circulating tumor DNA: the UC San Diego Moores Center for personalized cancer therapy experience with breast malignancies. Mol Cancer Ther. 2019;18(5):1001–1011.

    Article  CAS  PubMed  Google Scholar 

  8. Tjensvoll K, Lapin M, Buhl T, et al. Clinical relevance of circulating KRAS mutated DNA in plasma from patients with advanced pancreatic cancer. Mol Oncol. 2016;10(4):635–643.

    Article  CAS  PubMed  Google Scholar 

  9. Mardinian K, Okamura R, Kato S, Kurzrock R. Temporal and spatial effects and survival outcomes associated with concordance between tissue and blood KRAS alterations in the pan-cancer setting. Int J Cancer. 2020;146(2):566–576.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Baumgartner JM, Raymond VM, Lanman RB, et al. Preoperative circulating tumor DNA in patients with peritoneal carcinomatosis is an independent predictor of progression-free survival. Ann Surg Oncol. 2018;25(8):2400–2408.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Scholer LV, Reinert T, Orntoft MW, et al. Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin Cancer Res. 2017;23(18):5437–5445.

    Article  CAS  PubMed  Google Scholar 

  12. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–990.

    Article  CAS  PubMed  Google Scholar 

  13. 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(7):827.

    Article  CAS  PubMed  Google Scholar 

  14. Pu WY, Zhang R, Xiao L, et al. Prediction of cancer progression in a group of 73 gastric cancer patients by circulating cell-free DNA. BMC Cancer. 2016;16(1):943.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Sugarbaker PH, Stuart OA, Vidal-Jove J, Pessagno AM, DeBruijn EA. Pharmacokinetics of the peritoneal-plasma barrier after systemic mitomycin C administration. Cancer Treat Res. 1996;82:41–52.

    Article  CAS  PubMed  Google Scholar 

  16. Chua TC, Moran BJ, Sugarbaker PH, et al. Early- and long-term outcome data of patients with pseudomyxoma peritonei from appendiceal origin treated by a strategy of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. J Clin Oncol. 2012;30(20):2449–2456.

    Article  PubMed  Google Scholar 

  17. Yan TD, Deraco M, Baratti D, et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for malignant peritoneal mesothelioma: multi-institutional experience. J Clin Oncol. 2009;27(36):6237–6242.

    Article  PubMed  Google Scholar 

  18. Franko J, Ibrahim Z, Gusani NJ, Holtzman MP, Bartlett DL, Zeh HJ, 3rd. Cytoreductive surgery and hyperthermic intraperitoneal chemoperfusion versus systemic chemotherapy alone for colorectal peritoneal carcinomatosis. Cancer. 2010;116(16):3756–3762.

    Article  PubMed  Google Scholar 

  19. Verwaal VJ, Bruin S, Boot H, van Slooten G, van Tinteren H. 8-year follow-up of randomized trial: cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy in patients with peritoneal carcinomatosis of colorectal cancer. Ann Surg Oncol. 2008;15(9):2426–2432.

    Article  PubMed  Google Scholar 

  20. Baumgartner JM, Tobin L, Heavey SF, Kelly KJ, Roeland EJ, Lowy AM. Predictors of progression in high-grade appendiceal or colorectal peritoneal carcinomatosis after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Ann Surg Oncol. 2015;22(5):1716–1721.

    Article  PubMed  Google Scholar 

  21. 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(10):e0140712.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Odegaard JI, Vincent JJ, Mortimer S, et al. Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies. Clin Cancer Res. 2018;24(15):3539–3549.

    Article  CAS  PubMed  Google Scholar 

  23. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45(2):228–247.

    Article  CAS  PubMed  Google Scholar 

  24. Wickham H. Easily Install and Load the “Tidyverse.” tidyverse;2017.

  25. Maron SB, Chase LM, Lomnicki S, et al. Circulating tumor DNA sequencing analysis of gastroesophageal adenocarcinoma. Clin Cancer Res. 2019;25(23):7098–7112.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Khier S, Lohan L. Kinetics of circulating cell-free DNA for biomedical applications: critical appraisal of the literature. Future Sci OA. 2018;4(4):FSO295.

    Article  Google Scholar 

  27. Yang YC, Wang D, Jin L, et al. Circulating tumor DNA detectable in early- and late-stage colorectal cancer patients. Biosci Rep. 2018. https://doi.org/10.1042/BSR20180322.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Joel M. Baumgartner MD, MAS.

Ethics declarations

Financial support

Funded in part by the National Cancer Institute (grant P30 CA023100) and the Joan and Irwin Jacobs Fund philanthropic fund. Study also funded in part by the Guardant Health (Guardant Health, Inc., Redwood City, CA). The project described was partially supported by the National Institutes of Health (grant TL1TR001443 of CTSA funding beginning August 13, 2015 and beyond). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Disclosures

Razelle Kurzrock discloses 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 Fundingf [Incyte, Genentech, Merck Serono, Pfizer, Sequenom, Foundation Medicine, Guardant Health, Grifols, Konica Minolta, DeBiopharm, Boerhringer Ingelheim, and OmniSeq (all institutional)]; board member (CureMatch, Inc). Paul Riviere discloses consulting fees from Peptide Logic, LLC. Richard Lanman is an employee of Guardant Health, Inc.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 44 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Baumgartner, J.M., Riviere, P., Lanman, R.B. et al. Prognostic Utility of Pre- and Postoperative Circulating Tumor DNA Liquid Biopsies in Patients with Peritoneal Metastases. Ann Surg Oncol 27, 3259–3267 (2020). https://doi.org/10.1245/s10434-020-08331-x

Download citation

  • Received:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-020-08331-x

Keywords

Navigation