Skip to main content

Advertisement

Log in

Peritoneal Cell-Free Tumor DNA as Biomarker for Peritoneal Surface Malignancies

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

Abstract

Background

Disease burden in patients with peritoneal carcinomatosis (PC) is difficult to estimate. We evaluate whether peritoneal cell-free tumor DNA can be used as a measure of disease burden.

Patients and Methods

Malignant ascites or peritoneal lavage fluids were collected from patients with PC under approved IRB protocol. Cell-free DNA was extracted from peritoneal fluid. Droplet digital PCR (ddPCR) was performed using a commercially available KRAS G12/G13 screening kit. Mutant allele frequency (MAF) was calculated based on the numbers of KRAS wild-type and mutant droplets. Clinicopathological, treatment and outcome data were abstracted and correlated with MAF of cell-free KRAS mutant DNA.

Results

Cell-free KRAS mutant DNA was detected in 15/37 (40%) malignant peritoneal fluids with a MAF of 0.1% to 26.2%. While peritoneal cell-free KRAS mutant DNA was detected in all the patients with KRAS mutant tumors (N = 10), 3/16 (19%) patients with KRAS wild-type tumors also had peritoneal cell-free KRAS mutant DNA. We also found that 71% (5/7) of patients with disease amenable to cytoreductive surgery (CRS) had a MAF of < 1% (median: 0.5%, range: 0.1–4.7%), while 75% (6/8) of patients with unresectable disease had a MAF of > 1% (median: 4.4%, range: 0.1–26.2%).

Conclusions

This pilot proof-of-principle study suggests that peritoneal cell-free tumor DNA detected by ddPCR may enable prediction of disease burden and a measure of disease amenable to CRS in patients with PC.

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
Fig. 3

Similar content being viewed by others

References

  1. de Bree E, Koops W, Kroger R, van Ruth S, Witkamp AJ, Zoetmulder FA. Peritoneal carcinomatosis from colorectal or appendiceal origin: correlation of preoperative CT with intraoperative findings and evaluation of interobserver agreement. J Surg Oncol. 2004;86(2):64–73.

    Article  Google Scholar 

  2. Pasqual EM, Bertozzi S, Bacchetti S, et al. Preoperative assessment of peritoneal carcinomatosis in patients undergoing hyperthermic intraperitoneal chemotherapy following cytoreductive surgery. Anticancer Res. 2014;34(5):2363–8.

    PubMed  Google Scholar 

  3. Stroun M, Anker P, Lyautey J, Lederrey C, Maurice PA. Isolation and characterization of DNA from the plasma of cancer patients. Eur J Cancer Clin Oncol. 1987;23(6):707–12.

    Article  CAS  Google Scholar 

  4. Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14(9):985–90.

    Article  CAS  Google Scholar 

  5. Francis G, Stein S. Circulating cell-free tumour DNA in the management of cancer. Int J Mol Sci. 2015;16(6):14122–42.

    Article  CAS  Google Scholar 

  6. Hamakawa T, Kukita Y, Kurokawa Y, et al. Monitoring gastric cancer progression with circulating tumour DNA. Br J Cancer. 2015;112(2):352–6.

    Article  CAS  Google Scholar 

  7. Murtaza M, Dawson SJ, Tsui DW, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497(7447):108–12.

    Article  CAS  Google Scholar 

  8. Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368(13):1199–209.

    Article  CAS  Google Scholar 

  9. Buscail L, Bournet B, Cordelier P. Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2020.

  10. Shoda K, Masuda K, Ichikawa D, et al. HER2 amplification detected in the circulating DNA of patients with gastric cancer: a retrospective pilot study. Gastric Cancer. 2015;18(4):698–710.

    Article  CAS  Google Scholar 

  11. De Mattos-Arruda L, Olmos D, Tabernero J. Prognostic and predictive roles for circulating biomarkers in gastrointestinal cancer. Future Oncol. 2011;7(12):1385–97.

    Article  Google Scholar 

  12. Schwaederle M, Chattopadhyay R, Kato S, et al. Genomic alterations in circulating tumor DNA from diverse cancer patients identified by next-generation sequencing. Cancer Res. 2017;77(19):5419–27.

    Article  CAS  Google Scholar 

  13. Boysen AK, Wettergren Y, Sorensen BS, Taflin H, Gustavson B, Spindler KG. Cell-free DNA levels and correlation to stage and outcome following treatment of locally advanced rectal cancer. Tumour Biol. 2017;39(11):1010428317730976.

    Article  Google Scholar 

  14. Spindler KL, 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(9):2215–22.

    Article  CAS  Google Scholar 

  15. Perets R, Greenberg O, Shentzer T, et al. Mutant KRAS circulating tumor DNA is an accurate tool for pancreatic cancer monitoring. Oncologist. 2018;23(5):566–72.

    Article  CAS  Google Scholar 

  16. Hadano N, Murakami Y, Uemura K, et al. Prognostic value of circulating tumour DNA in patients undergoing curative resection for pancreatic cancer. Br J Cancer. 2016;115(1):59–65.

    Article  CAS  Google Scholar 

  17. 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  Google Scholar 

  18. Brychta N, Krahn T, von Ahsen O. Detection of KRAS mutations in circulating tumor DNA by digital PCR in early stages of pancreatic cancer. Clin Chem. 2016;62(11):1482–91.

    Article  CAS  Google Scholar 

  19. Spindler KL, Pallisgaard N, Vogelius I, Jakobsen A. Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan. Clin Cancer Res. 2012;18(4):1177–85.

    Article  CAS  Google Scholar 

  20. Fan G, Zhang K, Yang X, Ding J, Wang Z, Li J. Prognostic value of circulating tumor DNA in patients with colon cancer: Systematic review. PloS one. 2017;12(2):e0171991.

    Article  Google Scholar 

  21. Cheng H, Luo G, Jin K, et al. Kras mutation correlating with circulating regulatory T cells predicts the prognosis of advanced pancreatic cancer patients. Cancer Med. 2020.

  22. Portilla AG, Shigeki K, Dario B, Marcello D. The intraoperative staging systems in the management of peritoneal surface malignancy. J Surg Oncol. 2008;98(4):228–31.

    Article  Google Scholar 

  23. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–92.

    Article  CAS  Google Scholar 

  24. Normanno N, Rachiglio AM, Lambiase M, et al. Heterogeneity of KRAS, NRAS, BRAF and PIK3CA mutations in metastatic colorectal cancer and potential effects on therapy in the CAPRI GOIM trial. Ann Oncol. 2015;26(8):1710–4.

    Article  CAS  Google Scholar 

  25. Sclafani F, Chau I, Cunningham D, et al. KRAS and BRAF mutations in circulating tumour DNA from locally advanced rectal cancer. Sci Rep. 2018;8(1):1445.

    Article  Google Scholar 

  26. Watanabe F, Suzuki K, Tamaki S, et al. Longitudinal monitoring of KRAS-mutated circulating tumor DNA enables the prediction of prognosis and therapeutic responses in patients with pancreatic cancer. PloS one. 2019;14(12):e0227366.

    Article  CAS  Google Scholar 

  27. Sugimori M, Sugimori K, Tsuchiya H, et al. Quantitative monitoring of circulating tumor DNA in patients with advanced pancreatic cancer undergoing chemotherapy. Cancer Sci. 2020;111(1):266–78.

    Article  CAS  Google Scholar 

  28. Demuth C, Spindler KG, Johansen JS, et al. Measuring KRAS mutations in circulating tumor DNA by droplet digital PCR and next-generation sequencing. Transl Oncol. 2018;11(5):1220–4.

    Article  Google Scholar 

  29. Jung K, Fleischhacker M, Rabien A. Cell-free DNA in the blood as a solid tumor biomarker–a critical appraisal of the literature. Clin Chim Acta. 2010;411(21-22):1611–24.

    Article  CAS  Google Scholar 

  30. Ako S, Nouso K, Kinugasa H, et al. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology. 2017;17(2):285–90.

    Article  CAS  Google Scholar 

  31. Swisher EM, Wollan M, Mahtani SM, et al. Tumor-specific p53 sequences in blood and peritoneal fluid of women with epithelial ovarian cancer. Am J Obstet Gynecol. 2005;193(3 Pt 1):662–667.

    Article  CAS  Google Scholar 

  32. Haupts A, Roth W, Hartmann N. [Liquid biopsy in colorectal cancer: An overview of ctDNA analysis in tumour diagnostics]. Pathologe. 2019;40(Suppl 3):244–51.

    Article  CAS  Google Scholar 

  33. Riva F, Bidard FC, Houy A, et al. Patient-specific circulating tumor DNA detection during neoadjuvant chemotherapy in triple-negative breast cancer. Clin Chem. 2017;63(3):691–9.

    Article  CAS  Google Scholar 

  34. Galbiati S, Damin F, Ferraro L, et al. Microarray approach combined with ddPCR: an useful pipeline for the detection and quantification of circulating tumour DNA mutations. Cells. 2019;8(8):769.

    Article  CAS  Google Scholar 

  35. Pishvaian MJ, Bender RJ, Halverson D, et al. Molecular profiling of patients with pancreatic cancer: initial results from the know your tumor initiative. Clin Cancer Res. 2018;24(20):5018–27.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This study was supported by the Holden Comprehensive Cancer Center through funds from the National Cancer Institute of the National Institutes of Health under award no. P30 CA086862 for supporting the Molecular Epidemiology Resource Core. A.G.K. was supported by the National Heart, Lung, and Blood Institute of the National Institutes of Health under award no. T35 HL007485. A.M.M. was supported by the National Institutes of Health Free Radical and Radiation Biology T32 CA078586 training grant. H.R.S. was supported by the Iowa Center for Research by Undergraduates Fellowship Awards.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlos H. F. Chan MD, PhD.

Ethics declarations

Disclosure

P.M.K.: consultancy/advisory board (Taiho, Ipsen, Foundation Medicine, Natera); Honoraria (AstraZeneca, Foundation Medicine, Natera). Foundation Medicine, a company focusing on genomics and ctDNA in blood/tissue, is not directly related to the submitted work but to ctDNA testing. Natera, a company focusing on ctDNA for stage II/III colorectal cancer, is not directly related to the submitted work but to the field of liquid biopsies. All other authors declare that they have no conflicts of interest.

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 19 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Leick, K.M., Kazarian, A.G., Rajput, M. et al. Peritoneal Cell-Free Tumor DNA as Biomarker for Peritoneal Surface Malignancies. Ann Surg Oncol 27, 5065–5071 (2020). https://doi.org/10.1245/s10434-020-08832-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1245/s10434-020-08832-9

Navigation