CC BY 4.0 · Glob Med Genet 2020; 07(02): 027-029
DOI: 10.1055/s-0040-1716333
Editorial

Targeted Sequencing Analysis of Matched Cell-Free DNA and White Blood Cells: A Facile Method for Detection of Residual Disease in Gastric Cancer

Feiyu Diao
1   Department of General Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, People's Republic of China
› Author Affiliations
Funding None.
 

Gastric cancer (GC) is one of the most diagnosed cancers in the world and is mainly cured by surgical resection. However, a large proportion of patients still presents with microscopic residual disease after surgical resection. Patients with microscopic residual disease can be offered appropriate treatment to improve their long-term survival, but for such, it is imperative to timely identify those with residual disease after surgery. Unfortunately, the traditional detection methods for residual disease have poor sensitivity. Recently, with the development of liquid biopsy technology, circulating tumor-derived deoxyribonucleic acid (ctDNA) detection has shown great promise in bringing new hope for increasing the detection rate of microscopic residual disease. In a study recently published in Nature Communications, Leal et al developed a novel liquid biopsy strategy to distinguish tumor-specific ctDNA alterations from cell-free DNA variants associated with clonal hematopoiesis and evaluated the utility of ctDNA as a biomarker for predicting the prognosis of postoperative GC patients.

Gastric cancer (GC) is the fifth common cancer and third leading cause of cancer-related death worldwide.[1] Although the incidence of GC has declined rapidly over recent decades; however, compared with other countries, this decline has been less striking in China.[2] Additionally, the prognosis of GC patients remains poor in China.[3] The curative treatment for GC is based on R0 resection with D2 lymphadenectomy.[4] However, a large proportion of patients still presents with microscopic residual disease after surgery and eventually succumb due to local recurrence or distant metastases.[5] Patients with microscopic residual disease can be offered appropriate treatment to improve their long-term survival if they are timely identified.[4] Unfortunately, the currently available detection methods for residual diseases, such as traditional blood biomarker detection and imaging techniques, have poor sensitivity, specificity, and cannot be reliably used.[6]

With the development of liquid biopsy technology, the latest approaches, such as circulating tumor-derived deoxyribonucleic acid (ctDNA) detection via liquid biopsy, may bring new hope for detecting microscopic residual disease after GC surgery. Theoretically, the tumor-specific alterations in the circulation of postoperative cancer patients can dynamically and specifically reflect whether these patients have microscopic residual disease or preclinical metastases.[7] [8] Recently, ctDNA released from cancer cells into the peripheral blood has been noninvasively detected, not only in late-stage cancer patients but also in those at early stage.[9] The most critical step of liquid biopsy is to distinguish tumor-specific ctDNA from large amounts of cell-free DNA. Most of the previous studies only focused on alterations in ctDNA during anticancer treatment and mainly on the occurrence of metastatic disease. All of them have only analyzed a limited number of genomic positions that only represent a small subset of tumor clones.[10] [11] More recent studies have started to apply blood-based deep sequencing approaches to detect white blood cell-derived variants that associate with clonal hematopoiesis in cell-free DNA.[9] [12] Additionally, such studies also used similar approaches to evaluate the white blood cell DNA and cell-free DNA from cancer patients at a single time point.[9] [13] However, no study has yet attempted to evaluate these during anticancer treatment to predict prognosis. In a study recently published in Nature Communications, titled as “White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer,” Leal et al[14] developed a novel liquid biopsy strategy to distinguish tumor-specific ctDNA alterations from cell-free DNA variants associated with clonal hematopoiesis and evaluated the utility of ctDNA as a biomarker for predicting the prognosis of postoperative GC patients.

All GC patients enrolled in that study were from the CRITICS trial (NCT00407186).[15] The plasma from each patient was collected at the time of trial enrollment (baseline), preoperatively, and after surgery but before postoperative chemotherapy (median time after surgery = 6.5 weeks). The novel approach developed in this study was as follows: the cell-free DNA and white blood cells in blood samples were first analyzed by parallel depth sequencing, and then the tumor-specific alterations in blood samples were identified by removing the hematopoietic-associated alterations detected in white blood cells from the cell-free DNA data. The blood samples from 50 GC patients were evaluated using this new approach. Among them, 27 patients harbored tumor-specific alterations at baseline. In addition, the detection of ctDNA variants at baseline did not show statistically significant differences in both event-free and overall survival. Next, the authors evaluated ctDNA levels in the blood samples collected at the baseline and after preoperative chemotherapy. They found that the preoperative ctDNA level was positively correlated with pathological response in GC patients. Finally, the authors evaluated microscopic residual disease in all 20 patients who had blood samples collected after surgery. They found that tumor-specific alterations in cell-free DNA from 4 patients with major tumor responses disappeared completely after surgery. However, postoperative tumor-specific alterations were detectable in 9 out of 16 patients with minor tumor responses. All 11 patients without detectable tumor-specific alterations after surgery were free of recurrence, while 6 out of 9 patients with detectable tumor-specific alterations after surgery developed metastatic disease.

Furthermore, the detection of microscopic residual disease without a white blood cell filter did not predict disease recurrence. In contrast, with a white blood cell filter, a significantly shorter median event-free survival, and a significantly higher risk of disease recurrence for patients with detectable tumor-specific mutations after surgery could be observed. All results mentioned above suggested that microscopic residual disease could be accurately detected by the new detection method developed in the study by Leal et al, and microscopic residual disease could be a predictive indicator for the prognosis of postoperative GC patients.

Patients with microscopic residual disease would need adjuvant treatment after surgery. It is important to diagnose them as soon as possible. At present, we still rely on traditional methods, such as pathological staging and microscopic residual disease scoring system, to estimate the risk of GC recurrence after surgery.[16] However, these methods have some limitations, which prevent them from being used in clinical practice.[16] Furthermore, the sensitivity of currently available imaging and blood biomarker detection methods is poor.[6] The above-discussed study is the first to develop a tissue-independent detection method that detects tumor-specific mutations in the cell-free DNA of GC patients before and after surgery by sequencing the matched white blood cell and cell-free DNA. In this study, the authors first investigated the value of white blood cell and cell-free DNA parallel deep sequencing to detect clonal hematopoiesis-associated cell-free DNA alterations. After that, they used this method to infer the bona fide alterations of tumor longitudinally. Although the detection of microscopic residual disease by analyzing ctDNA has been used in a variety of tumors,[7] [10] the new strategy developed in this study still has many advantages. For example, this strategy can identify ctDNA without tumor tissue at any time point before and after surgery. In addition, its detection data will not be affected by the heterogeneity within the tumor mass. Even though this study only analyzed a small number of GC patients with sufficient plasma samples, the extensive follow-up time of this study was sufficient to accurately determine clinical recurrences. Moreover, although the panel used in this study was not developed for GC, at the baseline time point, the majority of the GC patients were still detected at the baseline time point. About 60% of GC who were not detected at baseline had a diffuse subtype morphology. This finding suggests that the histological features of GC may not be related to tumor-specific DNA shedding. Furthermore, some GC patients did not relapse although their ctDNA results detected by the new method were positive. The best explanation for this observation could be that these patients were cured after receiving adjuvant therapy, but Leal et al did not assess their ctDNA levels after receiving adjuvant therapy.

Overall, the discussed study developed a facile approach for detecting microscopic residual disease in GC by distinguishing ctDNA alterations from other cell-free DNA alterations. However, when using this new approach to detect postoperative microscopic residual disease of GC, there are some limitations, especially in GC patients who have localized diseases. Therefore, in the future, it is necessary to improve this approach by using targeted panels specifically designed for GC and incorporating cell-free DNA fragmentation analyses.


#

Conflict of Interest

None declared.

Note

The author read and approved the final manuscript.


  • Reference

  • 1 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69 (01) 7-34
  • 2 Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics?. Cancer Commun (Lond) 2019; 39 (01) 22
  • 3 Gao K, Wu J. National trend of gastric cancer mortality in China (2003-2015): a population-based study. Cancer Commun (Lond) 2019; 39 (01) 24
  • 4 Wang FH, Shen L, Li J. , et al. The Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer. Cancer Commun (Lond) 2019; 39 (01) 10
  • 5 Songun I, Putter H, Kranenbarg EM, Sasako M, van de Velde CJ. Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol 2010; 11 (05) 439-449
  • 6 Aurello P, Petrucciani N, Antolino L, Giulitti D, D'Angelo F, Ramacciato G. Follow-up after curative resection for gastric cancer: is it time to tailor it?. World J Gastroenterol 2017; 23 (19) 3379-3387
  • 7 McDonald BR, Contente-Cuomo T, Sammut SJ. , et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci Transl Med 2019; 11 (504) eaax7392
  • 8 Shen L. Liquid biopsy: a powerful tool to monitor trastuzumab resistance in HER2-positive metastatic gastric cancer. Cancer Commun (Lond) 2018; 38 (01) 72
  • 9 Phallen J, Sausen M, Adleff V. , et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med 2017; 9 (403) eaan2415
  • 10 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 (346) 346ra92
  • 11 Rago C, Huso DL, Diehl F. , et al. Serial assessment of human tumor burdens in mice by the analysis of circulating DNA. Cancer Res 2007; 67 (19) 9364-9370
  • 12 Xie M, Lu C, Wang J. , et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014; 20 (12) 1472-1478
  • 13 Hu Y, Ulrich BC, Supplee J. , et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res 2018; 24 (18) 4437-4443
  • 14 Leal A, van Grieken NCT, Palsgrove DN. , et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun 2020; 11 (01) 525
  • 15 Cats A, Jansen EPM, van Grieken NCT. , et al; CRITICS investigators. Chemotherapy versus chemoradiotherapy after surgery and preoperative chemotherapy for resectable gastric cancer (CRITICS): an international, open-label, randomised phase 3 trial. Lancet Oncol 2018; 19 (05) 616-628
  • 16 Langer R, Becker K. Tumor regression grading of gastrointestinal cancers after neoadjuvant therapy. Virchows Arch 2018; 472 (02) 175-186

Address for correspondence

Feiyu Diao, MD
Department of General Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University
Guangzhou 510000
People's Republic of China   

Publication History

Article published online:
31 August 2020

© 2020. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/).

Georg Thieme Verlag KG
Stuttgart · New York

  • Reference

  • 1 Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019; 69 (01) 7-34
  • 2 Feng RM, Zong YN, Cao SM, Xu RH. Current cancer situation in China: good or bad news from the 2018 Global Cancer Statistics?. Cancer Commun (Lond) 2019; 39 (01) 22
  • 3 Gao K, Wu J. National trend of gastric cancer mortality in China (2003-2015): a population-based study. Cancer Commun (Lond) 2019; 39 (01) 24
  • 4 Wang FH, Shen L, Li J. , et al. The Chinese Society of Clinical Oncology (CSCO): clinical guidelines for the diagnosis and treatment of gastric cancer. Cancer Commun (Lond) 2019; 39 (01) 10
  • 5 Songun I, Putter H, Kranenbarg EM, Sasako M, van de Velde CJ. Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol 2010; 11 (05) 439-449
  • 6 Aurello P, Petrucciani N, Antolino L, Giulitti D, D'Angelo F, Ramacciato G. Follow-up after curative resection for gastric cancer: is it time to tailor it?. World J Gastroenterol 2017; 23 (19) 3379-3387
  • 7 McDonald BR, Contente-Cuomo T, Sammut SJ. , et al. Personalized circulating tumor DNA analysis to detect residual disease after neoadjuvant therapy in breast cancer. Sci Transl Med 2019; 11 (504) eaax7392
  • 8 Shen L. Liquid biopsy: a powerful tool to monitor trastuzumab resistance in HER2-positive metastatic gastric cancer. Cancer Commun (Lond) 2018; 38 (01) 72
  • 9 Phallen J, Sausen M, Adleff V. , et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med 2017; 9 (403) eaan2415
  • 10 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 (346) 346ra92
  • 11 Rago C, Huso DL, Diehl F. , et al. Serial assessment of human tumor burdens in mice by the analysis of circulating DNA. Cancer Res 2007; 67 (19) 9364-9370
  • 12 Xie M, Lu C, Wang J. , et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat Med 2014; 20 (12) 1472-1478
  • 13 Hu Y, Ulrich BC, Supplee J. , et al. False-positive plasma genotyping due to clonal hematopoiesis. Clin Cancer Res 2018; 24 (18) 4437-4443
  • 14 Leal A, van Grieken NCT, Palsgrove DN. , et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat Commun 2020; 11 (01) 525
  • 15 Cats A, Jansen EPM, van Grieken NCT. , et al; CRITICS investigators. Chemotherapy versus chemoradiotherapy after surgery and preoperative chemotherapy for resectable gastric cancer (CRITICS): an international, open-label, randomised phase 3 trial. Lancet Oncol 2018; 19 (05) 616-628
  • 16 Langer R, Becker K. Tumor regression grading of gastrointestinal cancers after neoadjuvant therapy. Virchows Arch 2018; 472 (02) 175-186