Next Article in Journal
Cell-Free DNA Analysis by Whole-Exome Sequencing for Hepatocellular Carcinoma: A Pilot Study in Thailand
Next Article in Special Issue
Validation of the Combined Biomarker for Prediction of Response to Checkpoint Inhibitor in Patients with Advanced Cancer
Previous Article in Journal
3T MRI-Radiomic Approach to Predict for Lymph Node Status in Breast Cancer Patients
Previous Article in Special Issue
Characterization of Cell-Bound CA125 on Immune Cell Subtypes of Ovarian Cancer Patients Using a Novel Imaging Platform
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Clinical Implication of Liquid Biopsy in Colorectal Cancer Patients Treated with Metastasectomy

1
Division of Medical Oncology and Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Korea
2
Korea University Cancer Research Institute, Korea University College of Medicine, Seoul 02841, Korea
3
Department of Surgery, Korea University College of Medicine, Seoul 02841, Korea
4
Department of Thoracic and Cardiovascular Surgery, Korea University College of Medicine, Seoul 02841, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally as co-first authors.
Current affiliation: Department of Surgery, Seoul National University Boramae Medical Center, Seoul National University College of Medicine Seoul, Seoul 03080, Korea.
Submission received: 28 March 2021 / Revised: 26 April 2021 / Accepted: 4 May 2021 / Published: 6 May 2021
(This article belongs to the Collection Cancer Biomarkers)

Abstract

:

Simple Summary

Circulating tumor DNA (ctDNA) is tumor-derived fragmented DNA in the bloodstream that was shed from primary and/or metastatic tumors. ctDNA in patients with oligometastatic colorectal cancer (CRC) was detected before and after metastasectomy depending on the clinical context. The detection rate of ctDNA was higher in liver metastasis than lung metastasis and tumors measuring ≥1 cm and small tumors <1 cm. After metastasectomy for oligometastatic lesions with good response to neoadjuvant chemotherapy, most ctDNA was cleared or existed below the detection level. Biological characteristics affecting tumor DNA release should be considered when applying ctDNA assays in clinical settings.

Abstract

Background & Aims: The application of circulating tumor DNA (ctDNA) has been studied for predicting recurrent disease after surgery and treatment response during systemic treatment. Metastasectomy can be curative for well-selected patients with metastatic colorectal cancer (mCRC). This prospective study investigated the ctDNA level before and after metastasectomy in patients with mCRC to explore its potential as a predictive biomarker. Methods: We collected data on 98 metastasectomies for mCRC performed from March 2017 to February 2020. Somatic mutations in the primary and metastatic tumors were identified and tumor-informed ctDNAs were selected by ultra-deep targeted sequencing. Plasma samples were mandatorily collected before and 3–4 weeks after metastasectomy and serially, if patients agreed. Results: Data on 67 of 98 metastasectomies (58 patients) meeting the criteria were collected. ctDNA was detected in 9 (29%) of 31 cases treated with upfront metastasectomy and in 7 (19.4%) of 36 cases treated with metastasectomy after upfront chemotherapy. The detection rate of ctDNA was higher in liver metastasis (p = 0.0045) and tumors measuring ≥1 cm (p = 0.0183). ctDNA was less likely to be detected if the response to chemotherapy was good. After metastasectomy, ctDNA was found in 4 (6%) cases with rapid progressive disease. Conclusion: The biological factors affecting the ctDNA shedding from the tumor should be considered when applying ctDNA assays in a clinical setting. After metastasectomy for oligometastatic lesions in good responders of chemotherapy, most ctDNA was cleared or existed below the detection level. To assist clinical decision making after metastasectomy for mCRC using ctDNA, further studies for improving specific outcomes are needed.

1. Introduction

Colorectal cancer (CRC) is the third most common cancer in the world and Korea [1]. Approximately 25% patients with CRC present with overt metastasis and 25–35% patients develop metastasis during the course of the disease [2]. The majority of these patients have a poor prognosis. However, some patients with metastatic CRC (mCRC) are diagnosed with isolated liver and lung metastases. Surgical resection is the treatment of choice, with a survival rate of 25–50% [3]. Metastasectomy can be performed if the liver or lung volume after resection is sufficient to maintain organ function while removing all macroscopic disease with microscopically negative margins and preserving adequate vascular inflow and outflow [4]. Patients with widely metastatic disease may be suitable candidates for local treatment depending on the systemic chemotherapy response. mCRC treatment strategies aim at converting irresectable disease into resectable disease. The 5-year survival after conversion ranges from 35% to 50% [5,6], which is similar to that for patients who underwent resection at presentation.
Although the survival of patients with resectable stage IV cancer has significantly improved with effective chemotherapy regimens and advances in surgical techniques, 75% patients who undergo metastasectomy developed recurrence within 18 months of surgery [7]. Moreover, there is no consensus on standard treatment guidelines regarding the role of postoperative chemotherapy, treatment period, and additional intervention for preventing recurrence after metastasectomy. In addition, difficulty in detecting tiny lesions during postmetastasectomy follow-up, lack of reliable longitudinal surveillance methods, and multiple genetic changes because of clonal evolution and corresponding intratumoral heterogeneity are limitations for customizing treatment strategies to mCRC.
Liquid biopsy has emerged as a novel method for tumor mutation profiling. Various tumor-derived products such as circulating tumor cells, circulating cell-free DNA, and circulating tumor DNA (ctDNA) can be detected in the blood, which has been increasingly used in clinical practice [8]. The DNA fragments from tumor cells are released into the circulation through apoptosis, necrosis, and secretion. Furthermore, tumor-specific genetic alterations such as driver mutations, chromosome copy number alterations, and methylation can be detected in ctDNA, which can be of high value for cancer detection, prognostication, and treatment monitoring [9]. Several studies have confirmed the predictive value of ctDNA levels in mCRC and the prognostic significance of postoperative ctDNA levels in early and locally advanced CRC [10,11,12,13,14]. The presence of ctDNA after radical surgery correlates with recurrence, and elevated ctDNA levels in R0 and R1 resections may signal the occurrence of micrometastases [15,16,17].
In this study, we analyzed tumor-specific DNA mutations found in primary and metastatic tumor tissues and investigated the presence of tumor-guided ctDNA in patients with oligometastatic CRC before and after metastasectomy in a clinical setting and explored its practical usefulness.

2. Patients and Methods

2.1. Study Design and Participants

We prospectively enrolled patients with mCRC who underwent surgical resection of the primary tumor and metastasectomy at Korea University Anam Hospital between March 2017 and February 2020 with the objective of assessing premetastasectomy and postmetastasectomy ctDNA levels and clinical outcomes. Patients aged >19 years with stage Ⅳ CRC who underwent metastasectomy with definitive intent and with or without systemic chemotherapy were included. The decision regarding on chemotherapy timing and regimen was made by the treating physicians based on the current treatment guidelines. The treating physicians were blinded to the ctDNA results. All participants’ samples were collected with informed consent. This study was approved by the Institutional Review Board of Korea University Anam Hospital (2017AN0070).

2.2. Tumor Tissue and Blood Collection and Mutation Analysis by Next-Generation Sequencing

All primary and metastatic tumor samples were obtained from formalin-fixed paraffin-embedded surgical specimens based on a cutoff value of 80% for tumor sample purity. The resected primary and metastatic tumor specimens were evaluated by next-generation sequencing (average depth × 2328; on-target rate: 96.61%; and average uniformity: 97.69%) using the Ion AmpliSeq Cancer Hotspot Panel (ICP) v2 with the Ion Torrent Proton system. The ICP v2 covers 2800 COSMIC mutations in 50 cancer-related genes, which are presented in Table S1. The mutational status of tumor tissues was determined as previously described [18].
Blood was drawn from patients with mCRC before and 3–4 weeks after metastasectomy. Serial blood sampling after metastasectomy was conducted if the patients agreed. At each time point, 10 mL of blood was collected in EDTA tubes and processed for 1 h in the circulating cancer biomarker laboratory in the study institution. Ultra-deep targeted sequencing of ctDNA (average depth × 25,804; on-target rate: 90.18%; and average uniformity: 97.89%) was performed using the ICP v2 for paired plasma samples, as reported previously [19].

2.3. Tumor-Guided ctDNA Identification

We developed a personalized approach for tumor-guided ctDNA detection and quantification called targeted sequencing. We selected pathogenic somatic variants in primary and metastatic CRC tumor samples and then matched plasma sample with same pathogenic variants from tumor cells. Pathogenic variants were annotated as “likely pathogenic” or “pathogenic” in the COSMIC, ClinVar, and OncoKB databases. Each alteration identified de novo was reviewed manually to exclude false positives. The cutoffs of variant allele frequency (VAF) for tissue sequencing and ctDNA sequencing were 5% and 1%, respectively, if the mutation was identified in the tumor specimen. Patients with a VAF below these thresholds were considered negative, whereas those with missing VAF data for a mutation were excluded from further analysis. We selected tumor-ctDNA matches and comprehensive filtering of mutations through clustering maximized the likelihood that the reported alterations originated from tumors and not from other sources such as clonal hematopoiesis of indeterminate potential [20]. The germline mutations were not analyzed in this study and were filtered when plasma variants uniformly represented in a VAF of 50–100%.

2.4. Statistical Analysis

Descriptive statistics for categorical variables are presented as frequencies and those for continuous variables are presented as means. Categorical variables are presented using contingency tables. They were compared using the chi-square test or Fisher’s exact test. The ctDNA level was classified as detectable (ctDNA positive, VAF: ≥1%) or undetectable (ctDNA negative, VAF: <1%). The relationships between clinicopathological variables and ctDNA detection and VAF levels were assessed using Fisher’s exact test and the Wilcoxon rank-sum tests. Progression-free survival (PFS) was measured from the time of metastasectomy to recurrence or death from disease. Patients alive at the last follow-up and with no evidence of disease were censored. Overall survival (OS) was measured from the time of metastasectomy to death from the disease. p < 0.05 was considered statistically significant. All statistical analyses were performed using SPSS ver. 25.0 (IBM Corp., Armonk, NY, USA).

3. Results

3.1. Patient Characteristics

This study enrolled consecutive patients with mCRC who underwent metastasectomy with or without chemotherapy. Table 1 summarizes the preoperative features of the 58 evaluable patients. The mean age of the patients was 56 years, and two-thirds of the enrolled patients were male (n = 37, 63.8%). The distribution of the primary tumor sites was as follows: the ascending colon (n = 8, 13.8%), descending colon (n = 20, 34.5%), and rectum (n = 30, 51.7%). In all patients, except one, the primary lesion was surgically removed. At the time of diagnosis, metachronous metastasis occurred in one-third of patients and synchronous metastasis occurred in two-thirds of patients. Metastasectomy was most commonly performed for liver metastasis (66%), followed by lung (43%), peritoneal (7%), and lymph node (3%) metastases.

3.2. ctDNA Detection before and after Metastasectomy

Data on 98 metastasectomies were collected; however, data on 31 of 98 metastasectomies (3 cases showing no cancer cells after metastasectomy, 10 cases that failed quality control during the sample preparation process, and 18 cases lacking pathogenic variants in the metastatic tumor tissue; Figure 1) were excluded. The clinical data on every metastasectomy and pathogenic variants in ctDNA are presented in Table S2. Overall, ctDNA was detected in 16 of 67 (24%) cases before metastasectomy and in 4 of 67 (6%) cases with rapid progressive disease immediately after metastasectomy.
In the clinical context, metastasectomy of mCRC can be performed in 2 situations. First, if the metastatic lesion is isolated and small, upfront metastasectomy without systemic chemotherapy can be performed (groups 1 and 2). Second, if the metastatic disease is extensive and not resectable, systemic chemotherapy is preferred and, thereafter, the decision to perform metastasectomy can be reassessed depending on the chemotherapy response (groups 3 and 4).

3.3. Group 1: Upfront Metastasectomy without Neoadjuvant Chemotherapy (R0 Resection = 28 Cases)

Patients in this group underwent metastasectomy without neoadjuvant chemotherapy (31 of 67 cases). R0 resection failed in 3 cases. The majority of 28 successful R0 resection cases were found to be oligometastatic and metachronous (26 cases). ctDNA was detected in 8 (28.5%) of 28 R0 resection cases before metastasectomy, but it was not detected in any case after metastasectomy (Table 2). All 8 cases positive for ctDNA were cases of liver metastasis and the tumor size was >1 cm. The VAF in primary tumor was often not detected (case number #2, #34, #68) when the primary tumor was exposed to chemotherapy or concurrent chemoradiotherapy in metachronous mCRC (26 cases). The detection of ctDNA before metastasectomy was not related to PFS and OS.

3.4. Group 2: Upfront Metastasectomy without Neoadjuvant Chemotherapy (R0 Resection Failed = 3 Cases)

Patients in this group underwent metastasectomy without neoadjuvant chemotherapy. ctDNA was detected in 1 (33.3%) of the 3 R0 resection cases. One ctDNA positive case had liver metastasis with a 2.1 cm sized tumor. Although residual lesions in the lymph node, peritoneum, and lung (small sized multiple nodules) were found on postoperative imaging, ctDNA was not detected immediately after metastasectomy (Table 3).
We compared the median VAFs in the tumor tissue and ctDNA between groups 1 and 2. The amount of ctDNA released from the tumor can be considered to reflect the tumor burden if the size of metastatic lesions is large and chemotherapy is not performed. The median VAF in the tumor was 28.7% and that in ctDNA was 1.1%, meaning that the detection rate of ctDNA was less than one-tenth of that of tumor.

3.5. Group 3: Neoadjuvant Chemotherapy Followed by Metastasectomy (R0 Resection = 25 Cases)

This group of patients received upfront chemotherapy because curative surgery was not feasible at the time of mCRC diagnosis due to metastatic tumor burden and extent. Most cases (22 of 25 cases) had synchronous metastasis. More than 3 cycles of chemotherapy were administered to treat all cases. The responses to chemotherapy were as follows: complete response, 1 case; partial responses, 13 cases; stable disease, 6 cases; and progressive disease, 5 cases (resistance was acquired after an initial good response). All cases achieved complete resection.
After neoadjuvant chemotherapy, ctDNA was detected in 4 (16%) of 25 R0 resection cases, of which 3 cases were cases of liver metastasis and one case was a case of lung metastasis. Three of 4 cases acquired resistance and the disease progressed before metastasectomy. A good response to chemotherapy was observed and ctDNA was not detected in all, except 2 (#11 and #51b), of the remaining 21 cases. After metastasectomy, no ctDNA-positive case was found (Table 4).

3.6. Group 4: Neoadjuvant Chemotherapy Followed by Metastasectomy (R0 Resection Failed = 11)

This group of patients received chemotherapy, followed by metastasectomy; however, R0 resection failed. Nine of 11 cases had synchronous metastasis. Most cases (8 of 11 cases) showed poor response to chemotherapy. Three (27.3%) cases were found to be ctDNA positive before liver metastasectomy or peritonectomy, with tumors measuring >1 cm and disease progression at the time of ctDNA sampling. After metastasectomy, ctDNA was detected in 4 (36.4%) cases with rapid progressive disease in the liver or in the liver and bone (Table 5).

3.7. Longitudinal Tracking with Serial ctDNA Analysis

We conducted longitudinal ctDNA tracking in 14 cases. During serial sampling, the VAF in ctDNA increased before metastasis was radiologically confirmed in the case of recurrence (case number 6, 8, 20, 25, 26, 37, 40, 51, 53, 61, 65, 79, 92; all cases are listed in Supplementary Table S1); however, ctDNA was not detected after local interventions such as repeated surgery or radiofrequency ablation for metastatic lesions (case number 6, 20, 25, 37, 40, 51, 79, 92).
Moreover, although gross lesions are present, ctDNA might not be detected if the effective chemotherapy and radiotherapy are continued. In case number 26, ctDNA was not detected during maintenance chemotherapy for lung metastasis, but when a new brain metastasis occurred, ctDNA was detected. An increase in the VAF in ctDNA during treatment is highly associated with tumor growth and is detectable postmetastasectomy.
In case number 19, advanced gastric cancer occurred after 4.5 years of the diagnosis of mCRC with a novel pathogenic variant (TP53) from the gastric cancer tissue, which was not found in colon cancer tissue. During follow-up after gastrectomy, this pathogenic variant was repeatedly detected in the blood, confirming the recurrence of gastric cancer in the supraclavicular lymph nodes.
The detection rate of ctDNA differed based on the size and location of the metastatic lesion. Among R0 resection cases, the detection of ctDNA was less in small tumors (<1 cm) and lung metastasis and was high in large tumors (≥1 cm) and liver metastasis (Table 6; Figure 2). As demonstrated in this study, biological factors such as metastatic milieu shedding ctDNA other than tumor burden should be considered. In case of concurrent chemoradiotherapy or neoadjuvant chemotherapy for primary tumor, the VAF in the tumor was extremely low or under the detection level. This could be related to the lack of ctDNA detection in group 1 (case number 2, 7, 34, 40, 58, 60, 68, 72). Although not statistically presented, ctDNA was detected in several cases before several months of bone metastasis (case number 6) or confirmation of disease progression with radiological imaging (case number 40, 53).

4. Discussion

The optimal treatment strategies for mCRC differ based on the metastatic patterns. In patients with resectable and synchronous metastasis, surgical resection can offer a clear and significant long-term survival benefit. However, in patients with synchronous, extensive, large-volume, and disseminated disease, systemic chemotherapy is preferred and metastasectomy may be considered depending on the response to chemotherapy. No definite biomarker is available whether chemotherapy should be performed or the effectiveness of therapy regimen after successful R0 resection.
Numerous reports have demonstrated that the baseline ctDNA level is a prognostic factor in a wide range of patients with metastatic cancers undergoing chemotherapy and sensitive assessment for early response monitoring [21,22,23]. ctDNA levels are associated with tumor burden. However, other factors influencing ctDNA levels are poorly characterized. Additionally, limited studies have reported data specifically on the use of ctDNA in patients with mCRC who have undergone metastasectomy.
To determine whether the presence of ctDNA before and after metastasectomy can be a prognostic or predictive biomarker that aids in determining the treatment direction, we used a personalized approach for tumor-guided ctDNA detection. Nearly one-third of the samples were excluded from the analysis because of low yield. Among the 98 metastasectomies, 10 failed quality control and 18 did not have clinically significant pathogenic variants. ctDNA was detected in 16 (24%) of 67 events before metastasectomy; 9 (29%) of 31 cases treated with upfront metastasectomy without chemotherapy (groups 1 and 2) and 7 (19.4%) of 36 cases treated with neoadjuvant chemotherapy followed by metastasectomy (groups 3 and 4). After metastasectomy, ctDNA was detected in 4 (6%) of 67 events, all of which were found in group 4, and these were patients with gross lesions after metastasectomy. Compare with postoperative ctDNA analysis in the stage l to lll CRC, ctDNA-positive events were 10(8%) out of 125 patients at postoperative day 30 [24].
Further, we evaluated whether administration of chemotherapy was not statistically significantly related to ctDNA positivity before metastasectomy; the ctDNA detection rate tended to be low in cases of good response to chemotherapy. The detection of ctDNA rate was higher in liver metastasis and metastasized tumors measuring ≥1 cm than in lung metastasis and metastasized tumors measuring <1 cm. ctDNA was detected in 4 cases immediately after metastasectomy. All the 4 cases belonged to group 4, which underwent metastasectomy (failed R0 resection) with neoadjuvant chemotherapy.
In our study, the ctDNA detection rate was lower than that reported in a previous study on mCRC [9]. Considering the ctDNA dynamics corresponding to the clinical condition of patients with mCRC [25,26], most patients’ tumor burden was low and chemotherapy response was the best at the time of ctDNA blood sampling. The mean diameter of the resected tumor was 2.7 cm in ctDNA-positive cases and 1.4 cm in ctDNA-negative cases. Primary tumor burdens of 1, 10, and 100 cm3 result in mean clonal plasma VAFs of 0.008%, 0.1%, and 1.4%, respectively [27]. In a previous study, considering the ctDNA shedding rate per cancer cell, the probabilities of a false-negative result for a particular actionable mutation clonally present in tumors with diameters of 1, 1.5, and 2 cm were 82%, 44%, and 9.3%, respectively [28]. In the case of patients with small, isolated metastatic tumors who did not receive chemotherapy (groups 1 and 2), the median VAFs in the tumor and plasma ctDNA were 28.7% and 1.1%, respectively, which the detection rate in plasma ctDNA can be less than one-tenth of that of tumor DNA. The difference of the above frequencies could be partially explained by the vulnerability of ctDNA. According to the studies on early dynamics of ctDNA, changes in ctDNA mutation frequencies were generally observed during the first 2 cycles of chemotherapy [29,30]. This is the reason that the neoadjuvant chemotherapy groups (groups 3 and 4) showed a lower ctDNA positive rate than the upfront metastasectomy groups (groups 1 and 2). ctDNA is typically undetectable in mid-treatment samples even in patients with metastatic cancer with measurable disease on imaging [31].
To the best of our knowledge, this is the first prospective study to investigate ctDNA levels before and after metastasectomy in patients with potentially resectable mCRC. We aimed to detect ctDNA before and after metastasectomy with the same detection method and identify the clinicopathological determinants of ctDNA detection. The released ctDNA may be influenced by various mechanisms such as the metastatic tumor site, invasion through blood vessels, administration of effective local and systemic chemotherapy, and tumor burden.
This study has several limitations. The relatively small sample size of this exploratory study impairs the generalization of our conclusion. Further, we did not collect serial ctDNA samples from all patients. Therefore, the interpretation of serial ctDNA tracking is limited. There is also the potential that our ctDNA sequencing assay was not sensitive enough to detect the true landscape of plasma mutations and molecular alterations. Finally, our study was not powered to examine the association between ctDNA change and clinical outcomes such as PFS and OS. Further studies should include the systemic sample collection, a larger sample to allow clinical factors associated with prognosis to be controlled, such as receiving neoadjuvant chemotherapy before metastasectomy, components of the clinical risk factors, and R0 resection status.

5. Conclusions

After metastasectomy for oligometastatic lesions and a good response to chemotherapy, most of the ctDNA was cleared and existed below the detection level. The biological characteristics affecting the release of tumor DNA should be considered when applying ctDNA assays in a clinical setting. Unexpectedly low fractions of ctDNA were found in a substantial proportion of patients with metastatic disease [32]. To expand the clinical application of ctDNA as a biomarker, it would be more cost-effective to follow up specific tumor-derived ctDNA with a highly sensitive tool such as droplet digital PCR rather than whole exome or targeted sequencing in the context of clinical setting.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/cancers13092231/s1, Table S1: Ion AmpliSeq™ Comprehensive Cancer Panel target gene list (50 genes). Table S2: Clinical characteristics of all participants and their genomic information with tumor and plasma.

Author Contributions

Conceptualization, Y.-S.P., W.-J.C. and Y.H.K.; methodology, S.L., Y.-S.P., W.-J.C. and Y.H.K.; formal analysis, S.L., Y.-S.P., A.L., B.K., S.-B.L., J.-W.L. and Y.H.K.; investigation, S.L., W.-J.C., J.Y.C., Y.H.K., S.-H.K., J.K., J.-M.K., K.-C.Y., and S.-H.L., resources, Y.H.K.; writing—original draft preparation, S.L., Y.-S.P.; writing—review and editing, Y.H.K.; funding acquisition, Y.H.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (NRF-2017R1A-2B3004624) and Korea University research grant.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of Korea University Anam Hospital (2017AN0070).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to a privacy issue from the patients.

Acknowledgments

We thank the patients who participated in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hong, S.; Won, Y.J.; Park, Y.R.; Jung, K.W.; Kong, H.J.; Lee, E.S. The Community of Population-Based Regional Cancer Registries. Cancer Statistics in Korea: Incidence, Mortality, Survival, and Prevalence in 2017. Cancer Res. Treat. 2020, 52, 335–350. [Google Scholar] [CrossRef]
  2. Van Cutsem, E.; Nordlinger, B.; Adam, R.; Köhne, C.-H.; Pozzo, C.; Poston, G.; Ychou, M.; Rougier, P. Towards a pan-European consensus on the treatment of patients with colorectal liver metastases. Eur. J. Cancer 2006, 42, 2212–2221. [Google Scholar] [CrossRef] [PubMed]
  3. Biasco, G.; Derenzini, E.; Grazi, G.; Ercolani, G.; Ravaioli, M.; Pantaleo, M.; Brandi, G. Treatment of hepatic metastases from colorectal cancer: Many doubts, some certainties. Cancer Treat. Rev. 2006, 32, 214–228. [Google Scholar] [CrossRef]
  4. Garden, O.J.; Rees, M.; Poston, G.J.; Mirza, D.; Saunders, M.; Ledermann, J.; Primrose, J.N.; Parks, R.W. Guidelines for resection of colorectal cancer liver metastases. Gut 2006, 55 (Suppl. 3), iii1–iii8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lee, W.-S.; Yun, S.H.; Chun, H.-K.; Lee, W.-Y.; Yun, H.-R.; Kim, J.; Kim, K.; Shim, Y.M. Pulmonary resection for metastases from colorectal cancer: Prognostic factors and survival. Int. J. Colorectal Dis. 2007, 22, 699–704. [Google Scholar] [CrossRef]
  6. Kemeny, N.E.; Chou, J.F.; Boucher, T.M.; Capanu, M.; DeMatteo, R.P.; Jarnagin, W.R.; Allen, P.J.; Fong, Y.C.; Cercek, A.; D’Angelica, M.I. Updated long-term survival for patients with metastatic colorectal cancer treated with liver resection followed by hepatic arterial infusion and systemic chemotherapy. J. Surg. Oncol. 2016, 113, 477–484. [Google Scholar] [CrossRef] [Green Version]
  7. Penna, C.; Nordlinger, B. Colorectal metastasis (liver and lung). Surg. Clin. N. Am. 2002, 82, 1075–1090. [Google Scholar] [CrossRef]
  8. Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L. 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. [Google Scholar] [CrossRef] [PubMed]
  9. Bach, S.; Sluiter, N.R.; Beagan, J.J.; Mekke, J.M.; Ket, J.C.; van Grieken, N.C.; Steenbergen, R.D.; Ylstra, B.; Kazemier, G.; Tuynman, J.B. Circulating tumor DNA analysis: Clinical implications for colorectal cancer patients. A systematic review. JNCI Cancer Spectr. 2019, 3, pkz042. [Google Scholar] [CrossRef]
  10. Tie, J.; Kinde, I.; Wang, Y.; Wong, H.-L.; Roebert, J.; Christie, M.; Tacey, M.; Wong, R.; Singh, M.; Karapetis, C. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann. Oncol. 2015, 26, 1715–1722. [Google Scholar] [CrossRef]
  11. Wang, Y.; Li, L.; Cohen, J.D.; Kinde, I.; Ptak, J.; Popoli, M.; Schaefer, J.; Silliman, N.; Dobbyn, L.; Tie, J.; et al. Prognostic Potential of Circulating Tumor DNA Measurement in Postoperative Surveillance of Nonmetastatic Colorectal Cancer. JAMA Oncol. 2019, 5, 1118–1123. [Google Scholar] [CrossRef] [PubMed]
  12. Tie, J.; Cohen, J.D.; Wang, Y.; Li, L.; Christie, M.; Simons, K.; Elsaleh, H.; Kosmider, S.; Wong, R.; Yip, D. Serial circulating tumour DNA analysis during multimodality treatment of locally advanced rectal cancer: A prospective biomarker study. Gut 2019, 68, 663–671. [Google Scholar] [CrossRef] [PubMed]
  13. Tie, J.; Cohen, J.D.; Wang, Y.; Christie, M.; Simons, K.; Lee, M.; Wong, R.; Kosmider, S.; Ananda, S.; McKendrick, J.; 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. [Google Scholar] [CrossRef] [PubMed]
  14. Tie, J.; Wang, Y.; Tomasetti, C.; Li, L.; Springer, S.; Kinde, I.; Silliman, N.; Tacey, M.; Wong, H.-L.; Christie, M. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci. Transl. Med. 2016, 8, 346ra392. [Google Scholar] [CrossRef] [Green Version]
  15. Carrato, A. Adjuvant treatment of colorectal cancer. Gastrointest. Cancer Res. 2008, 2, S42–S46. [Google Scholar]
  16. Grávalos, C.; García-Escobar, I.; García-Alfonso, P.; Cassinello, J.; Malón, D.; Carrato, A. Adjuvant chemotherapy for stages II, III and IV of colon cancer. Clin. Transl. Oncol. 2009, 11, 526–533. [Google Scholar] [CrossRef]
  17. Schøler, L.V.; Reinert, T.; Ørntoft, M.-B.W.; Kassentoft, C.G.; Árnadóttir, S.S.; Vang, S.; Nordentoft, I.; Knudsen, M.; Lamy, P.; Andreasen, D. Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin. Cancer Res. 2017, 23, 5437–5445. [Google Scholar] [CrossRef] [Green Version]
  18. Tie, J.; Cohen, J.D.; Lo, S.N.; Wang, Y.; Li, L.; Christie, M.; Lee, M.; Wong, R.; Kosmider, S.; Skinner, I.; et al. Prognostic significance of postsurgery circulating tumor DNA in nonmetastatic colorectal cancer: Individual patient pooled analysis of three cohort studies. Int. J. Cancer 2020. [Google Scholar] [CrossRef]
  19. Sung, J.S.; Chong, H.Y.; Kwon, N.J.; Kim, H.M.; Lee, J.W.; Kim, B.; Lee, S.B.; Park, C.W.; Choi, J.Y.; Chang, W.J.; et al. Detection of somatic variants and EGFR mutations in cell-free DNA from non-small cell lung cancer patients by ultra-deep sequencing using the ion ampliseq cancer hotspot panel and droplet digital polymerase chain reaction. Oncotarget 2017, 8, 106901–106912. [Google Scholar] [CrossRef] [Green Version]
  20. Genovese, G.; Kahler, A.K.; Handsaker, R.E.; Lindberg, J.; Rose, S.A.; Bakhoum, S.F.; Chambert, K.; Mick, E.; Neale, B.M.; Fromer, M.; et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 2014, 371, 2477–2487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Dawson, S.J.; Tsui, D.W.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef] [Green Version]
  22. Pecuchet, N.; Zonta, E.; Didelot, A.; Combe, P.; Thibault, C.; Gibault, L.; Lours, C.; Rozenholc, Y.; Taly, V.; Laurent-Puig, P.; et al. Base-Position Error Rate Analysis of Next-Generation Sequencing Applied to Circulating Tumor DNA in Non-Small Cell Lung Cancer: A Prospective Study. PLoS Med. 2016, 13, e1002199. [Google Scholar] [CrossRef] [PubMed]
  23. Santiago-Walker, A.; Gagnon, R.; Mazumdar, J.; Casey, M.; Long, G.V.; Schadendorf, D.; Flaherty, K.; Kefford, R.; Hauschild, A.; Hwu, P.; et al. Correlation of BRAF Mutation Status in Circulating-Free DNA and Tumor and Association with Clinical Outcome across Four BRAFi and MEKi Clinical Trials. Clin. Cancer Res. 2016, 22, 567–574. [Google Scholar] [CrossRef] [Green Version]
  24. Reinert, T.; Henriksen, T.V.; Christensen, E.; Sharma, S.; Salari, R.; Sethi, H.; Knudsen, M.; Nordentoft, I.; Wu, H.T.; Tin, A.S.; et al. Analysis of Plasma Cell-Free DNA by Ultradeep Sequencing in Patients With Stages I to III Colorectal Cancer. JAMA Oncol. 2019. [Google Scholar] [CrossRef] [Green Version]
  25. Levy, M.; Benesova, L.; Lipska, L.; Belsanova, B.; Minarikova, P.; Veprekova, G.; Zavoral, M.; Minarik, M. Utility of cell-free tumour DNA for post-surgical follow-up of colorectal cancer patients. Anticancer Res. 2012, 32, 1621–1626. [Google Scholar]
  26. Flamini, E.; Mercatali, L.; Nanni, O.; Calistri, D.; Nunziatini, R.; Zoli, W.; Rosetti, P.; Gardini, N.; Lattuneddu, A.; Verdecchia, G.M.; et al. Free DNA and carcinoembryonic antigen serum levels: An important combination for diagnosis of colorectal cancer. Clin. Cancer Res. 2006, 12, 6985–6988. [Google Scholar] [CrossRef] [Green Version]
  27. Abbosh, C.; Birkbak, N.J.; Wilson, G.A.; Jamal-Hanjani, M.; Constantin, T.; Salari, R.; Le Quesne, J.; Moore, D.A.; Veeriah, S.; Rosenthal, R.; et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature 2017, 545, 446–451. [Google Scholar] [CrossRef] [PubMed]
  28. Avanzini, S.; Kurtz, D.M.; Chabon, J.J.; Moding, E.J.; Hori, S.S.; Gambhir, S.S.; Alizadeh, A.A.; Diehn, M.; Reiter, J.G. A mathematical model of ctDNA shedding predicts tumor detection size. Sci. Adv. 2020, 6. [Google Scholar] [CrossRef]
  29. Oikkonen, J.; Zhang, K.; Salminen, L.; Schulman, I.; Lavikka, K.; Andersson, N.; Ojanpera, E.; Hietanen, S.; Grenman, S.; Lehtonen, R.; et al. Prospective Longitudinal ctDNA Workflow Reveals Clinically Actionable Alterations in Ovarian Cancer. JCO Precis. Oncol. 2019, 3, 1–12. [Google Scholar] [CrossRef]
  30. Garlan, F.; Laurent-Puig, P.; Sefrioui, D.; Siauve, N.; Didelot, A.; Sarafan-Vasseur, N.; Michel, P.; Perkins, G.; Mulot, C.; Blons, H.; 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. [Google Scholar] [CrossRef] [Green Version]
  31. Katoh, M.; Unakami, M.; Hara, M.; Fukuchi, S. Bone metastasis from colorectal cancer in autopsy cases. J. Gastroenterol. 1995, 30, 615–618. [Google Scholar] [CrossRef] [PubMed]
  32. Bettegowda, C.; Sausen, M.; Leary, R.J.; Kinde, I.; Wang, Y.; Agrawal, N.; Bartlett, B.R.; Wang, H.; Luber, B.; Alani, R.M.; et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014, 6, 224ra224. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Workflow of the exploratory prospective study.
Figure 1. Workflow of the exploratory prospective study.
Cancers 13 02231 g001
Figure 2. VAF difference between tumor and plasma under various conditions. (a) Mean VAF difference between tumor and plasma; (b) VAF difference between tumor and plasma under metastatic organ; (c) VAF difference between tumor and plasma under neoaduvant chemotherapy response.
Figure 2. VAF difference between tumor and plasma under various conditions. (a) Mean VAF difference between tumor and plasma; (b) VAF difference between tumor and plasma under metastatic organ; (c) VAF difference between tumor and plasma under neoaduvant chemotherapy response.
Cancers 13 02231 g002
Table 1. Patient Characteristics.
Table 1. Patient Characteristics.
Clinical VariablesNpatient = 58 (%)
Age years (median, range)56 (35–78)
GenderMale37 (63.8)
Female21 (36.2)
Primary tumor siteAscending colon8 (13.8)
Descending colon20 (34.5)
Rectum30 (51.7)
Primary tumor surgeryYes57 (98.3%)
No1 (1.7%)
Synchronicity of metastasisMetachronous18 (31.1)
Synchronous40 (68.9)
Metastasectomy organLiver38 (65.5)
Lung25 (43.1)
Peritoneum4 (6.9)
Lymph node2 (3.4)
Table 2. Metastasectomy (R0 resection) Without Chemotherapy (Nevent = 28).
Table 2. Metastasectomy (R0 resection) Without Chemotherapy (Nevent = 28).
NoSynchronicityVariantPrimary Tissue
VAF
Metastatic
Tissue
VAF
Metastatic SitePreM
ctDNA
VAF
PostM
ctDNA
VAF
Recurence
No CTxCTx
ctDNA positive before metastasectomy
020bMTP53 R196 * 38.565.1Liver
2.5 cm
1.20.0N
APC Q1294 * 25.451.60.00.0
037bMTP53 T211I14.9 59.1Liver
2.5 cm
46.80.0N
073MAPC T1445Qfs21.0 26.0Liver
7.4 cm
0.00.0N
APC R876 *31.2 0.00.00.0
TP53 R248W23.0 21.01.10.0
025aMFXW7 R465C33.0 33.4Liver
2.1 cm
0.00.0N
KRAS G12D23.8 28.40.00.0
TP53 R196 *35.9 51.82.10.0
APC L1488fs26.1 00.00.0
025bMFXW7 R465C33.0 31.0Liver
1.5 cm
1.10.0Liver
LN
KRAS G12D23.8 36.32.00.0
TP53 R196 *35.9 49.01.80.0
APC L1488fs26.1 00.00.0
050MAPC Q1367 *32.6 34.0Liver
3.0 cm
0.00.0Liver
TP53 R237H26.2 39.01.00.0
BRAF G469E0.0 1.30.00.0
CTNNB1 G34E0.0 1.40.00.0
VHL R161 *0.0 1.40.00.0
062MAPC R1450 *35.3 52.0Liver
3.2 cm
5.90.0Lung
KRAS G12S55.0 68.08.60.0
TP53 R342 *50.9 52.09.00.0
092MSMAD4 G419RQC failed * 53.6Liver
4.7 cm
11.00.0Lung
Liver
ctDNA negative before metastasectomy
002MNRAS G12D 0.019.0Lung
0.9 cm
0.00.0N
005MTP53 R282W43.0 22.0Lung
0.8 cm
0.00.0N
APC R876 *34.0 14.00.00.0
028bMKRAS G12V14.3 0.0PT
4.0 cm
0.00.0N
PIK3CA E545K14.8 0.00.00.0
SMAD4 R361H16.4 0.00.00.0
SMAD4 E330Q16.6 50.20.00.0
PIK3CA G914R14.9 00.00.0
034MTP53 R175H 0.017.2LN
0.9 cm
0.00.0N
KRAS G12C 0.018.00.00.0
PIK3CA E545V 0.03.30.00.0
041MKRAS A59T59.0 44.0Lung
1.0 cm
0.00.0N
TP53 R342 *44.0 22.00.00.0
056MKRAS G12D24.0 0.0Liver
0.7 cm
0.00.0N
TP53 C238Y35.7 0.00.00.0
060MTP53 N239 * 21.032.0Liver
2.0 cm
0.00.0N
066MTP53 R282W23.4 3.9Lung
0.4 cm
0.00.0N
APC R876 *31.2 0.00.00.0
APC E1379 *26.4 0.00.00.0
068MTP53 R213 * 0.025.1Lung
1.7 cm
0.00.0N
081SKRAS G12S30.0 29.0Lung
1.2 cm
0.00.0N
APC Q1378 *23.0 25.00.00.0
093MPIK3CA V344MBDL * 17.4Lung
1.4 cm
0.00.0N
098MKRAS G13D0.0 21.1Liver
2.2 cm
0.00.0N
APC L1488fs22.9 0.00.00.0
APC E1494fs0.0 20.40.00.0
007MAPC C1289 * 6.920.8Lung
1.4 cm
0.00.0PT
PIK3CA E542K 3.820.50.00.0
TP53 R282W 2.614.00.00.0
TP53 S99fs * 2.111.00.00.0
031aSAPC R1114 * 8.129.4Lung
1.4 cm
0.00.0Lung
KRAS G13C 13.442.60.00.0
APC R1463fs 2.00.00.00.0
APC E1345 * 8.130.70.00.0
032MKRAS G12D22.3 29.2Lung
1.2 cm
0.00.0LN
TP53 R282W0.0 34.60.00.0
TP53 S127F14.3 0.00.00.0
APC L1488fs13.2 0.00.00.0
035MKRAS G12C37.7 15.6Lung
0.5 cm
0.00.0Lung
TP53 I195T41.4 17.10.00.0
APC R1463fs1.9 0.00.00.0
APC S1501fs24.0 8.90.00.0
040MKRAS G12D 3.418.5Lung
0.6 cm
0.00.0Lung
Bone
TP53 S241P 1.210.40.00.0
PTEN R173H 1.00.00.00.0
058MNRAS G12D 24.632.7Lung
1.0 cm
0.00.0Lung
TP53 H297fs 26.214.60.00.0
TP53 R333fs 22.510.00.00.0
072MAPC R876 * 17.810.6PT
2.7 cm
0.00.0PT
LN
TP53 R248Q 16.810.80.00.0
APC Q1303 * 14.112.00.00.0
078MKRAS G12V41.0 12.9Lung
1.2 cm
0.00.0Lung
TP53 R175H54.1 20.80.00.0
Abbreviations: VAF, variant allele frequency; PreM, premetastasectomy; PostM, postmetastasectomy; CTx, chemotherapy; M, metachronous stage 4; S, synchronous stage 4; N, no recurrence; PT, peritoneum; LN, lymph node; BDL, below detection level. QC failed *: this patient received concurrent chemoradiation followed by surgery; *, indicate a translation termination (stop) codon.
Table 3. Metastasectomy without Chemotherapy and Residual Disease (Nevent = 3).
Table 3. Metastasectomy without Chemotherapy and Residual Disease (Nevent = 3).
NoSynchronicityVariantPrimary Tissue
VAF
Metastatic Tissue
VAF
Metastatic SitePreM
ctDNA
VAF
PostM
ctDNA
VAF
Residual
No CTxCTx
ctDNA positive before metastasectomy
046MKRAS G12D 038.1Liver
2.1 cm
4.40.0Lung
<0.5 #4
Pelvic LNs
TP53 G245C 1.046.35.60.0
APC K1561 * 050.55.00.0
ctDNA negative before metastasectomy
022MTP53 V272M39.4 55.4Liver
3.0 cm
0.00.0LN
2.2 cm
SMAD47.4 0.0
061cMTP53 51.525.1Lung
2.1 cm
0.00.0PT
APC H1490fs 30.216.20.00.0
Abbreviations: VAF, variant allele frequency; PreM, premetastasectomy; PostM, postmetastasectomy; CTx, chemotherapy; M, metachronous stage 4; PT, peritoneum; LN, lymph node; *, indicate a translation termination (stop) codon.
Table 4. Neoadjuvant Chemotherapy Followed by Metastasectomy (R0 resection) (Nevent = 25).
Table 4. Neoadjuvant Chemotherapy Followed by Metastasectomy (R0 resection) (Nevent = 25).
NoSynchronicityVariantPrimary Tissue
VAF
Metastatic Tissue
VAF
Meta
Site
PreM
ctDNA
PostM
ctDNA
Recurence
No CTx CTx RxCTx Rx
ctNDA positive before metastasectomy
001STP53 I255F 49.8SD0.0PRLiver
0.1 cm
1.60.0N
036SAPC R1114 *44.2 31.0PDLiver
2.5 cm
21.00.0N
APC R1450 *21.5 17.011.00.0
KRAS G12D46.5 34.019.00.0
008bSKRAS Q61R48.8 53.7PDLung
2.5 cm
9.60.0Lung
TP53 R175H49.5 54.16.40.0
028aSKRAS G12V14.3 7.8PDLiver 2.2 cm0.00.0PT
PIK3CA E545K14.8 11. 11.30.0
PIK3CA G914R14.9 10.20.00.0
SMAD4 E330Q16.6 8.90.00.0
SMAD4 R361H/C16.4 1.70.00.0
ctNDA negative before metastasectomy
004STP53 R273C60.4 4.1PRLiver
0.8 cm
0.00.0N
APC K1308 *40.2 2.80.00.0
012SKRAS G12V14.6 0.0CRLiver
0 cm
0.00.0N
PIK3CA H1047R20.6 0.00.00.0
031bSAPC R1114 *8.1 11.6SDLung
1.4 cm
0.00.0N
APC E1345 *8.1 15.40.00.0
KRAS G13C13.4 9.10.00.0
042MAPC Q1378 *47.5 36.2PRLiver
2.5 cm
0.00.0N
KRAS G12D29.9 18.30.00.0
TP53 R175H30.5 19.30.00.0
044SCTNNB1 S45F36.8 6.5PRLiver
2.2 cm
0.00.0N
KRAS G12D17.3 0.00.00.0
PTEN R335 *5.2 0.00.00.0
057STP53 C275Y26.5 36.9SDLung
1.6 cm
0.00.0N
091SAPC E1306 *18.0 0.0PRLiver
0.8 cm
0.00.0N
TP53 R175H40.2 6.20.00.0
APC Q886 *26.9 4.60.00.0
006SKRAS G13C37.2 10.7PRLiver
1.6 cm
0.00.0Bone
TP53 R306 *34.9 9.10.00.0
011MAPC R876 *30.6 0.0PDLung
1.0 cm
0.00.0Lung
KRAS E545K34.3 2.20.00.0
TP53 R342 *0.0 2.50.00.0
SMAD4 R361H0.0 3.80.00.0
SMAD4 A118V64.8 0.00.00.0
014SKRAS Q61H25.3 5.6SDLiver
2.3 cm
0.00.0PT
SMAD4 R361C15.2 3.00.00.0
019SPIK3CA G1049R 3.1SD3.6SDLiver
0.9 cm
0.00.0Liver
020aSTP53 R196 * 38.5SD21.2PRLiver
2.5 cm
0.00.0Liver
APC Q1294 * 25.413.80.00.0
027STP53 R175H 6.1PR2.6PRLiver
3.2 cm
0.00.0Liver
Lung
PTEN R335 * 1.10.00.00.0
037aSTP53 T211I14.9 1.0PRLiver
0.5 cm
0.00.0Liver
048SAPC R1450 * 1.5 0.0PRLung
0.6 cm
0.00.0Lung
CTNNB1 S37Y 0.0 21.20.00.0
KRAS G12D 1.4 0.00.00.0
PIK3CA E545K 2.0 0.00.00.0
TP53 R209Kfs 1.8 00.00.0
051bMKRAS G13S0.0 1.1PDLung
0.9 cm
0.00.0Lung
PTEN R130Q0.0 1.00.00.0
SMAD4 C115Y0.0 1.20.00.0
TP53 R175H10.7 40.00.00.0
052SRB1 A201fs 0.0 5.5PRLung
0.4 cm
0.00.0Lung
055SKRAS G13D60.3 16.6PRLiver
3.5 cm
0.00.0Liver
061bSTP53? 51.5SD27.1SDLung
0.5 cm
0.00.0Lung
APC H1490fs 30.219.50.00.0
063SKRAS G12VBDL 5.3PRLiver
2.2 cm
0.00.0Lung
KRAS A146TBDL 25.00.00.0
TP53 V173LBDL 2.40.00.0
APC P1381fsBDL 15.50.00.0
067STP53 M246R 23.2SD39.3SDRec 3.4 cm0.00.0PT
Abbreviations: VAF, variant allele frequency; PreM, premetastasectomy; PostM, postmetastasectomy; CTx, chemotherapy; Rx, chemotherapy response; CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease; M, metachronous stage 4; S, synchronous stage 4; N, no recurrence; PT, peritoneum; LN, lymph node; Rec, rectum; BDL, below detection level; *, indicate a translation termination (stop) codon.
Table 5. Neoadjuvant Chemotherapy Followed by Metastasectomy and Residual Disease (Nevent = 11).
Table 5. Neoadjuvant Chemotherapy Followed by Metastasectomy and Residual Disease (Nevent = 11).
NoSynchronicityVariantPrimary Tissue
VAF
Metastatic Tissue
VAF
Meta
Site
PreM
ctDNA
VAF
PostM
ctDNA
VAF
Residual
No CTx CTx RxCTxRx
ctDNA positive before metastasectomy
053SEBRR2 D769Y0 71.7PDLiver
#2 (2.3)
32.950Liver
multiple
Bone
076STP53 I195fs19.2 0PDPT
4.1 cm
20.416.9Huge liver
079MAPC Q1378 *BDL 23.2PDLiver
#7
21.60Lung
0.9 cm
TP53 S215GBDL 19.821.70
096SKRAS G13D31.4 8.6PRLiver
#9
00Liver #2 (<0.5)
TP53 V73fs50.8 000
ctDNA negative before metastasectomy
003STP53 R196 *30.2 22.8PDLiver
2.3 cm
00Liver
1.6 cm
008aSKRAS Q61R48.8 10.4SDLung
0.6 cm
0
0
0
0
Lung
0.8 cm
TP53 R175H49.5 10.1
026STP53 R306 *39.7 25.2PDLiver #5
PT #5
00Liver
0.5 cm
051aSTP53 R175H10.7 11.5PDLiver
#4
00Lung
0.5 cm
BRAF V471F0 12.300
054MKRAS G12D45.5 49.9PDLiver 1.6 cm
PT 2.6 cm
010.1Liver #6 (<1.0 cm)
061aSTP53? 51.5SD14.5PDLung
1.0 cm
00Lung
<0.5 cm #2
APC H1490fs 30.212.400
065SAPC R1114 *37.5 37.5PRLiver
#4
03.6Liver
1.4 cm
TP53 G244S52.7 46.2 02.2
APC S1356 *29.3 15.8 02.9
Abbreviations: VAF, variant allele frequency; PreM, premetastasectomy; PostM, postmetastasectomy; CTx, chemotherapy; Rx, chemotherapy response; PR, partial response; SD, stable disease; PD, progressive disease; M, metachronous stage 4; S, synchronous stage 4; N, no recurrence; PT, peritoneum; LN, lymph node; Rec, rectum; BDL, below detection level; *, indicate a translation termination (stop) codon.
Table 6. ctDNA Detection before Metastasectomy (Nevents = 67).
Table 6. ctDNA Detection before Metastasectomy (Nevents = 67).
Clinical ConditionctDNA Positive
(N = 16)
ctDNA Negative
(N = 51)
p Value
Neoadjuvant chemotherapy
before R0 resection
Yes7290.3587
No9 22
Metastasectomy organLiver14210.0045
Lung124
Other16
Tumor burden (tumor diameter)>1 cm15280.0183
≤1 cm123
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lee, S.; Park, Y.-S.; Chang, W.-J.; Choi, J.Y.; Lim, A.; Kim, B.; Lee, S.-B.; Lee, J.-W.; Kim, S.-H.; Kim, J.; et al. Clinical Implication of Liquid Biopsy in Colorectal Cancer Patients Treated with Metastasectomy. Cancers 2021, 13, 2231. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13092231

AMA Style

Lee S, Park Y-S, Chang W-J, Choi JY, Lim A, Kim B, Lee S-B, Lee J-W, Kim S-H, Kim J, et al. Clinical Implication of Liquid Biopsy in Colorectal Cancer Patients Treated with Metastasectomy. Cancers. 2021; 13(9):2231. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13092231

Chicago/Turabian Style

Lee, Soohyeon, Young-Soo Park, Won-Jin Chang, Jung Yoon Choi, Ahreum Lim, Boyeon Kim, Saet-Byeol Lee, Jong-Won Lee, Seon-Hahn Kim, Jin Kim, and et al. 2021. "Clinical Implication of Liquid Biopsy in Colorectal Cancer Patients Treated with Metastasectomy" Cancers 13, no. 9: 2231. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers13092231

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop