Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Review

Clinical application of circulating tumour DNA in colorectal


cancer
Matthew Loft, Yat Hang To, Peter Gibbs, Jeanne Tie

Liquid biopsies that detect circulating tumour DNA (ctDNA) have the potential to revolutionise the personalised Lancet Gastroenterol Hepatol
management of colorectal cancer. For patients with early-stage disease, emerging clinical applications include the 2023

assessment of molecular residual disease after surgery, the monitoring of adjuvant chemotherapy efficacy, and early Published Online
July 24, 2023
detection of recurrence during surveillance. In the advanced disease setting, data highlight the potential of ctDNA
https://doi.org/10.1016/
levels as a prognostic marker and as an early indicator of treatment response. ctDNA assessment can complement S2468-1253(23)00146-2
standard tissue-based testing for molecular characterisation, with the added ability to monitor emerging mutations Division of Personalised
under the selective pressure of targeted therapy. Here we provide an overview of the evidence supporting the use of Oncology, Walter and Eliza Hall
ctDNA in colorectal cancer, the studies underway to address some of the outstanding questions, and the barriers to Institute of Medical Research,
Parkville, VIC, Australia
widespread clinical uptake.
(M Loft MBBS, Y H To MBBS,
Prof P Gibbs MD, Prof J Tie MD);
Introduction Curable disease Department of Medical
The term cell-free DNA (cfDNA) refers to genetic MRD detection and monitoring efficacy of adjuvant Biology, University of
Melbourne, Parkville, VIC,
fragments in the form of nucleic acid chains found in treatment
Australia (M Loft, Y H To,
many bodily fluids. The presence of cfDNA in the blood MRD refers to occult tumour remaining after surgery Prof P Gibbs, Prof J Tie);
was initially described in 1948 by Mandel and Metais,1 with curative intent, typically at a distant site, where the Department of Medical
with reports in 1977 of increased serum concentrations in tumour bulk is below the resolution of radiological Oncology, Western Health,
Footscray, VIC, Australia
patients with cancer.2 In patients with malignancy, a detection but measurable by molecular techniques. The
(M Loft, Prof P Gibbs);
proportion of cfDNA is derived from cancer cells and is possibility of using ctDNA to detect MRD was first Department of Medical
referred to as circulating tumour DNA (ctDNA; figure 1). described by Diehl and colleagues4 in a study of Oncology, Peter MacCallum
Technological advances have addressed many of the 18 patients with resected colorectal cancer liver Cancer Centre, Parkville, VIC,
Australia (Y H To, Prof J Tie);
initial issues related to sensitivity and specificity, making metastases, where 15 (94%) of 16 patients with a positive Sir Peter MacCallum
ctDNA a promising clinical tool across cancer types and postoperative ctDNA had a disease recurrence, and no Department of Oncology,
stages (figure 2). recurrences were observed in the two patients with University of Melbourne,
In contrast to many standard blood-based biomarkers, negative postoperative ctDNA.4 Observational studies, Parkville, VIC, Australia
(Prof J Tie)
such as carcinoembryonic antigen, ctDNA has a half-life using various fit for purpose ctDNA assays, have
Correspondence to:
of only 2 h,3,4 conferring a distinct advantage for ctDNA repeatedly shown that a high proportion of postoperative
Prof Jeanne Tie, Sir Peter
as a dynamic biomarker of tumour burden. This shorter patients with detectable ctDNA after surgery for colorectal MacCallum Department of
half-life, along with the considerably superior sensitivity cancer (approaching 100% in some series) will later Oncology, University of
and specificity of ctDNA as a cancer biomarker, and develop clinical recurrence if they do not receive adjuvant Melbourne, Parkville 3000, VIC,
Australia
the ability to characterise tumour-specific genetic and chemotherapy.7–10 Completed observational studies are jeanne.tie@petermac.org
epigenetic abnormalities, extends the potential clinical presented in table 1. The clinical utility of preoperative
utility of ctDNA far beyond that of traditional markers. ctDNA assessment for colorectal cancer remains
Many ctDNA assays are commercially available, with unproven; most series9,12,21 report a high rate of pre­
many more in development. Beyond the fundamental operative ctDNA positivity and no statistical prognostic
differences between a tumour-agnostic (plasma analysis significance, probably because the dominant source of
only) and a tumour-informed (initial genomic profiling of ctDNA was the primary tumour. By contrast, few patients
a patient’s tumour tissue to identify patient-specific remain ctDNA positive post-operatively,12,21 and ctDNA-
somatic mutations that could be monitored in the plasma) positivity is highly correlated with recurrence risk,
approach, ctDNA detection methods vary considerably. independent of any preoperative ctDNA result.
Notably, plasma only ctDNA assays developed for Patients are currently not monitored during the
metastatic disease genotyping, typically with a limit of 3–6 months of adjuvant therapy, with carcinoembryonic
detection of 0·5–1%, are not suitable for the assessment antigen concentrations potentially being falsely elevated
of molecular residual disease (MRD).5,6 Also, the clinical by chemotherapy administration.22 Currently, there are
utility of each distinct ctDNA test should be independently no biomarkers that can indicate the success (or failure) of
validated through well conducted clinical trials. Further adjuvant treatment in any individual patient. When
consideration of ctDNA methods is beyond the scope of adjuvant chemotherapy is administered ctDNA can
this Review, but for each result reported we will specify become undetectable, correlating with a reduced risk of
the testing platform used. The potential of ctDNA in recurrence.21,23 Emerging evidence shows that ctDNA is
population-based cancer screening is another exciting not only useful as a prognostic biomarker post-surgery,
application, but is also beyond the scope of this review, but that serial ctDNA analysis holds promise as an early
which focuses on patients with colorectal cancer. real-time biomarker of adjuvant treatment benefit.

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 1


Review

Tumour tissue Healthy tissue


Factors affecting ctDNA concentrations and
(eg, haematopoietic cells)
detection:
• Metastatic disease sites (liver > lung,
Apoptosis
peritoneum, and bone)*
or necrosis
• Cancer burden
• Disease status (eg, before or after surgery,
responding or stable vs progressive disease)
• Types of variant (clonal > sub-clonal)*
• Timing of blood sampling in relation to
local or systemic treatment
• Non-tumour-related factors
(eg, inflammation, infection, and exercise)

ctDNA cfDNA

CTC

Analysis of ctDNA to detect cancer-related changes

A T G C G A C T

Point mutations Translocations and fusions Amplifications and deletions

Me1

Me1

Chromosomal abnormalities Epigenetic modifications (eg, methylation) DNA fragment lengths

Figure 1: The origin of ctDNA, cancer-related changes in ctDNA, and factors affecting ctDNA concentrations and detection
cfDNA=cell free DNA. CTC=circulating tumour cells. ctDNA=circulating tumour DNA. *The greater-than sign represents differences in ctDNA concentrations by
various factors.

Stage I–III colon cancer a small number will have clincial occult disease and have
The likelihood of clinical occult disease increases with a recurrence. Potentially any patient with clinical occult
advancing tumour stage and the presence of adverse disease, regardless of stage, could benefit from adjuvant
clinicopathological factors. For stage III colon cancer, chemotherapy.
historical data suggest clinical occult disease that can Current studies in stage III colon cancer are exploring
cause subsequent recurrence is present in around 50% treatment escalation in patients with detectable ctDNA
of patients.24,25 Adjuvant chemotherapy is standard of care and de-escalation in those without detectable ctDNA. For
because it can eradicate clinical occult disease in a stage II colon cancer, the focus is on avoiding adjuvant
proportion of these patients, reducing recurrence risk chemotherapy for patients in whom adjuvant chemo­
and improving survival.24–27 However, adjuvant chemo­ therapy is unlikely to be beneficial (no detectable ctDNA)
therapy provides no benefit in the 50% of patients and exploring treatment benefit in those who are ctDNA
without clinical occult disease and will only prevent positive. Patients with stage I disease are not the focus of
recurrence in about a third of those with clinical occult current interventional trials.
disease.26,27 Ultimately, few patients benefit from adjuvant In an initial observational study of 230 patients with
treatment. resected stage II colorectal cancer, ctDNA detection (using
Whether there is a benefit for adjuvant chemotherapy the tumour-informed Safe Sequencing System [Safe-SeqS]
in patients with stage II colorectal cancer remains assay) was associated with a markedly reduced recurrence-
contentious, even for patients with adverse features free survival, with an estimated 3-year recurrence-free
associated with a high risk of recurrence, with no clear survival of 0% for ctDNA-positive patients versus 90% for
overall survival benefit shown in a population with low ctDNA-negative patients not receiving adjuvant
overall recurrence risk.28 For patients with low-risk chemotherapy (hazard ratio [HR] 18, 95% CI 8–40;
stage II cancers, and even for patients with stage I cancers, p<0·0001).7 The same authors reported on an observational

2 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

Curable disease Molecular residual disease Adjuvant treatment response Surveillance


ctDNA testing Serial ctDNA testing
ctDNA ctDNA
Positive Negative Positive Negative

Starting Escalate adjuvant De-escalate or avoid Starting Further Surveillance Improved lead time vs CEA
3–4 weeks after treatment adjuvant treatment 2–8 weeks after treatment? or imaging for progression,
surgical completion of all with opportunity to increase
resection with Refined patient selection in addition to curable-intent Opportunity to escalate or consider detection of potentially
Definitive treatment curable intent clinicopathological parameters treatment second-line treatment resectable or ablatable disease

Non-curable Prognostication Treatment selection Response monitoring Personalised


disease At diagnosis Refine upfront prognosis prediction Single agent Identify responses as treatment
in addition to conventional markers Doublet early as second cycle Switch
such as BRAF status Triplet De-escalate
Anti-VEGF Rechallenge
Anti-EGFR Novel therapies
Immunotherapy Trials
Genotyping Anti-BRAF Resistance monitoring
Detection influenced Starting
Alternative to tumour tissue Anti-HER2 Non-invasive and
by disease site: 2–4 weeks
genotyping, especially when tissue Other targets dynamic monitoring
↑Liver after
biopsies are suboptimal or rapid of emerging resistance
↓Lung only treatment
↓Peritoneal only turnaround time is crucial
initiation
Palliative treatment ↓Nodal only

Figure 2: Potential clinical applications of ctDNA assessment for management of colorectal cancer with curative intent (stage I—III or resectable oligometastatic stage IV) or palliative intent
(stage IV)
ctDNA=circulating tumour DNA. CTC=circulating tumour cells.

study of 96 patients with stage III colon cancer that Research by Henriksen and colleagues20 assessed
measured ctDNA using the same assay.8 All patients 140 patients with stage III colorectal cancer with serial
underwent surgery and received standard adjuvant ctDNA measurements (tumour-informed Signatera assay)
chemotherapy. A positive postoperative ctDNA result was both before and after adjuvant chemotherapy. Post­
associated with an inferior 3-year recurrence-free survival operative ctDNA was detected in 20 (14%) patients, of
compared with a negative postoperative ctDNA result (HR whom 18 (90%) received adjuvant chemotherapy. Of the
3·8, 95% CI 2·4–21·0; p<0·001) Additionally, patients with 13 patients with assessable ctDNA after adjuvant chemo­
detectable ctDNA following chemotherapy similarly had therapy, only three (23%) showed durable ctDNA
an inferior 3-year recurrence-free survival (30% vs 77%, clearance and did not relapse at 3 years. However, all ten
HR 6·8, 95% CI 11·0–157·0; p<0·001). The median time patients who had no or only transient ctDNA clearance
to recurrence from the post-chemotherapy blood draw was relapsed. Correspondingly, persistently detectable ctDNA
short at 51 days (range, 9–470 days).8 A separate after adjuvant chemotherapy was associated with a
observational study of 130 patients with significantly lower recurrence-free survival (HR 50·76,
stage I–III colorectal cancer reported that patients with 95% CI 15·4–167·0; p<0·001).20 Other observational
positive ctDNA (tumour-informed Signatera assay) after studies have shown similar results, with the ability of
definitive therapy during surveillance were at a higher risk adjuvant chemotherapy to convert postoperative ctDNA-
of disease recurrence than were ctDNA-negative patients positivity into ctDNA-negativity after chemotherapy in
(HR 43·5, 95% CI 9·8–193·5; p<0·001).9 A later pooled patients with early-stage colorectal cancer ranging from
analysis29 of three cohort studies of 485 patients with stage 17% to 50%.8,9,18
II–III colorectal cancer found that 5-year recurrence-free The large observational GALAXY trial reported data on
survival was significantly lower in postoperative patients 315 patients with stage II colorectal cancer and 397 patients
testing positive for ctDNA (38·6%, 95% CI with stage III colorectal cancer and analysed ctDNA via
27·7–53·9) compared with those testing negative the Signatera assay.21 Postoperative ctDNA detection was
(85·5%, 81·8–89·3; HR 7·56, 95% CI 4·85–11·79; associated with inferior 18-month disease-free survival
p<0·001) 5-year overall survival was also significantly lower compared with no postoperative ctDNA detection for both
in patients with detectable (64·6%, 95% CI 47·8–87·4) stage II (34·7%, 95% CI 15·7–54·7 vs 94·0%, 90·6–96·2;
versus undetectable (89·4%, 85·9–93·1) postoperative HR 18·0, 95% CI 8·7–35·0; p<0·0001) and stage III
ctDNA (HR 3·77, 95% CI 1·96–7·25; p<0·001). ctDNA was (49·0%, 38·3–58·9 vs 92·4%, 88·6–95·0; HR 9·6,
a more accurate predictor of recurrence than standard 5·8–16·0; p<0·0001). Additionally, ctDNA positivity
individual clinicopathological factors.29 4 weeks after surgery was the most significant independent

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 3


Review

Patient population Sample size ctDNA assay ctDNA sampling Neoadjuvant or Key results
(approach, number timepoints adjuvant
of variants tracked chemotherapy
in plasma) given
Tie et al, 20167 Stage II 230 Safe-SeqS (TI, 1) 4–10 weeks Adjuvant (23%) In patients not treated with adjuvant therapy, presence of ctDNA
postoperative; after surgery was associated with an inferior 3-year RFS
serial follow-up (0% vs 90%; HR 18 [95% CI 8–40]; p<0·0001); ctDNA detection
for 24 months during surveillance identified recurrence with a median lead time
of 5·5 months
Reinert et al, Stage I–III 125 (5 with Signatera (TI, 16) Preoperative; Adjuvant (62%) Preoperative ctDNA detected in 89% of all patients;
20199 stage I; 39 with postoperative postoperative ctDNA-positive patients were significantly more
stage II; 81 with day 30 likely to have disease recurrence than ctDNA-negative patients
stage III) (HR 7·2 [95% CI 2·7–19·0]; p<0·001); ctDNA detection during
surveillance identified recurrence with a median lead time of
8·7 months
Murray et al, Stage I–V 172 (93 with Triplex real-time Within NR Patients with any feature suggestive of residual disease (close
201811 stage I or II; qPCR assay (TA, 12 months resection margins, apical node involved, distant metastases)
79 with stage III 2 methylated postoperative were 5·3-times more likely to be ctDNA-positive (p=0·008);
or IV) markers) postoperative ctDNA-positivity associated with an increased
recurrence risk (HR 3·8; p=0·004).
Tie et al, 201912 Locally advanced 159 Safe-SeqS (TI, 1) Pre-CRT; Neoadjuvant (CRT; Pre-treatment ctDNA detected in 77%; no difference in RFS
rectal cancer 4–6 weeks after 100%); adjuvant between detected and undetected (HR 1·1; p=0·82); 3-year
CRT; 4–10 weeks (36%) RFS was significantly worse if ctDNA was positive post-CRT
postoperative (HR 6·6 [95% CI 2·6–17·0]; p<0·001) or postoperatively (HR 13·0
[5·5–31·0]; p<0·001); 3-year RFS was 33% (95% CI 16–72) for
postoperative ctDNA-positive patients and 87% (79–95) for
postoperative ctDNA-negative patients
Mason et al, Stage IV post-liver 63 Guardant360 (TA, Postoperative, Neoadjuvant (87%); 2-year OS from date of liver resection was significantly worse for
202113 metastasectomy 70 cancer-related median adjuvant (59%) ctDNA-positive vs ctDNA-negative patients (70% vs 100%;
genes) 13 months p=0·005)
Tie et al, 202114 Stage IV CRC with 54 Safe-SeqS (TI, 1) Pre-treatment Neoadjuvant (43%); Pre-treatment ctDNA detected in 85%; no difference in RFS
upfront resectable (surgery or adjuvant (78%) (HR 4·72; p=0·13) or OS (HR 1·39; p=0·66) between detected and
liver metastases chemotherapy); undetected; median 41-fold (p<0·001) decrease in ctDNA mutant
before each cycle allele fraction with neoadjuvant chemotherapy, but ctDNA
of neoadjuvant clearance was not associated with improved RFS; RFS was
treatment; significantly worse if ctDNA was positive postoperatively
4–10 weeks (HR 6·3 [95% CI 2·6–15·2]; p<0·001), as was OS (HR 4·2 [1·5–11·8];
postoperative; p<0·001); RFS after all treatment (surgery with or without adjuvant
after adjuvant chemotherapy) was significantly better in those with a post-
therapy treatment ctDNA-negative result (0% vs 76%; HR 14·9; p<0·001).
Lee et al, 202115 Stage IV post 58 (38 liver; Ultra-deep targeted Preoperative; Neoadjuvant (47%); Preoperative ctDNA detected in 24%; detection rate of ctDNA
metastasectomy 25 lung; sequencing in 50 3–4 weeks adjuvant (NR) was higher in patients with liver metastases (p=0·0045) or
4 peritoneum; cancer-related genes postoperative tumours measuring 1 cm or more (p=0·018); ctDNA was less
2 lymph node) (TI, NR) likely to be detected after a complete or partial response to
chemotherapy.
Chen et al, Stage II–III 240 (112 with Geneseeq Prime (TI, Preoperative; Adjuvant (73%) Preoperative ctDNA detected in 64%; preoperative detection
202116 stage II; 128 with median 6) once between associated with reduced RFS (HR 5·7; p=0·004); postoperative
stage III) days 3–7 ctDNA-positive patients had a much higher recurrence risk
postoperative; (HR 10·98; p<0·001); ctDNA detection during surveillance
serial follow-up identified recurrence with an accuracy of 92% and mean lead
for 24 months time of 5 months
Loupakis et al, Stage IV post-liver 112 Signatera (TI, 16) Postoperative, Neoadjuvant 96·7% of ctDNA-positive patients had progression, with ctDNA-
202117 metastasectomy median 27 days (49%); adjuvant positivity associated with inferior OS (HR 16·0 [95% CI 3·9–68·0];
(44%) p<0·001); in multivariate analysis, ctDNA-positivity was the
most significant prognostic factor for DFS (HR 5·8 [3·34–10·0];
p<0·001); for ctDNA-positive patients that progressed, the
median lead time was 3·2 months
Parikh et al, Stage I–IV 84 (8 with stage Guardant Reveal Preoperative; Neoadjuvant (45%); ctDNA-positivity after definitive treatment (either surgery only or
202118 I; 20 with stage II; (TA, 40 genes plus 4 weeks adjuvant (55%) completion of adjuvant therapy) predicted relapse in 15 (100%)
40 with stage III; methylation panel) postoperative; of 15 patients; 12 (25%) of 49 patients who were ctDNA-negative
16 with stage IV) 4 weeks after after treatment had recurrence
adjuvant
treatment
(Table 1 continues on next page)

4 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

Patient population Sample size ctDNA assay ctDNA sampling Neoadjuvant or Key results
(approach, number timepoints adjuvant
of variants tracked chemotherapy
in plasma) given
(Continued from previous page)
Taieb et al, Stage III 1017 Multiplex ddPCR Postoperative Adjuvant (100%) 3-year DFS was 66% for ctDNA-positive patients and 77% for
202119 (TA, 2 methylation ctDNA-negative patients (p=0·015), with ctDNA an independent
markers) prognostic marker for DFS (adjusted HR 1·55 [95% CI 1·13–2·12];
p=0·006) and OS (HR 1·65 [1·12–2·43]; p=0·011)
Henriksen et al, Stage III 160 Signatera (TI, 16) Preoperative; Adjuvant (87%) Preoperative ctDNA detected in 91%; ctDNA-positive patients
202220 postoperative had a much higher recurrence risk both postoperatively (HR 7·0
(within 8 weeks [95% CI 3·7–13·5]; p<0·001) and after adjuvant treatment (HR 51
of surgery, [15·4–167]; p<0·001); ctDNA detection during surveillance
median 2 weeks); identified recurrence with a median lead time of 9·8 months
after adjuvant
treatment
(within
3 months)
Kotani et al, Stage I–IV 1039 (97 with Signatera (TI, 16) Preoperative; Adjuvant (45% of Preoperative ctDNA detected in 91% but not associated with RFS
202321 stage I; 315 with 4, 12, 24, 36, 48, high-risk stage II (HR 0·89; p=0·62); postoperative ctDNA-positivity was
stage II; 397 with and 72 weeks and stage III) associated with increased recurrence risk (HR 10·0 [95% CI
stage III; 230 with postoperative 7·7–14·0]; p<0·0001); stage II (HR 18·0 [8·7–35·0]; p<0·0001);
stage IV) stage III (HR 9·6 [5·8–16·0]; p<0·001); stage IV (HR 5·9 [3·9–9·0];
p<0·0001); postoperative ctDNA-positivity identified patients
with stage II or III CRC who derived benefit from adjuvant
chemotherapy (HR 6·6 [3·53–12·3]; p<0·0001)
CRC=colorectal cancer. ctDNA=circulating tumour DNA. ddPCR=digital droplet PCR. DFS=disease-free survival. HR=hazard ratio. NR=not reported. OS=overall survival. qPCR=quantitative PCR. RFS=recurrence-
free survival. Safe-SeqS=Safe-Sequencing System. TI=tumour-informed. TA=tumour-agnostic. CRT=chemoradiation.*Including non-metastatic studies if the sample size was 100 or more and oligometastatic
studies if the sample size was 50 or more.

Table 1: Selected observational ctDNA studies assessing molecular residual disease in curatively resected colorectal cancer*

prognostic factor associated with increased risk of To our knowledge, the DYNAMIC trial was the first
recurrence (HR 10·82, 95% CI 7·07–16·60; p<0·001).21 randomised controlled trial to report on a ctDNA-guided
Among patients with high-risk stage II disease with approach to the initiation of adjuvant chemotherapy in
adverse risk features (eg, T4 disease, perforation, patients with stage II colon cancer.30 In the trial,
obstruction, or lympho­vascular invasion) and all patients 455 patients were randomly assigned (2:1) to have
with stage III disease who had detectable ctDNA after adjuvant treatment decisions guided by ctDNA results
surgery, those who received any adjuvant chemotherapy (using the tumour-informed Safe-SeqS assay) or by
had significantly higher 18-month disease-free survival standard clinicopathological features, at the discretion of
than those who did not (61·6%, 95% CI 49·0–71·9 vs the treating clinician. Patients who were ctDNA-positive
22·0%, 10·9–35·5; HR 6·59, 95% CI 3·53–12·30; at 4 weeks or 7 weeks after surgery received adjuvant
p<0·0001), suggesting benefit from adjuvant chemotherapy; ctDNA-negative patients were not treated.
chemotherapy in ctDNA-positive patients.21 Fewer patients in the ctDNA-guided group than in
In a subset of the GALAXY trial consisting of 182 patients the standard group received adjuvant chemotherapy
with stage I–III disease and resectable stage IV disease (45 [15%] of 294 patients vs 41 [28%] of 147 patients; relative
who had positive ctDNA 4 weeks after surgery and ctDNA risk 1·82, 95% CI 1·25–2·65), without compromising
clearance data available, patients who received adjuvant 2-year recurrence-free survival (93·5% vs 92·4%). In the
chemotherapy had a higher ctDNA clearance rate ctDNA-guided group, 3-year recurrence-free survival was
(63 [68%] of 92) than those who did not (11 [12%] of 90; 92·5% among ctDNA-negative patients and 86·4%
HR 8·5, 95% CI 4·2–17·3; p<0·0001).21 Not achieving among ctDNA-positive patients treated with adjuvant
ctDNA clearance on adjuvant chemo­ therapy was chemotherapy, suggesting a treatment benefit in this
associated with inferior disease-free survival compared patient subset. A prespecified analysis of the DYNAMIC
with achieving ctDNA clearance (HR 11·0, 5·2–23·0; study presented at the European Society for Medical
p<0·0001). Not receiving adjuvant chemo­ therapy was Oncology 2022 Congress found a ctDNA clearance rate
found to be the most significant negative prognostic factor of 87% (n=34) at 4 weeks after completing adjuvant
in multivariate analysis for MRD-positive, ctDNA-positive chemotherapy in the 39 patients with positive post­
patients across stages II–IV of disease.21 Preliminary data operative ctDNA for whom post-adjuvant chemotherapy
from both the GALAXY study and an Australian pooled samples were available.23 Patients who had ctDNA
analysis29 suggest that postoperative ctDNA concentrations clearance had a far superior outcome to those who had
(MRD) might impact the efficacy of adjuvant treatment. persistently detectable ctDNA, with an estimated 1-year

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 5


Review

recurrence-free survival from the end of treatment of Reveal assay) in 43 (83%) of 52 baseline samples and in
100% versus 20% (HR 55·7, 95% CI 5·8–532·2; seven (16%) of 45 samples following total neoadjuvant
p<0·001).23 To our knowledge, the DYNAMIC study therapy.34 Although no association was found between
represents the first evidence from a randomised trial ctDNA status before and after total neoadjuvant therapy
that a ctDNA guided approach to adjuvant treatment and pathological complete response, ctDNA positivity
selection can reduce adjuvant chemotherapy admin­ after total neoadjuvant therapy but before surgery was
istration without compromising survival outcomes. associated with increased recurrence risk (HR 4·0,
95% CI 1·0–16·2; p=0·033) and reduced overall survival
Locally advanced rectal cancer (HR 23·0, 2·4–212·0; p<0·0001).34 A study by Wang and
The treatment approach for locally advanced rectal colleagues35 combined ctDNA assessment (tumour-
cancer is rapidly evolving, with data supporting the use agnostic next-generation sequencing panel) with MRI,
of total neoadjuvant therapy (the delivery of both showing that patients with locally advanced rectal
systemic chemotherapy and chemoradiotherapy before cancer who cleared their ctDNA following neoadjuvant
surgery).31,32 Patients who have a clinical complete chemoradiotherapy had a low probability of residual
response to neoadjuvant therapy (per endoscopy and tumour (odds ratio 0·11, 95% CI 0·01–0·60; p=0·04).
clinical imaging) are increasingly being offered an Conversely, detectable ctDNA after neoadjuvant therapy
organ preservation approach as opposed to proceeding was associated with decreased recurrence-free survival
with surgery, although mature survival data are absent (HR 9·29, 95% CI 3·74–23·10; p<0·001) and those with
and there are no reliable biomarkers to inform this detectable ctDNA and high-risk MRI features of the
decision. primary tumour at baseline had the highest recurrence
Most ctDNA data are from observational studies of risk (HR 90·29, 17·01–479·26; p<0·001).35 Overall, these
patients undergoing standard long-course chemo­ data suggest that ctDNA clearance after chemoradiation
radiation followed by surgery. In the largest series to date or total neoadjuvant therapy is a positive prognostic
of 159 patients with locally advanced rectal cancer, 3-year marker for distant recurrence but does not predict patho­
recurrence-free survival was significantly lower when logical complete response. A potential application might
ctDNA (using the Safe-SeqS assay) was detectable be to identify those who should not be recommended
(50%, 95% CI 28–88) compared with undetectable for organ preservation if ctDNA does not clear after
(85%, 79–93) following long-course neoadjuvant chemo­ neoadjuvant therapy.
radiation (HR 6·6, 95% CI 2·6–17·0; p<0·001) and also Neoadjuvant immunotherapy is an emerging strategy
post-surgery (33%, 16–72 vs 87%, 79–95; HR 13·0, in the small number of patients with colorectal cancers
5·5–31·0; p<0·001).12 Post-surgery ctDNA detection with deficient mismatch repair.36 In a study of
predicted recurrence independently of conventional 35 patients (27 with colorectal cancer), Ludford and
clinicopathological risk factors, and ctDNA-positive colleagues37 reported pathological complete response in
patients not treated with adjuvant chemotherapy had 11 (79%) of 14 patients with deficient mismatch repair
the highest recurrence risk.12 Patients who were and microsatellite instability high colorectal cancer
ctDNA-negative after neoadjuvant chemoradiation had who received neoadjuvant pembrolizumab. Among
numerically higher rates of pathological complete 19 patients in the overall study with serial ctDNA samples
response than patients who were ctDNA-positive after at baseline and after 3 weeks of pembrolizumab (assessed
neoadjuvant chemoradiation (28 [21%] of 132 patients vs with a 70-gene liquid biopsy panel), 15 (79%) had a
one [8%] of 12; p=0·46). In an analysis of 60 patients with decrease in serial ctDNA measurements. Three (75%)
locally advanced rectal cancer treated on the STELLAR of four patients with increasing or stable ctDNA
trial, by use of a tumour-informed approach tracking up concentrations between baseline and after 3 weeks of
to 22 variants, detectable ctDNA following neoadjuvant treatment showed progression, suggesting that early
therapy (pre-surgery) was associated with an increased serial ctDNA assessment might identify patients with
risk of recurrence versus undetectable ctDNA (HR 27·38, immunotherapy-resistant tumours.
95% CI 8·61–87·06; p<0·0001), but the absence of ctDNA
was not associated with pathological complete response.33 Resectable colorectal liver metastases
Notably, patients who had clearance of ctDNA (a ratio For patients with metastatic colorectal cancer who present
of post-neoadjuvant therapy ctDNA fraction to baseline with liver-only disease, surgery with curative intent might
ctDNA fraction of <2%) were more likely to have either a be possible either initially or after a response to
pathological complete response or a clinical complete neoadjuvant therapy. The utility of adjuvant chemo­
response than were those who had ctDNA persistence therapy after surgery remains uncertain, irrespective of
(seven [44%] of 16 patients vs one [5%] of 19 patients; whether neoadjuvant therapy is given.38
p=0·013). The potential use of ctDNA analysis at different
The GEMCAD 1402 trial analysed 72 patients timepoints in the management of oligometastatic liver
undergoing total neoadjuvant therapy for locally disease has been explored by several groups. Tie and
advanced rectal cancer, detecting ctDNA (Guardant colleagues14 analysed serial samples from 54 patients

6 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

with upfront, resectable colorectal cancer liver 1·7–11·5 months).7–9,20,42–45 The range of lead times
metastases, of whom 23 (43%) received neoadjuvant reported probably reflect several factors including the
chemotherapy, 42 (78%) received adjuvant chemotherapy, variable timing and frequency of ctDNA sampling,
and 16 (30%) received both. ctDNA (using the Safe-SeqS imaging protocols, and assay sensitivities. Prospective
assay) was detectable in 46 (85%) of 54 patients prior to studies showing the clinical effect of adding ctDNA
any treatment and in 12 (24%) of 49 patients after analysis to standard surveillance are needed before
surgery. Neoadjuvant chemotherapy resulted in a clinical adoption.
median 41-fold decrease (19·10–87·3, p<0·001) in ctDNA
mutant allele fraction, but ctDNA clearance during Non-curable disease
neoadjuvant therapy was not associated with improved ctDNA concentrations are elevated at presentation in the
recurrence-free survival. Patients with detectable versus great majority of patients with metastatic colorectal
undetectable ctDNA after surgery had a significantly cancer,46 but it should be noted that the sites of metastatic
lower recurrence-free survival (HR 6·3, 95% CI disease can affect ctDNA burden and detection rate.47–49
2·6–15·2; p<0·001) and overall survival (HR 4·2, Bando and colleagues48 showed that in patients with
1·5–11·8; p<0·001). Among the 11 (27%) patients metastatic colorectal cancer with single organ metastatic
with detectable with detectable postoperative ctDNA and disease, the ctDNA fraction (expressed as the maximum
available post-adjuvant chemotherapy samples, 5-year variant allelic fraction by Guardant360 assay) was
recurrence-free survival was longer for patients who significantly lower in patients with lung-only or peritoneal-
cleared their ctDNA after adjuvant chemotherapy than only disease compared with those with liver metastases.
for those with persistently positive ctDNA (66·7% vs
0·0%; HR 7·87, 0·95–63·70; p=0·056). Prognostication
Other studies have also consistently shown inferior An elevated ctDNA at diagnosis of metastatic colorectal
progression-free survival associated with ctDNA cancer is associated with a poorer prognosis. Research by
detection after surgery.21,39,40 Loupakis and colleagues17 Manca and colleagues49 used variant allele frequency to
analysed a cohort of 112 patients with metastatic quantify a high or low ctDNA burden in patients with
colorectal cancer who had undergone metastatic RAS wild-type (RASWT) metastatic colorectal cancer from
resection with curative intent, detecting ctDNA the Valentino study. Manca and colleagues showed that a
(Signatera assay) in 61 (54%) patients, 59 (97%) of whom higher pre-treatment ctDNA was associated with poorer
ultimately had progression. Postoperative ctDNA progression-free survival and markedly shorter overall
detection was associated with inferior recurrence-free survival than lower pre-treatment ctDNA (median overall
survival (HR 5·8, 95% CI 3·5–9·7; p<0·001), as well as survival 21·8 months vs 36·5 months; HR 1·82, 95% CI
overall survival (HR 16·0, 3·9–68·0; p<0·001).17 In 1·20–2·76, p=0·005).49 Carcinoembryonic antigen and
multivariate analysis, MRD status based on ctDNA result radiological measurement of initial disease burden were
was the most significant prognostic factor associated not prognostic. Other studies have identified that high
with disease-free survival (HR 5·78, 3·34–10·00; p<0·001). pre-treatment ctDNA concentrations are independently
In a prospective ancillary study to the Prodige-14 trial associated with poorer survival.50,51 However, there
(droplet digital PCR assay for KRAS mutation), in which remains no standard definition of what constitutes a
patients with potentially resectable colorectal cancer liver high ctDNA, or guidance as to how this result should
metastases were treated with first-line triplet or doublet inform therapy.
chemotherapy combined with targeted therapy, those
who had clearance of ctDNA after 4 weeks of preoperative Tumour genotyping at diagnosis
treatment had higher R0/R1 resection rates compared Anti-EGFR therapy, either in combination with chemo­
with those who did not have clearance (11 [85%] of therapy or as mono­therapy, is an important part of the
13 vs eight [36%] of 22; p=0·01), but ctDNA clearance did treatment for left-sided RASWT metastatic colorectal
not correlate with significantly improved survival.41 cancer.52,53 Multiple studies have shown excellent con­
cordance between RAS status in plasma and in tumour
Molecular recurrence monitoring tissue, consistently reporting agreement rates exceeding
Following completion of treatment with curative intent 90%.54–61 However, lower concordance rates were noted in
for colorectal cancer, patients are followed up with patients with peritoneal-only (14 [56%] of 25) and lung-
serial carcinoembryonic antigen tests and CT imaging, only (29 [66%] of 44) metastases, in whom RASmut is less
with the intent of detecting any recurrence before likely to be detectable in the circulation.48 Importantly,
clinical symptoms emerge, the expectation being that progression-free survival on anti-EGFR therapy appeared
earlier intervention could improve survival outcomes. similar whether RASWT status was determined by tissue
Multiple observational studies have shown that during analysis or by ctDNA.55 Some studies suggest that low-
this surveillance period, ctDNA might become level RAS mutations, measured either in tissue or ctDNA,
detectable months before radiological recurrence or a might not exclude a benefit from an EGFR inhibitor, but a
carcino­embryonic antigen increase (median lead time threshold remains to be confirmed.61

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 7


Review

Data suggest that the benefit of first-line anti-EGFR potentially identify patients with microsatellite-stable
therapy is confined to patients with metastatic tumours who will benefit from this treatment. The
colorectal cancer with a left-sided RASWT primary CO.26 trial randomly assigned patients with advanced
tumour. However, a ctDNA biomarker sub-study of colorectal cancer with treatment-refractory microsatellite-
PARADIGM (a randomised phase 3 trial comparing stable tumours to dual CTLA-4 plus PD-L1 blockade or
FOLFOX [leucovorin, fluorouracil, and oxaliplatin] plus best supportive care, and showed that, in patients treated
panitumumab vs FOLFOX plus bevacizumab as first- with immune checkpoint inhibitors, elevated ctDNA-
line treatment in patients with tissue-confirmed RASWT assessed tumour mutational burden (28 variants per
metastatic colorectal cancer) found that hyperselection megabase or more) was associated with the greatest
of patients on the basis of the absence for genomic overall survival benefit (HR 0·34, 90% CI 0·18–0·63;
alterations in cfDNA in KRAS, NRAS, BRAFV600E, PTEN, p=0·004).71 The ARETHUSA study examined the efficacy
the ectodomain (ECD) of EGFR, HER2 (also known as of priming patients with microsatellite-stable, RASmut
ERBB2), MET, ALK, RET, or NTRK1 might define metastatic colorectal cancer with temozolomide to induce
patients with RASWT metastatic colorectal cancer who response to subsequent pembrolizumab therapy.72 Plasma
could benefit most from anti-EGFR therapy regardless tumour mutational burden analysis of patients with
of tumour sidedness.62,63 These negatively hyperselected microsatellite-stable disease was similar to tissue
patients also had longer survival times compared with analysis and reflected an increase in tumour mutational
patients who had at least one of the genetic alterations. burden values after temozolomide priming, potentially
The ongoing LIBImAb trial (NCT04776655) uses both identifying those most likely to benefit from immune
tissue and ctDNA analysis to identify patients with checkpoint inhibitor treatment.72 However, successful
metastatic colorectal cancer who are most likely to derive analysis was largely limited to patients with liver-
benefit from cetuximab therapy. predominant disease.72
ctDNA analysis has also shown high concordance with
results for tissue-based testing of other molecular Assessing early treatment response
targets.64 A ctDNA substudy from the SWOG S1406 trial Serial carcinoembryonic antigen testing is cheap and
using a Guardant 68-gene next-generation sequencing widely accessible but is not a reliable marker of therapy
panel showed a concordance rate of 88% in 160 patients response in metastatic colorectal cancer. The reason
with metastatic colorectal cancer and tumour-confirmed for this unreliability is because carcinoembryonic
BRAFV600E mutation.65 The prospective TRIUMPH study antigen is not initially elevated in many patients, and
(using Guardant360) enrolled patients with either tissue- early elevations on treatment (a carcinoembryonic
confirmed or ctDNA-confirmed HER2 amplification, antigen surge) can occur in patients responding to
reporting an agreement rate of 73%.66 Patients treated chemotherapy.22 Decreasing carcinoembryonic antigen
with pertuzumab and trastuzumab had similar response concentrations are evidence of response, but knowing
rates and progression-free survival whether they had this will not change management and carcinoembryonic
tissue-confirmed or ctDNA-confirmed HER2-positive antigen changes are not necessarily proportional to the
disease.66 Similarly, the Guardant360 assay was able to RECIST-defined response seen on imaging.
detect HER2 amplification with 97·9% sensitivity (95% CI Tie and colleagues73 reported that significant reductions
87·2–99·8) in the HERACLES-A study of patients with reductions in ctDNA concentrations could be observed
metastatic colorectal cancer.67 Clinical response correlated as early as before the second cycle of first-line systemic
with increased initial plasma HER2 and increased HER2 chemotherapy in patients treated for metastatic colorectal
copy numbers.67 Exploratory analysis of patients recruited cancer. A major reduction in ctDNA mutant allele
to the DESTINY trial also observed increased response fraction (≥10-fold from baseline to after one treatment
rates associated with high ctDNA HER2 copy number but cycle) compared with pre-treatment concentrations was
noted decreased response rates associated with high also significantly associated with radiological response
blood tumour mutational burden.68 In a large study 8–10 weeks after treatment initiation.73 Carcinoembryonic
involving 3808 patients with metastatic colorectal cancer, antigen did not show any statistically significant change
ctDNA analysis using the Guardant360 assay identified a in concentration between baseline and before cycle
potentially actionable fusion, such as in RET, FGFR3, or two and correlation with radiological response was
ALK, in 40 (1·1%) patients.69 statistically inferior to that of ctDNA. Other studies have
In a pan-cancer study, results indicated that ctDNA consistently shown improved outcomes associated with
analysis with Guardant360 identified 71 (87%) of 82 cases ctDNA reduction; however, the timing of post-treatment
with high tissue microsatellite instability, but sensitivity assessment has varied from 7 days to 60 days post-
was highly dependent on ctDNA tumour fraction, and treatment.51,61,74–80 Longitudinal studies with more frequent
with markedly reduced sensitivity at tumour fraction less ctDNA testing have also indicated that serial monitoring
than 0·2%.70 Although immune checkpoint inhibitor of ctDNA is more predictive of survival than changes in
treatment is usually reserved for patients with tumours carcinoembryonic antigen or imaging.76,77 During second-
with high microsatellite instability, ctDNA might line treatment, an absence of a reduction in ctDNA

8 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

concentration as early as before cycle two correlated with therapy in combination with chemotherapy (four [6·6%]
poorer survival.79,80 of 61) compared with later lines (62%).
The optimal protocols for ctDNA testing during Results from prospective studies of rechallenge with an
treatment of metastatic colorectal cancer are yet to be anti-EGFR antibody in the third-line setting have shown
defined. Serial ctDNA analysis should have maximum the utility of ctDNA-based RAS assessment to identify
utility in later-line therapy, given the increased rates of a patient’s likelihood of benefitting from treatment.
early treatment failure. Here, identifying treatment The CHRONOS single-arm phase 2 study screened
failure within the first month of treatment could allow an 52 patients with metastatic colorectal cancer for a panel
early switch to an alternative therapy that might be more of KRAS, BRAF, and EGFR-ECD mutations before
active, while also minimising the toxicity and financial considering a panitumumab rechallenge.91 Using a
cost of persisting with a futile treatment. Such an digital droplet PCR-based assay, in an initial screen,
approach is being explored in the TACT-D and 36 (69%) of the 52 patients did not have detectable
Rapid 1 trials (table 2). resistant mutations. Of the 27 patients with no detectable
resistant mutations proceeding to treatment, partial
Detecting resistance to targeted therapy and timing of responses were observed in eight (30%), with a further
treatment re-challenge 11 (41%) having stable disease. Median duration of
The non-invasive nature of ctDNA testing lends itself response was 17 weeks and median overall survival was
well to serial testing to identify the emergence of resistant 55 weeks. In the CRICKET study, a combination of
clones. Two landmark studies concurrently showed the cetuximab and irinotecan was given as third-line
ability of serial ctDNA testing to detect mutations treatment in patients with metastatic colorectal cancer
associated with resistance in patients with RASWT who initially had responded well to first-line
colorectal cancer treated with anti-EGFR therapy. A study cetuximab-based and irinotecan-based therapy and
by Diaz and colleagues81 reported that nine (38%) of had progressed on second-line treatment.95 No RAS
24 patients developed KRAS mutations at various mutations were detected in the ctDNA of patients who
timepoints, as determined by ctDNA analysis. In a had a partial response to the rechallenge, and patients
similar study, Misale and colleagues82 described with ctDNA RASWT disease had an improved progression-
acquisition of secondary KRAS mutations in six (60%) of free survival compared with patients with ctDNA RASmut
ten analysed cases and the emergence of KRAS disease.95 The phase 2, single-arm CAVE trial rechallenged
amplification in an additional case.82 Other studies have patients with RASWT metastatic colorectal cancer with
used ctDNA to identify other molecular mechanisms third-line cetuximab and avelumab following previous
of resistance to targeted therapies, predominantly complete or partial response to first-line anti-EGFR
in the MAPK pathway, such as KRAS, NRAS, therapy.96 Exploratory biomarker analysis showed that
BRAF, MAP2K1, MAP2K2, EGFR-ECD, and MET and patients with RASWT and BRAFWT ctDNA before treatment
HER2 amplifications.83–90 Among acquired EGFR-ECD had improved survival compared with those with RASmut
mutations, Ser492Leu, Ser464Leu, Gly465Arg, Gly465Glu, or BRAFmut ctDNA, showing a clear benefit of anti-EGFR
Val441Asp, and Val441Gly are common targets used in rechallenge in patients with wild-type genes. Of these
trials examining anti-EGFR treatment rechallenge, such studies, only the CHRONOS trial enrolled patients on
as Sym00405, CHRONOS, and CITRIC.88,91,92 the basis of ctDNA screening results, showing the
Patterns of genomic alteration can also vary depending potential utility of ctDNA analysis in identifying patients
on treatment factors. A 2023 study screened for likely to benefit from anti-EGFR rechallenge.
mechanisms of resistance (including KRAS, NRAS, Mathematical modelling of resistant subclonal pop­
BRAF, MAP2K1, and EGFR-ECD) in paired plasma ulation expansion and mutant allele decay might provide
samples of patients with RASWT colorectal cancer treated insight into the ideal frequency of ctDNA monitoring and
with anti-EGFR as monotherapy in the third-line setting, the appropriate timing of anti-EGFR rechallenge. Khan
or in combination with chemotherapy in the first-line and colleagues97 developed a predictive model based on
setting.93 Acquired mutations were more likely in those ctDNA-assessed evolution of acquired subclonal RAS
treated with third-line single-agent anti-EGFR (181 [46%] pathway mutations in patients with metastatic colorectal
of 395) than in those treated with the combination in cancer treated with cetuximab. The model suggested that
the first line (seven [9%] of 77).93 A preclinical model ctDNA testing every 4 weeks or more frequently provided
showed that transcriptional mechanisms of resistance the greatest predictive power in forecasting time to
predominate when anti-EGFR therapy is combined radiologically evident progression.97 However, validation of
with chemotherapy, whereas acquired MAPK pathway this model in prospective trials is needed. Regarding the
resistance mutations did not affect sensitivity to cyto­ timing of anti-EGFR rechallenge, analysis of post-
toxic chemotherapy.93 Similarly, Raghav and colleagues94 progression ctDNA profiles of treated patients with
showed that fewer patients acquired genomic alterations metastatic colorectal cancer showed that acquired RAS or
(RAS, BRAF, and EGFR-ECD mutations, or HER2 or EGFR-ECD mutant alleles exponentially decayed with a
MET amplifications) following first-line anti-EGFR half-life of 4·4 months.98 Validation based on retrospective

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 9


Review

Patient population Sample size ctDNA assay Intervention Primary objective


ctDNA-guided anti-EGFR rechallenge
PULSE KRASWT or NRASWT mCRC, as 84 Guardant360 Control arm: physician’s choice of To assess overall survival in
(NCT03992456) assessed by ctDNA after regorafenib or TAS-102; experimental molecularly selected patients
randomised phase 2 progression on previous arm: panitumumab rechallenge with mCRC receiving
anti-EGFR therapy panitumumab rechallenge vs
standard therapy (TAS-102 or
regorafenib)
PARERE RASWT or BRAFWT mCRC, as 214 Idylla ctKRAS- Control arm: panitumumab followed Overall survival
(NCT04787341) assessed by ctDNA after NRAS-BRAF by regorafenib; experimental arm:
randomised phase 2 progression on previous mutation test regorafenib followed by panitumumab
anti-EGFR therapy
CAPRI II GOIM RASWT or BRAFWT mCRC 200 NR Dynamic and longitudinal ctDNA To assess response rates for
(NCT05312398) prior to starting treatment assessment over three lines of each treatment line over
single-arm phase 2 treatment; second-line and third-line three treatment lines
treatment guided by ctDNA assessment
of RAS and BRAF status; first-line:
FOLFIRI plus cetuximab; second-line:
FOLFIRI plus cetuximab (RASWT or
BRAFWT) or FOLFOX plus bevacizumab
(RASmut or BRAFmut); third-line: irinotecan
plus cetuximab (RASWT or BRAFWT) or
regorafenib or TAS-102 (RASmut or
BRAFmut)
CAVE II GOIM RASWT or BRAFWT, as 173 NR Control arm: cetuximab; experimental Overall survival
(NCT05291156) assessed by ctDNA after arm: cetuximab and avelumab
randomised phase 2 progression on anti-EGFR
therapy
A phase 2 study using RASWT or BRAFWT CRC 60 NR ctDNA testing following progression at Objective response rates and
cfDNA in the first line to assess emerging mutations; progression-free survival
detection of RAS second-line therapy determined
mutations in patients by physician; upon second-line
with advanced CRC progression, ctDNA will be tested again;
(NCT04775862) RASWT: investigator choice of either
anti-EGFR or third-line chemotherapy;
RASmut: investigator choice of third-line
chemotherapy
CITRIC (EudraCT RASWT, BRAFWT or EGFR- 66 NR Control arm: investigator’s choice of Objective response rate
2020–000443–31) ECDWT, as assessed by third-line therapy; experimental arm:
phase 2 ctDNA following cetuximab and irinotecan
progression on two
previous regimens of
standard therapy,
including anti-EGFR
monoclonal antibodies
Other ctDNA-guided treatments
Rapid-1 mCRC after progression 78 Signatera Both groups treated with a prespecified Overall survival
(NCT04786600) on oxaliplatin-based sequence of FDA-approved drugs and
randomised phase 2 therapy drug combinations; patients moved to
next-line treatment determined by
treatment arm; control arm (scan-
guided): results of imaging scans show
progressive disease; experimental arm
(ctDNA-guided): substantial increase in
ctDNA concentration
TACT-D mCRC with at least 100 Guardant 360 Both groups receive either regorafenib Assessment of early change
(NCT03844620) two previous therapies or TAS 102; control arm (SOC): assigned in ctDNA as a predictor of
randomised phase 2 treatment as per SOC; experimental radiographic progression
arm (ctDNA): patients undergo ctDNA (in control arm); evaluate
testing and receive either regorafenib differences in clinically
or TAS-102 on the basis of the result significant treatment-related
adverse events between SOC
and ctDNA arms
(Table 2 continues on next page)

10 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

Patient population Sample size ctDNA assay Intervention Primary objective


(Continued from previous page)
LIBImAb RASWT or BRAFWT mCRC on 280 Oncomine Cohort 1: patients with incongruent Progression-free survival in
(NCT04776655) tumour tissue Pan-Cancer RAS status (RASWT on tumour tissue and patients with RASWT or BRAFWT
randomised phase 3 Cell-Free assay RASmut on ctDNA) will be randomly on tumour tissue and RASmut on
assigned to either control (bevacizumab ctDNA
plus FOLFIRI) or experimental
(cetuximab plus FOLFIRI) groups; in
cohort 2, patients with congruent RAS
status will be treated with 8 weeks of
cetuximab plus FOLFIRI; repeat ctDNA
testing will be completed with RASmut
patients subsequently randomly
assigned to control (continuing
cetuximab) or experimental (switching
to bevacizumab) arms
COLOMATE Treatment-refractory 500 Guardant 360 Blood-based genomic profiling on Evaluate proportion of patients
(NCT03765736) mCRC patients with treatment-refractory who have an actionable
cohort mCRC to facilitate accrual to genomic profile; evaluate
molecularly assigned therapies subsequent proportion of
enrolment into companion trial
ctDNA=circulating tumour DNA. cfDNA=cell-free DNA. ECD=ectodomain. FDA=US Food and Drug Administration FOLFIRI=leucovorin, fluorouracil, and irinotecan.
FOLFOX=leucovorin, fluorouracil, and oxaliplatin. mCRC=metastatic colorectal cancer. mut=mutant-type. NR=not reported. SOC=standard of care. TAS-102=trifluridine plus
tipiracil. WT=wild-type.

Table 2: Ongoing ctDNA-based prospective trials in non-resectable metastatic colorectal cancer

analysis of 80 rechallenged patients showed that objective of the higher risk subset of patients with T4 staging.
response rates were highest in patients who waited for Further randomised trials are needed to explore various
more than two half-lives (32%) from previous anti-EGFR management scenarios based on ctDNA status for each
compared with those who waited for less than one (16%) or tumour stage, and to address key clinical management
one to two half-lives (20%), but these differences were not questions, such as how therapy should optimally be
statistically significant.98 escalated (if at all) in ctDNA-positive patients or de-
Regarding other clinically relevant molecular targets, escalated (if at all) in those with undetectable ctDNA. The
serial ctDNA monitoring of patients with BRAFV600E value of repeated ctDNA-based MRD testing in increasing
metastatic colorectal cancer receiving BRAF-targeted the negative predictive value of this assay is also being
treatment has also uncovered distinct patterns of explored in ongoing trials (table 3).
convergent genomic evolution associated with acquired Current consensus guidelines recommend that
treatment resistance. The frequent emergence of MAPK validated ctDNA assays can be used to genotype advanced-
pathway alterations could help to rationalise future stage cancers and to select patients for targeted therapies
combination treatment.99–101 Longitudinal analyses of in scenarios in which rapid results are paramount, or
patients treated with HER2 dual inhibitors have identified tissue is unavailable.105,106 The most substantial limitations
the emergence of resistant mutations, such as HER2 are the lower sensitivity for fusion events and copy
amplification and PIK3CA mutations.67,68,102 Following number changes,107,108 and the potential for non-
treatment targeting KRAS12G→C, plasma analysis after informative ctDNA results in certain patient subsets (with
progression has shown acquired resistance due to low disease burden, brain, peritoneal, or lung metastases,
alterations in the RTK pathway or secondary RAS or locoregional recurrence).109–111 For patients who are at
mutations.103,104 In cases of tyrosine kinase receptor high risk of a non-informative result, tumour tissue-
alterations, further studies are needed to explore the based genomic analysis is still advisable where possible.
efficacy of upstream inhibitors, such as those targeting Further work is also needed to address the health
SHP2.103 economics effect of ctDNA-informed management, given
the large associated costs. Securing reimbursement in
Barriers to real-world adoption of ctDNA guided some jurisdictions will usually require cost-effectiveness
decision making studies. This approach is feasible in scenarios in which
The DYNAMIC study30 in stage II colon cancer is a ctDNA-based testing can lead to avoiding treatment
landmark trial as it represents the world’s first randomised and the associated financial and other costs without
controlled trial in any solid tumour to directly compare a compromising outcomes. This possibility was explored
ctDNA-guided approach to adjuvant treatment selection in an analysis of an observational study of patients with
with standard of care. However, many questions remain, stage II colorectal cancer,112 and a health economics
including the role of ctDNA analysis in guiding treatment analysis is planned for the DYNAMIC study. An EGFR

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 11


Review

Patient population Sample size ctDNA assay Intervention Primary objective


Postoperative ctDNA-guided adjuvant management
DYNAMIC-III Stage III colon cancer 1000 Safe-SeqS Control arm: SOC (clinician-determined); To assess the effect of chemotherapy
(ACTRN12617001566325) experimental arm: ctDNA-positive, de-escalation and escalation strategies guided
escalation of chemotherapy; ctDNA- by ctDNA, including: acceptable rate of
negative, de-escalation of chemotherapy de-escalation in the ctDNA-informed negative
cohort (phase 2); non-inferior recurrence with
ctDNA-guided management in the de-escalated
ctDNA-informed negative cohort (phase 3);
superior recurrence with ctDNA-informed
management in the escalated ctDNA-informed
positive cohort (phase 3)
DYNAMIC-RECTAL Locally advanced rectal 450 Safe-SeqS Control arm: SOC (clinician-determined); Show that ctDNA results plus pathological nodal
(ACTRN12617001560381) cancer experimental arm: ctDNA-positive, adjuvant status can reduce the number of patients
chemotherapy; ctDNA-negative (ypN0), receiving adjuvant chemotherapy without
surveillance; ctDNA-negative (ypN+), compromising RFS
surveillance or adjuvant chemotherapy
(clinician’s discretion)
CIRCULATE-US Stage II and III colon 1912 Signatera If ctDNA-negative (cancer stage IIIA and IIIB To assess the effect of chemotherapy
(NCT05174169) cancer only): control arm, SOC chemotherapy de-escalation and escalation strategies guided
(3–6 months’ FOLFOX or 3 months’ CAPOX); by ctDNA
experimental arm, no chemotherapy;
if ctDNA-positive (cancer stage II or III):
control arm, 6 months’ FOLFOX or CAPOX;
experimental arm, 6 months’ FOLFOXIRI
CIRCULATE AIO-KRK-0217 ctDNA-positive stage II 2310 (total); 231 NR Control arm: surveillance; experimental arm: To assess the effect of chemotherapy on DFS in
(NCT04089631) colon cancer (ctDNA-positive) adjuvant chemotherapy (capecitabine or patients with ctDNA-positive stage II colon
CAPOX [clinician’s choice]) cancer
CIRCULATE PRODIGE 70 ctDNA-positive stage II 1980 (total); 198 ddPCR Control arm: surveillance; experimental arm: Show superior DFS for patients with ctDNA-
(NCT04120701) colon cancer (ctDNA-positive) (2 methylated adjuvant 6 months’ FOLFOX positive stage II colon cancer treated with
markers WIF1 and adjuvant FOLFOX
NPY)
COBRA (NRG GI-005; Stage IIA colon cancer 1408 Guardant LUNAR-1 Control arm: surveillance; experimental arm: To assess clearance of ctDNA for ctDNA-positive
NCT04068103) if ctDNA-positive, adjuvant FOLFOX or patients and the effect of ctDNA-guided
CAPOX; if ctDNA-negative, surveillance treatment on RFS
VEGA (UMIN000039205) ctDNA-negative high- 1240 Signatera Control arm: 3 months’ CAPOX; Show non-inferiority of observation vs adjuvant
risk stage II and low-risk experimental arm: surveillance; patients CAPOX in patients with ctDNA-negative high-
stage III colon cancer who become ctDNA-positive at 3 months risk stage II and low-risk stage III colon cancer
subsequently enrolled in ALTAIR trial
TRACC (NCT04050345) High-risk stage II colon 1000 Next-generation Control arm: 6 months’ capecitabine Show non-inferiority of DFS between SOC and
cancer, stage III colon sequencing-based or 3 months’ CAPOX; experimental arm: ctDNA-guided adjuvant treatment
cancer, and locally 22-gene colorectal if ctDNA-positive, standard adjuvant
advanced rectal cancer panel chemotherapy (as in control condition);
if ctDNA-negative, de-escalation of
chemotherapy (but re-escalate if ctDNA
becomes positive at 3 months)
MEDOCC-CrEATE Stage II colon cancer 1320 PDGx elio Control arm: surveillance; experimental arm: Investigate willingness of patients to be treated
(NL6281/NTR6455) if ctDNA-positive, 6 months’ CAPOX or with chemotherapy guided by ctDNA testing
FOLFOX; if ctDNA-negative, surveillance and the effect on recurrence rate
CIRCULATE-SPAIN ctDNA-positive stage II 164 NR Control arm: SOC CAPOX; experimental arm: To assess the proportion of patients who clear
(EudraCT 2021–000507– and III colon cancer FOLFOXIRI ctDNA and the effect on DFS with standard vs
20) intensified adjuvant treatment
OPTIMISE Curative-intent stage IV 350 NR Control arm: SOC adjuvant therapy; To assess the effect of chemotherapy
(NCT04680260) colon cancer experimental arm: if ctDNA-positive, de-escalation and escalation strategies guided
escalation of treatment to 4 months’ by ctDNA on RFS
FOLFOXIRI followed by 2 months’
fluorouracil monotherapy; if ctDNA-negative,
de-escalation based on shared decision
making (either monotherapy or observation)
Risk-stratified adjuvant Patients with stage IV 120 NR Two experimental arms; if ctDNA- To assess RFS of both ctDNA-negative and
therapy (NCT05062317) colon cancer undergoing negative, de-escalation of chemotherapy ctDNA-positive patients who received risk-
elective curative-intent (monotherapy); if ctDNA-positive, stratified adjuvant chemotherapy
hepatectomy after escalation of chemotherapy (resumption
neoadjuvant of neoadjuvant platinum-based doublet
chemotherapy with or without bevacizumab)
(Table 3 continues on next page)

12 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

Patient population Sample size ctDNA assay Intervention Primary objective


(Continued from previous page)
ctDNA-guided second-line adjuvant therapy
ALTAIR (NCT04457297) ctDNA-positive stage III 240 Signatera Control arm: surveillance; experimental arm: Show benefit of TAS-102 over surveillance in
colon cancer after second-line TAS-102 patients who remained ctDNA-positive after
3 months of CAPOX standard adjuvant chemotherapy
ACT-3 (NCT04259944) ctDNA-positive stage III 500 Guardant LUNAR-1 Control arm: surveillance; experimental arm: Show benefit of FOLFIRI over surveillance in
colon cancer after for MSS or BRAFWT, 6 months’ FOLFIRI; patients who remained ctDNA-positive after
standard adjuvant for BRAFmut, 6 months’ encorafenib– standard adjuvant therapy
chemotherapy binimetinib–cetuximab; for MSI-H,
6 months’ nivolumab
Postoperative ctDNA-guided adjuvant and second-line adjuvant management
CTAC (NCT05529615) High-risk stage II and 2684 NR Patients diagnosed with high-risk stage II Determine non-inferior recurrence risk of
stage III colon cancer and low-risk stage III cancer: if ctDNA- surveillance vs adjuvant chemotherapy in
negative, surveillance; if ctDNA-positive: patients with ctDNA-negative low-risk cancer
control arm, surveillance; experimental arm: and whether second-line chemotherapy can
CAPOX for 3 months; for patients diagnosed improve the DFS of ctDNA-positive patients vs
with high-risk stage III cancers, all receive standard chemotherapy in the high-risk group
CAPOX for 3 months; at 3 months: if ctDNA-
negative, surveillance; if ctDNA-positive:
control arm, further CAPOX for 3 months
(total 6 months); experimental arm, second-
line treatment decided by clinician
ERASE-CRC ctDNA-positive high- 300 NR Part 1: control arm, 6 months’ FOLFOX or Part 1: evaluate the effect of escalating adjuvant
(NCT05062889) risk stage II and stage III CAPOX as adjuvant treatment; experimental chemotherapy in ctDNA-positive patients on
colon cancer arm, FOLFOXIRI for 6 months as adjuvant ctDNA clearance rates and DFS; part 2: evaluate
treatment; part 2: if ctDNA-positive whether further adjuvant treatment with
after adjuvant treatment: control arm, TAS-102 in patients who remained ctDNA-
surveillance; experimental arm, six cycles of positive after adjuvant treatment can increase
TAS-102 ctDNA clearance rates and improve DFS
ctDNA-guided surveillance
IMPROVE-IT2 High-risk stage II and 254 ddPCR (colorectal Control arm: surveillance; experimental arm: Show combination of ctDNA-guided and high-
(NCT04084249) stage III colon cancer panel) 3-monthly PET-CT for ctDNA-positive intensity radiological surveillance could result in
patients earlier detection of recurrent disease and hence
more eligible patients for curative treatment
CAPOX=capecitabine and oxaliplatin. ctDNA=circulating tumour DNA. ddPCR=droplet digital PCR. DFS=disease-free survival. FOLFIRI=folinic acid, fluorouracil, and irinotecan. FOLFOXIRI=folinic acid,
fluorouracil, irinotecan, and oxaliplatin. FOLFOX=leucovorin, fluorouracil, and oxaliplatin. MSI-H=microsatellite instability-high; MSS=microsatellite stable. NR=not reported. RFS=recurrence-free survival.
Safe-SeqS=Safe-Sequencing System. SOC=standard of care. TAS-102=trifluridine plus tipiracil. ypN0=pathologic node negative disease following neoadjuvant systemic or radiation therapy. ypN+=pathologic
node positive disease following neoadjuvant systemic or radiation therapy.

Table 3: Ongoing ctDNA-based randomised trials in curatively resected colorectal cancer

inhibitor treatment rechallenge is another scenario in whom ctDNA becomes detectable during surveillance
which ctDNA-based testing might be cost-saving, in that (molecular recurrence) without detectable recurrence on
avoiding futile treatment in patients with a RAS mutation imaging would also be appealing.
would probably save sufficient money to offset the cost of It is notable that many different ctDNA assays are
a ctDNA test. being developed. Analytical performance has the
potential to differ between ctDNA MRD assays, which
Ongoing trials should preclude data from one ctDNA assay being used
Multiple ongoing studies are further exploring the utility to validate the use of a different assay. The clinical use of
of ctDNA in informing the curative intent management each individual ctDNA assay should be independently
of colorectal cancer (table 3), including after surgery, validated in carefully designed prospective studies that
during and at the completion of adjuvant therapy, and cover each specific clinical scenario.
during surveillance. Studies also continue in the
metastatic disease setting (table 2). Conclusion
Of particular interest are studies in the MRD setting ctDNA has the potential to change the treatment frame­
exploring additional therapy when patients have works for early-stage through to late-stage colorectal
completed standard adjuvant therapy without clinical cancer, allowing a higher degree of personalisation. For
recurrence but have detectable ctDNA as a marker of early disease, robust evidence currently supports ctDNA
persistent MRD—the new concept of second-line as a marker of recurrence risk, adjuvant treatment
adaptive-adjuvant therapy. Trials exploring the benefit of efficacy, and impending clinical recurrence. In the
early introduction of systemic therapy for patients for metastatic setting, ctDNA testing can also aid in

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 13


Review

13 Mason MC, Tzeng CD, Tran Cao HS, et al. Preliminary analysis of
Search strategy and selection criteria liquid biopsy after hepatectomy for colorectal liver metastases.
J Am Coll Surg 2021; 233: 82–89.e1.
Data for this Review were identified by searches of MEDLINE 14 Tie J, Wang Y, Cohen J, et al. Circulating tumor DNA dynamics and
and PubMed, and references from relevant articles using the recurrence risk in patients undergoing curative intent resection of
colorectal cancer liver metastases: a prospective cohort study.
search terms “colorectal neoplasm” and “circulating tumour PLoS Med 2021; 18: e1003620.
DNA”. Abstracts and reports from meetings were included 15 Lee S, Park YS, Chang WJ, et al. Clinical implication of liquid
only when they related directly to previously published work. biopsy in colorectal cancer patients treated with metastasectomy.
Cancers (Basel) 2021; 13: 2231.
Only articles published in English were included with no
16 Chen G, Peng J, Xiao Q, et al. Postoperative circulating tumor DNA
restriction on year of publication. as markers of recurrence risk in stages II to III colorectal cancer.
J Hematol Oncol 2021; 14: 80.
17 Loupakis F, Sharma S, Derouazi M, et al. Detection of molecular
molecular characterisation, such as identifying patients residual disease using personalized circulating tumor DNA assay in
patients with colorectal cancer undergoing resection of metastases.
who might benefit from an anti-EGFR rechallenge, and JCO Precis Oncol 2021; 5: 5.
in early monitoring of treatment response. With the 18 Parikh AR, Van Seventer EE, Siravegna G, et al. Minimal residual
results of multiple randomised trials on the horizon, the disease detection using a plasma-only circulating tumor DNA assay
in patients with colorectal cancer. Clin Cancer Res 2021; 27: 5586–94.
use of ctDNA informed therapy will become increasingly 19 Taieb J, Taly V, Henriques J, et al. prognostic value and relation with
well defined. adjuvant treatment duration of ctDNA in stage III colon cancer:
a post hoc analysis of the PRODIGE-GERCOR IDEA-France trial.
Contributors
Clin Cancer Res 2021; 27: 5638–46.
All authors contributed to conceptualisation and design. ML and
20 Henriksen TV, Tarazona N, Frydendahl A, et al. Circulating tumor
YHT conducted the literature review and wrote the first draft. DNA in stage III colorectal cancer, beyond minimal residual disease
All authors contributed to editing the initial and subsequent revisions detection, toward assessment of adjuvant therapy efficacy and
of the manuscript and approved the final version. clinical behavior of recurrences. Clin Cancer Res 2022; 28: 507–17.
Declaration of Interests 21 Kotani D, Oki E, Nakamura Y, et al. Molecular residual disease and
PG has received consulting fees from Haystack Oncology and efficacy of adjuvant chemotherapy in patients with colorectal cancer.
honoraria from Amgen, Roche, Servier, Merck, Pierre-Fabre, and MSD. Nat Med 2023; 29: 127–34.
JT has received consulting fees from Haystack Oncology and Seres 22 Sørbye H, Dahl O. Transient CEA increase at start of oxaliplatin
Therapeutics, honoraria from Astra Zeneca, Amgen, and Servier, combination therapy for metastatic colorectal cancer. Acta Oncol
2004; 43: 495–98.
support for attending meetings from Roche, participated on advisory
boards for Beigene, Novartis, Astra Zeneca, Merck, Serono, MSD, 23 Tie J, Cohen J, Lahouel K, et al. 318MO — Circulating tumour DNA
(ctDNA) dynamics, CEA and sites of recurrence for the randomised
Pierre Fabre, BMS, Daichii Sankyo, Takeda, Illumina, and Gilead, and
DYNAMIC study: adjuvant chemotherapy (ACT) guided by ctDNA
has a leadership role on AGITG and ESMO. All other authors declare no analysis in stage II colon cancer (CC). Ann Oncol 2022; 33: 33.
competing interests.
24 Moertel CG, Fleming TR, Macdonald JS, et al. Levamisole and
References fluorouracil for adjuvant therapy of resected colon carcinoma.
1 Mandel P, Metais P. [Nuclear acids in human blood plasma]. N Engl J Med 1990; 322: 352–58.
C R Seances Soc Biol Fil 1948; 142: 241–43. 25 IMPACT. Efficacy of adjuvant fluorouracil and folinic acid in colon
2 Leon SA, Shapiro B, Sklaroff DM, Yaros MJ. Free DNA in the serum of cancer. Lancet 1995; 345: 939–44.
cancer patients and the effect of therapy. Cancer Res 1977; 37: 646–50. 26 André T, Boni C, Navarro M, et al. Improved overall survival with
3 Lu L, Bi J, Bao L. Genetic profiling of cancer with circulating tumor oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in
DNA analysis. J Genet Genomics 2018; 45: 79–85. stage II or III colon cancer in the MOSAIC trial. J Clin Oncol 2009;
4 Diehl F, Schmidt K, Choti MA, et al. Circulating mutant DNA to 27: 3109–16.
assess tumor dynamics. Nat Med 2008; 14: 985–90. 27 Yothers G, O’Connell MJ, Allegra CJ, et al. Oxaliplatin as adjuvant
5 Moding EJ, Nabet BY, Alizadeh AA, Diehn M. Detecting liquid therapy for colon cancer: updated results of NSABP C-07 trial,
remnants of solid tumors: circulating tumor DNA minimal residual including survival and subset analyses. J Clin Oncol 2011;
disease. Cancer Discov 2021; 11: 2968–86. 29: 3768–74.
6 Yu L, Lopez G, Rassa J, et al. Direct comparison of circulating 28 Rebuzzi SE, Pesola G, Martelli V, Sobrero AF. Adjuvant
tumor DNA sequencing assays with targeted large gene panels. chemotherapy for stage II colon cancer. Cancers 2020; 12: 2584.
PLoS One 2022; 17: e0266889. 29 Tie J, Cohen JD, Lo SN, et al. Prognostic significance of postsurgery
7 Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis circulating tumor DNA in nonmetastatic colorectal cancer:
detects minimal residual disease and predicts recurrence in patients individual patient pooled analysis of three cohort studies.
with stage II colon cancer. Sci Transl Med 2016; 8: 346ra92. Int J Cancer 2021; 148: 1014–26.
8 Tie J, Cohen JD, Wang Y, et al. Circulating tumor DNA analyses as 30 Tie J, Cohen JD, Lahouel K, et al. Circulating tumor DNA analysis
markers of recurrence risk and benefit of adjuvant therapy for guiding adjuvant therapy in stage II colon cancer. N Engl J Med
stage III colon cancer. JAMA Oncol 2019; 5: 1710–17. 2022; 386: 2261–72.
9 Reinert T, Henriksen TV, Christensen E, et al. Analysis of plasma 31 Bahadoer RR, Dijkstra EA, van Etten B, et al. Short-course
cell-free DNA by ultradeep sequencing in patients with stages I to radiotherapy followed by chemotherapy before total mesorectal
III colorectal cancer. JAMA Oncol 2019; 5: 1124–31. excision (TME) versus preoperative chemoradiotherapy, TME, and
optional adjuvant chemotherapy in locally advanced rectal cancer
10 Faulkner LG, Howells LM, Pepper C, Shaw JA, Thomas AL.
(RAPIDO): a randomised, open-label, phase 3 trial. Lancet Oncol
The utility of ctDNA in detecting minimal residual disease
2021; 22: 29–42.
following curative surgery in colorectal cancer: a systematic review
and meta-analysis. Br J Cancer 2023; 128: 297–309. 32 Conroy T, Bosset JF, Etienne PL, et al. Neoadjuvant chemotherapy
with FOLFIRINOX and preoperative chemoradiotherapy for
11 Murray DH, Symonds EL, Young GP, et al. Relationship between
patients with locally advanced rectal cancer (UNICANCER-
post-surgery detection of methylated circulating tumor DNA with
PRODIGE 23): a multicentre, randomised, open-label, phase 3 trial.
risk of residual disease and recurrence-free survival.
Lancet Oncol 2021; 22: 702–15.
J Cancer Res Clin Oncol 2018; 144: 1741–50.
33 Liu W, Li Y, Tang Y, et al. Response prediction and risk stratification
12 Tie J, Cohen JD, Wang Y, et al. Serial circulating tumour DNA
of patients with rectal cancer after neoadjuvant therapy through an
analysis during multimodality treatment of locally advanced rectal
analysis of circulating tumour DNA. EBioMedicine 2022; 78: 103945.
cancer: a prospective biomarker study. Gut 2019; 68: 663–71.

14 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2


Review

34 Vidal J, Casadevall D, Bellosillo B, et al. Clinical impact of 54 Bachet JB, Bouché O, Taieb J, et al. RAS mutation analysis in
presurgery circulating tumor DNA after total neoadjuvant treatment circulating tumor DNA from patients with metastatic colorectal
in locally advanced rectal cancer: a biomarker study from the cancer: the AGEO RASANC prospective multicenter study.
GEMCAD 1402 trial. Clin Cancer Res 2021; 27: 2890–98. Ann Oncol 2018; 29: 1211–19.
35 Wang Y, Yang L, Bao H, et al. Utility of ctDNA in predicting 55 Grasselli J, Elez E, Caratù G, et al. Concordance of blood- and
response to neoadjuvant chemoradiotherapy and prognosis tumor-based detection of RAS mutations to guide anti-EGFR
assessment in locally advanced rectal cancer: a prospective cohort therapy in metastatic colorectal cancer. Ann Oncol 2017;
study. PLoS Med 2021; 18: e1003741. 28: 1294–301.
36 Battaglin F, Naseem M, Lenz HJ, Salem ME. Microsatellite 56 Pinheiro M, Peixoto A, Rocha P, et al. KRAS and NRAS mutational
instability in colorectal cancer: overview of its clinical significance analysis in plasma ctDNA from patients with metastatic colorectal
and novel perspectives. Clin Adv Hematol Oncol 2018; 16: 735–45. cancer by real-time PCR and digital PCR. Int J Colorectal Dis 2022;
37 Ludford K, Ho WJ, Thomas JV, et al. Neoadjuvant pembrolizumab 37: 895–905.
in localized microsatellite instability high/deficient mismatch repair 57 Schmiegel W, Scott RJ, Dooley S, et al. Blood-based detection of
solid tumors. J Clin Oncol 2023; 41: 2181–90. RAS mutations to guide anti-EGFR therapy in colorectal cancer
38 Booth CM, Berry SR. Perioperative chemotherapy for resectable patients: concordance of results from circulating tumor DNA and
liver metastases in colorectal cancer: do we have a blind tissue-based RAS testing. Mol Oncol 2017; 11: 208–19.
spot? J Clin Oncol 2021; 39: 3767–69. 58 Vidal J, Muinelo L, Dalmases A, et al. Plasma ctDNA RAS mutation
39 Newhook TE, Overman MJ, Chun YS, et al. Prospective study of analysis for the diagnosis and treatment monitoring of metastatic
perioperative circulating tumor DNA dynamics in patients undergoing colorectal cancer patients. Ann Oncol 2017; 28: 1325–32.
hepatectomy for colorectal liver metastases. Ann Surg 2022; published 59 Maurel J, Alonso V, Escudero P, et al. Clinical impact of circulating
online July 6. https://doi.org/10.1097/SLA.0000000000005461. tumor RAS and BRAF mutation dynamics in patients with
40 Marmorino F, Prisciandaro M, Giordano M, et al. Circulating metastatic colorectal cancer treated with first-line chemotherapy
tumor DNA as a marker of minimal residual disease after radical plus anti-epidermal growth factor receptor therapy.
resection of colorectal liver metastases. JCO Precis Oncol 2022; JCO Precis Oncol 2019; 3: 1–16.
6: e2200244. 60 Normanno N, Esposito Abate R, Lambiase M, et al. RAS testing of
41 Bidard FC, Kiavue N, Ychou M, et al. Circulating tumor cells and liquid biopsy correlates with the outcome of metastatic colorectal
circulating tumor DNA detection in potentially resectable metastatic cancer patients treated with first-line FOLFIRI plus cetuximab in
colorectal cancer: a prospective ancillary study to the unicancer the CAPRI-GOIM trial. Ann Oncol 2018; 29: 112–18.
prodige-14 trial. Cells 2019; 8: 516. 61 Vidal J, Fernández-Rodríguez MC, Casadevall D, et al. Liquid
42 Overman MJ, Vauthey J-N, Aloia TA, et al. Circulating tumor DNA biopsy detects early molecular response and predicts benefit to
(ctDNA) utilizing a high-sensitivity panel to detect minimal residual first-line chemotherapy plus cetuximab in metastatic colorectal
disease post liver hepatectomy and predict disease recurrence. cancer: PLATFORM-B study. Clin Cancer Res 2023; 29: 379–88.
Proc Am Soc Clin Oncol 2017; 35 (suppl 15): 3522 62 Shitara K, Muro K, Watanabe J, et al. Negative hyperselection of
43 Tarazona N, Gimeno-Valiente F, Gambardella V, et al. Targeted next- patients with RAS wild-type metastatic colorectal cancer for
generation sequencing of circulating-tumor DNA for tracking panitumumab: a biomarker study of the phase III PARADIGM
minimal residual disease in localized colon cancer. Ann Oncol 2019; trial. Proc Am Soc Clin Oncol 2023; 41 (suppl 4): 11.
30: 1804–12. 63 Watanabe J, Muro K, Shitara K, et al. Panitumumab vs
44 Wang Y, Li L, Cohen JD, et al. Prognostic potential of circulating bevacizumab added to standard first-line chemotherapy and overall
tumor DNA measurement in postoperative surveillance of survival among patients with RAS wild-type, left-sided metastatic
nonmetastatic colorectal cancer. JAMA Oncol 2019; 5: 1118–23. colorectal cancer: a randomized clinical trial. JAMA 2023;
45 Henriksen TV, Tarazona N, Reinert T, et al. Circulating tumor 329: 1271–82.
DNA analysis for assessment of recurrence risk, benefit of 64 Strickler JH, Loree JM, Ahronian LG, et al. Genomic landscape of
adjuvant therapy, and early relapse detection after treatment in cell-free DNA in patients with colorectal cancer. Cancer Discov 2018;
colorectal cancer patients. Proc Am Soc Clin Oncol 2021; 8: 164–73.
39 (suppl 3): 11. 65 Kopetz S, Guthrie KA, Morris VK, et al. Randomized trial of
46 Thomsen CB, Hansen TF, Andersen RF, Lindebjerg J, Jensen LH, irinotecan and cetuximab with or without vemurafenib in
Jakobsen A. Early identification of treatment benefit by methylated BRAF-mutant metastatic colorectal cancer (SWOG S1406).
circulating tumor DNA in metastatic colorectal cancer. J Clin Oncol 2021; 39: 285–94.
Ther Adv Med Oncol 2020; 12: 1758835920918472. 66 Nakamura Y, Okamoto W, Kato T, et al. Circulating tumor
47 Bando H, Kagawa Y, Kato T, et al. A multicentre, prospective study DNA-guided treatment with pertuzumab plus trastuzumab for
of plasma circulating tumour DNA test for detecting RAS mutation HER2-amplified metastatic colorectal cancer: a phase 2 trial.
in patients with metastatic colorectal cancer. Br J Cancer 2019; Nat Med 2021; 27: 1899–903.
120: 982–86. 67 Siravegna G, Sartore-Bianchi A, Nagy RJ, et al. Plasma HER2
48 Bando H, Nakamura Y, Taniguchi H, et al. Effects of metastatic sites (ERBB2) copy number predicts response to HER2-targeted therapy
on circulating tumor DNA in patients with metastatic colorectal in metastatic colorectal cancer. Clin Cancer Res 2019; 25: 3046–53.
cancer. JCO Precis Oncol 2022; 6: e2100535. 68 Siena S, Raghav K, Masuishi T, et al. 386O Exploratory biomarker
49 Manca P, Corallo S, Lonardi S, et al. Variant allele frequency in analysis of DESTINY-CRC01, a phase II, multicenter, open-label
baseline circulating tumour DNA to measure tumour burden and study of trastuzumab deruxtecan (T-DXd, DS-8201) in patients (pts)
to stratify outcomes in patients with RAS wild-type metastatic with HER2-expressing metastatic colorectal cancer (mCRC).
colorectal cancer: a translational objective of the Valentino study. Ann Oncol 2021; 32: S532.
Br J Cancer 2022; 126: 449–55. 69 Clifton K, Rich TA, Parseghian C, et al. Identification of actionable
50 El Messaoudi S, Mouliere F, Du Manoir S, et al. Circulating DNA fusions as an anti-EGFR resistance mechanism using a circulating
as a strong multimarker prognostic tool for metastatic colorectal tumor DNA assay. JCO Precis Oncol 2019; 3: 3.
cancer patient management care. Clin Cancer Res 2016; 70 Willis J, Lefterova MI, Artyomenko A, et al. Validation of
22: 3067–77. microsatellite instability detection using a comprehensive plasma-
51 Parikh AR, Mojtahed A, Schneider JL, et al. Serial ctDNA based genotyping panel. Clin Cancer Res 2019; 25: 7035–45.
monitoring to predict response to systemic therapy in metastatic 71 Chen EX, Jonker DJ, Loree JM, et al. Effect of combined immune
gastrointestinal cancers. Clin Cancer Res 2020; 26: 1877–85. checkpoint inhibition vs best supportive care alone in patients with
52 Benson AB, Venook AP, Al-Hawary MM, et al. Colon cancer, advanced colorectal cancer: the Canadian Cancer Trials Group
version 2·2021, NCCN clinical practice guidelines in oncology. CO.26 study. JAMA Oncol 2020; 6: 831–38.
J Natl Compr Canc Netw 2021; 19: 329–59. 72 Crisafulli G, Sartore-Bianchi A, Lazzari L, et al. Temozolomide
53 Cervantes A, Adam R, Roselló S, et al. Metastatic colorectal cancer: treatment alters mismatch repair and boosts mutational burden in
ESMO clinical practice guideline for diagnosis, treatment and tumor and blood of colorectal cancer patients. Cancer Discov 2022;
follow-up. Ann Oncol 2023; 34: 10–32. 12: 1656–75.

www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2 15


Review

73 Tie J, Kinde I, Wang Y, et al. Circulating tumor DNA as an early 93 Parseghian CM, Sun R, Woods M, et al. Resistance mechanisms to
marker of therapeutic response in patients with metastatic anti-epidermal growth factor receptor therapy in RAS/RAF wild-
colorectal cancer. Ann Oncol 2015; 26: 1715–22. type colorectal cancer vary by regimen and line of therapy.
74 Max Ma X, Bendell JC, Hurwitz HI, et al. Disease monitoring using J Clin Oncol 2023; 41: 460–71.
post-induction circulating tumor DNA analysis following first-line 94 Raghav K, Ou F-S, Venook AP, et al. Acquired genomic alterations
therapy in patients with metastatic colorectal cancer. Clin Cancer Res on first-line chemotherapy with cetuximab in advanced colorectal
2020; 26: 4010–17. cancer: circulating tumor DNA analysis of the CALGB/
75 Kim S, Lim Y, Kang J-K, et al. Dynamic changes in longitudinal SWOG-80405 trial (alliance). J Clin Oncol 2023; 41: 472–78.
circulating tumour DNA profile during metastatic colorectal cancer 95 Cremolini C, Rossini D, Dell’Aquila E, et al. Rechallenge for
treatment. Br J Cancer 2022; 127: 898–907. patients with RAS and BRAF wild-type metastatic colorectal cancer
76 van ’t Erve I, Medina JE, Leal A, et al. Metastatic colorectal cancer with acquired resistance to first-line cetuximab and irinotecan:
treatment response evaluation by ultra-deep sequencing of cell-free a phase 2 single-arm clinical trial. JAMA Oncol 2019; 5: 343–50.
DNA and matched white blood cells. Clin Cancer Res 2022; 96 Martinelli E, Martini G, Famiglietti V, et al. Cetuximab rechallenge
29: 899–909. plus avelumab in pretreated patients with RAS wild-type metastatic
77 Lueong SS, Herbst A, Liffers S-T, et al. Serial circulating tumor colorectal cancer: the phase 2 single-arm clinical CAVE trial.
DNA mutational status in patients with KRAS-mutant metastatic JAMA Oncol 2021; 7: 1529–35.
colorectal cancer from the phase 3 AIO KRK0207 trial. Clin Chem 97 Khan KH, Cunningham D, Werner B, et al. Longitudinal liquid
2020; 66: 1510–20. biopsy and mathematical modeling of clonal evolution forecast time
78 Lyskjær I, Kronborg CS, Rasmussen MH, et al. Correlation between to treatment failure in the PROSPECT-C phase II colorectal cancer
early dynamics in circulating tumour DNA and outcome from clinical trial. Cancer Discov 2018; 8: 1270–85.
FOLFIRI treatment in metastatic colorectal cancer. Sci Rep 2019; 98 Parseghian CM, Loree JM, Morris VK, et al. Anti-EGFR-resistant
9: 11542. clones decay exponentially after progression: implications for anti-
79 Garlan F, Laurent-Puig P, Sefrioui D, et al. Early evaluation of EGFR re-challenge. Ann Oncol 2019; 30: 243–49.
circulating tumor DNA as marker of therapeutic efficacy in 99 Hazar-Rethinam M, Kleyman M, Han GC, et al. Convergent
metastatic colorectal cancer patients (PLACOL study). therapeutic strategies to overcome the heterogeneity of acquired
Clin Cancer Res 2017; 23: 5416–25. resistance in BRAFV600E colorectal cancer. Cancer Discov 2018;
80 Osumi H, Shinozaki E, Yamaguchi K, Zembutsu H. Early change in 8: 417–27.
circulating tumor DNA as a potential predictor of response to 100 Tan L, Tran B, Tie J, et al. Phase Ib/II trial of BRAF and EGFR
chemotherapy in patients with metastatic colorectal cancer. Sci Rep inhibition in BRAFV600E metastatic colorectal cancer and other
2019; 9: 17358. cancers: EVICT (erlotinib and vemurafenib in combination trial).
81 Diaz LA Jr, Williams RT, Wu J, et al. The molecular evolution of Clin Cancer Res 2023; 29: 1017-30
acquired resistance to targeted EGFR blockade in colorectal cancers. 101 Kopetz S, Murphy DA, Pu J, et al. 316O—Genomic mechanisms of
Nature 2012; 486: 537–40. acquired resistance of patients (pts) with BRAF V600E-mutant (mt)
82 Misale S, Yaeger R, Hobor S, et al. Emergence of KRAS mutations metastatic colorectal cancer (mCRC) treated in the BEACON study.
and acquired resistance to anti-EGFR therapy in colorectal cancer. Ann Oncol 2022; 33: 33.
Nature 2012; 486: 532–36. 102 Siravegna G, Lazzari L, Crisafulli G, et al. Radiologic and genomic
83 Bardelli A, Corso S, Bertotti A, et al. Amplification of the MET evolution of individual metastases during HER2 blockade in
receptor drives resistance to anti-EGFR therapies in colorectal colorectal cancer. Cancer Cell 2018; 34: 148–162.
cancer. Cancer Discov 2013; 3: 658–73. 103 Li BT, Velcheti V, Price TJ, et al. Largest evaluation of acquired
84 Arena S, Bellosillo B, Siravegna G, et al. Emergence of multiple resistance to sotorasib in KRAS p.G12C-mutated non–small cell
EGFR extracellular mutations during cetuximab treatment in lung cancer (NSCLC) and colorectal cancer (CRC): plasma
colorectal cancer. Clin Cancer Res 2015; 21: 2157–66. biomarker analysis of CodeBreaK100. Proc Am Soc Clin Oncol 2022;
85 Yonesaka K, Zejnullahu K, Okamoto I, et al. Activation of ERBB2 40 (suppl 16): 102.
signaling causes resistance to the EGFR-directed therapeutic 104 Awad MM, Liu S, Rybkin II, et al. Acquired resistance to KRASG12C
antibody cetuximab. Sci Transl Med 2011; 3: 99ra86. inhibition in cancer. N Engl J Med 2021; 384: 2382–93.
86 Peeters M, Price T, Boedigheimer M, et al. Evaluation of emergent 105 Pascual J, Attard G, Bidard FC, et al. ESMO recommendations on
mutations in circulating cell-free DNA and clinical outcomes in the use of circulating tumour DNA assays for patients with cancer:
patients with metastatic colorectal cancer treated with a report from the ESMO precision medicine working group.
panitumumab in the ASPECCT study. Clin Cancer Res 2019; Ann Oncol 2022; 33: 750–68.
25: 1216–25. 106 Dasari A, Morris VK, Allegra CJ, et al. ctDNA applications and
87 Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and integration in colorectal cancer: an NCI colon and rectal-anal task
resistance to EGFR blockade in the blood of colorectal cancer forces whitepaper. Nat Rev Clin Oncol 2020; 17: 757–70.
patients. Nat Med 2015; 21: 795–801. 107 Parikh AR, Leshchiner I, Elagina L, et al. Liquid versus tissue
88 Montagut C, Argilés G, Ciardiello F, et al. Efficacy of sym004 in biopsy for detecting acquired resistance and tumor heterogeneity in
patients with metastatic colorectal cancer with acquired resistance gastrointestinal cancers. Nat Med 2019; 25: 1415–21.
to anti-EGFR therapy and molecularly selected by circulating tumor 108 Ma M, Zhu H, Zhang C, Sun X, Gao X, Chen G. “Liquid biopsy”-
DNA analyses: a phase 2 randomized clinical trial. JAMA Oncology ctDNA detection with great potential and challenges.
2018; 4: e175245-e. Ann Transl Med 2015; 3: 235.
89 Van Emburgh BO, Arena S, Siravegna G, et al. Acquired RAS or 109 Xue J, Prabhakaran S, Prabhakaran S, et al. The utility of ctDNA in
EGFR mutations and duration of response to EGFR blockade in colorectal cancer with peritoneal metastases. ANZ J Surg 2023;
colorectal cancer. Nat Commun 2016; 7: 13665. 93: 506-09.
90 Montagut C, Dalmases A, Bellosillo B, et al. Identification of a 110 Tie J, Cohen J, Lahouel K, et al. 318MO—Circulating tumour DNA
mutation in the extracellular domain of the epidermal growth factor (ctDNA) dynamics, CEA and sites of recurrence for the randomised
receptor conferring cetuximab resistance in colorectal cancer. DYNAMIC study: adjuvant chemotherapy (ACT) guided by ctDNA
Nat Med 2012; 18: 221–23. analysis in stage II colon cancer (CC). Ann Oncol 2022; 33: S136–96.
91 Sartore-Bianchi A, Pietrantonio F, Lonardi S, et al. Circulating 111 Rehman AU, Khan P, Maurya SK, et al. Liquid biopsies to occult
tumor DNA to guide rechallenge with panitumumab in metastatic brain metastasis. Mol Cancer 2022; 21: 113.
colorectal cancer: the phase 2 CHRONOS trial. Nat Med 2022; 112 To YH, Degeling K, Kosmider S, et al. Circulating tumour DNA as a
28: 1612–18. potential cost-effective biomarker to reduce adjuvant chemotherapy
92 Vivas CS, Salva F, Fernández-Rodríguez C, et al. P-118 cetuximab overtreatment in stage II colorectal cancer. PharmacoEconomics
rechallenge in RAS, BRAF, EGFR-ECD wild type metastatic 2021; 39: 953–64.
colorectal cancer (mCRC) patients treated with anti-EGFR therapies
in first line: the CITRIC study. Ann Oncol 2022; 33: S291. © 2023 Published by Elsevier Ltd. All rights reserved.

16 www.thelancet.com/gastrohep Published online July 24, 2023 https://doi.org/10.1016/S2468-1253(23)00146-2

You might also like