Circulating Tumor DNA and Radiological Tumor Volum

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

ORIGINAL ARTICLE

Circulating tumor DNA and radiological tumor volume identify patients at


risk for relapse with resected, early-stage non-small-cell lung cancer
H. T. Tran1y , S. Heeke1y , S. Sujit2 , N. Vokes1 , J. Zhang1 , M. Aminu2 , V. K. Lam3 , A. Vaporciyan4 , S. G. Swisher4,
M. C. B. Godoy5, T. Cascone1 , B. Sepesi1 , D. L. Gibbons1 , J. Wu1,2z & J. V. Heymach1z
Departments of 1Thoracic Head & Neck Medical Oncology; 2Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston; 3Sidney Kimmel
Comprehensive Cancer Center, Johns Hopkins, Baltimore; Departments of 4Thoracic and Cardiovascular Surgery; 5Thoracic Imaging, The University of Texas MD
Anderson Cancer Center, Houston, USA

Available online 21 November 2023

Background: Predicting relapse and overall survival (OS) in early-stage non-small-cell lung cancer (NSCLC) patients
remains challenging. Therefore, we hypothesized that detection of circulating tumor DNA (ctDNA) can identify
patients with increased risk of relapse and that integrating radiological tumor volume measurement along with
ctDNA detectability improves prediction of outcome.
Patients and methods: We analyzed 366 serial plasma samples from 85 patients who underwent surgical resections and
assessed ctDNA using a next-generation sequencing liquid biopsy assay, and measured tumor volume using a computed
tomography-based three-dimensional annotation.
Results: Our results showed that patients with detectable ctDNA at baseline or after treatment and patients who did
not clear ctDNA after treatment had a significantly worse clinical outcome. Integrating radiological analysis allowed the
stratification in risk groups prognostic of clinical outcome as confirmed in an independent cohort of 32 patients.
Conclusions: Our findings suggest ctDNA and radiological monitoring could be valuable tools for guiding follow-up care
and treatment decisions for early-stage NSCLC patients.
Key words: MRD, early-stage NSCLC, ctDNA, tumor volume, liquid biopsy

INTRODUCTION relapse in multiple tumor entities, including colorectal


In contrast to the dismal prognosis of advanced-stage non- cancer3,4 and lung cancer.5-7
small-cell lung cancer (NSCLC), the 5-year overall survival In those studies, however, ctDNA was assessed using a
(OS) rate of early-stage NSCLC (I-IIIa) is 33%-60% with sur- customized or ‘bespoke’ assay informed by genomic
gery with curative intent.1 Recent studies indicate that the profiling of tumor. In contrast, tissue-agnostic assays are the
introduction of immunotherapy, given either as neo- same, independent of the patient, and might thus present
adjuvant chemoimmunotherapy or as monotherapy in the advantages for implementation in routine and timely clinical
adjuvant setting, has improved survival outcomes, but care, particularly when whole exome sequencing is not
relapse still occurs frequently. Thus, predicting the risk of available from the tumor.
relapse after standard-of-care (SOC) treatment remains a Interestingly, while ctDNA shedding is associated with
major challenge for these patients. tumor volume,8 we recently highlighted that in patients
Because relapse has been associated with the presence with advanced NSCLC, ctDNA levels also vary depending on
of minimal residual disease (MRD),2 many studies have the tumor genotype and location.9 Furthermore, some pa-
evaluated circulating tumor DNA (ctDNA) monitoring in tients with no detectable ctDNA ultimately relapse post-
plasma as a method to detect MRD and predict early operatively, presumably because their tumors shed lower
or undetectable levels of DNA into the circulation. Conse-
quently, we hypothesized that integrating ctDNA analysis
*Correspondence to: Dr John V. Heymach, Departments of Thoracic/Head and
Neck Medical Oncology and Cancer Biology, University of Texas MD Anderson
with radiologically measured tumor volume may also
Cancer Center, Unit 432, Houston, Texas 77230, USA. Tel: þ01-713-792-6363 improve prediction of relapse.
E-mail: jheymach@mdanderson.org (J. V. Heymach). We therefore retrospectively analyzed a cohort of 85 pa-
y
Contributed equally. tients with early-stage NSCLC who underwent plasma sam-
z
Co-senior authors. pling before surgery and shortly after surgery as well as
0923-7534/© 2023 The Author(s). Published by Elsevier Ltd on behalf of
European Society for Medical Oncology. This is an open access article under the
longitudinal treatment samples collected during follow-up to
CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). detect MRD and predict early relapse using a blood-based,

Volume 35 - Issue 2 - 2024 https://doi.org/10.1016/j.annonc.2023.11.008 183


Annals of Oncology H. T. Tran et al.

tissue-agnostic assay for robust ctDNA detection. We further TP53, and others) and a subset of w450 kb of epigenomic
integrated this analysis with radiological tumor volume regions that undergo differential methylation in lung cancer.
assessed before surgery to highlight how integrating ctDNA Extracted cfDNA fragments were partitioned based on the
and radiological data can improve prediction of relapse-free level of methylation, uniquely barcoded, and then recom-
survival (RFS) and OS in this clinical setting and confirmed bined for library preparation and next-generation sequencing
our results in an independent cohort of 32 NSCLC patients. (NovaSeq 600 System, Illumina, San Diego, CA). Sequencing
reads are analyzed using a proprietary bioinformatics algo-
PATIENTS AND METHODS rithm software trained on cancer and cancer-free control
From April 2016 to August 2018, 117 patients with early- samples for the binary detection of ctDNA (ctDNA detected
stage NSCLC were consented and enrolled in the Immuno- versus ctDNA not detected). The software algorithm iden-
genomic Profiling of NoneSmall Cell Lung Cancer protocol tifies genomic mutations (single-nucleotide variants and
(ICON) study that was approved by The University of Texas Indels, aligned to the reference human genome, hg19) and
MD Anderson Cancer Center Institution Review Board and filters only for mutations likely to be tumor derived (i.e. fil-
in accordance with the Code of Ethics of the World Medical ters germline variants as well as variants associated with
Association. All patients in this study received SOC therapies clonal hematopoiesis) based on a pre-defined threshold. The
for early-stage NSCLC (Table 1). bioinformatics algorithm simultaneously determines the
presence of tumor-derived differential methylation by eval-
uating the distribution of cfDNA molecules across the
Blood collection
methylation partitions. A statistical likelihood that the
Blood was collected in cell-free DNA (cfDNA) blood collec- observed patterns across all regions in a sample are likely to
tion tube tubes (Streck Corp, La Vista, NE) at the following be tumor derived is generated, and the samples must pass a
time points: before surgery and 4 weeks, 7 months, 10 pre-defined threshold to result in a ‘methylation-detected’
months, and 13 months after surgery. Blood samples were call. Detection of ctDNA was defined by the presence of a
processed within 24 h after collection and cell-free plasma somatic mutation or by the detection of a methylation
were frozen at 70 C until analysis. All samples were de- signal. This prototype assay has undergone partial analytic
identified using unique sample identification assignment validation with >90% specificity among 74 self-declared
by our institution’s biospecimen inventory system. Across cancer-free controls (96.6% for epigenomic caller, 95.9%-
both cohorts, 561 samples have been analyzed. 100% for genomic caller depending on mutation type,
combined caller 93.2%) and 100% detection at the lowest
ctDNA detection dilution limit tested in the limit of detection study of 0.3%
For cohort 1, cfDNA was extracted from 366 banked plasma mutant allele fraction.
samples (median volume: 5.9 ml; range 2-10 ml) and Additionally, 195 samples from 32 patients in cohort 2
analyzed with a 500-kb plasma-only assay prototype (Lunar have been analyzed using an earlier iteration of the assay
version 1.2, Guardant Health, Redwood City, CA) that eval- that omits the inclusion of DNA methylation (Lunar version
uates tumor-derived genomic and methylation signals from 1.1, Guardant Health).
an w500-kb panel without the need for separate tumor
tissue or peripheral blood mononuclear cell analysis. Briefly,
extracted cfDNA is subjected to automated methylation Radiological tumor volume assessment
partitioning followed by unique molecular barcoding for The tumor was contoured on computed tomography (CT)
single-molecule tracking. PCR-amplified libraries are then with the guidance of [18F]2-fluoro-2-deoxy-D-glucose posi-
enriched for the panel covering common truncal oncogenic tron emission tomography (18F-FDG PET) using the MIM
mutations (w45 kb, 37 genes including ALK, BRAF, EGFR, software (MIM Software Inc, Cleveland, OH, USA) by a
ERBB2, KRAS, MET, NF1, NRAS, PIK3CA, PTEN RB1, STK11, thoracic radiologist. The baseline CT and PET Digital Imaging

Table 1. Patient characteristics

Cohort 1 (n [ 85) Cohort 1 with recurrence (n [ 33; 39%) Cohort 2 (n [ 32)


Age Median (range) 69 (38-85) 68 (38-85) 65 (44-85)
Gender Female (%) 41 (48%) 15 (45%) 20 (63%)
Male (%) 44 (52%) 18 (55%) 12 (38%)
Histology Adenocarcinoma 58 (68%) 22 (67%) 25 (78%)
Squamous cell carcinoma 21 (25%) 7 (21%) 5 (16%)
other 6 (7%) 4 (12%) 2 (6%)
Clinical stage I 41 (48%) 10 (30%) 16 (50%)
II 29 (34%) 13 (39%) 12 (38%)
III 15 (18%) 10 (30%) 4 (13%)
Other therapies Neoadjuvant chemotherapies 24 (28%) 14 (42%) 6 (19%)
Adjuvant chemotherapies 23 (27%) 9 (27%) 13 (41%)
Post-operative radiation therapies 16 (19%) 10 (30%) 6 (19%)
Follow-up (months) Median (range) 51 (2-73) 43 (2-73) 65 (5-74)

184 https://doi.org/10.1016/j.annonc.2023.11.008 Volume 35 - Issue 2 - 2024


H. T. Tran et al. Annals of Oncology

and Communications in Medicine images were curated neoadjuvant therapies, respectively (Supplementary
from picture archiving and communication system. The Figure S1A, available at https://doi.org/10.1016/j.annonc.
Digital Imaging and Communications in Medicine scans 2023.11.008). Before surgery, ctDNA was detected in 18/
were converted to Neuroimaging Informatics Technology 24 (75%) patients who received neoadjuvant therapies and
Initiative by dcm2nii with the use of the MRIcron tool 30/56 (54%) patients who did not receive neoadjuvant
(www.nitrc.org). The three-dimensional (3D) manual con- therapies (Supplementary Figure S1A and B, available at
tours were computationally extracted for subsequent ana- https://doi.org/10.1016/j.annonc.2023.11.008). Detection
lyses. The visual quality control of segmentations was done rates in squamous cell carcinoma were highest (87%), fol-
by superimposing tumor on imaging using ITK-SNA (version lowed by other histologies (67%) and adenocarcinoma
4.0, www.itksnap.org).10,11 Images closest to surgery date (39%; Supplementary Figure S1C, available at https://doi.
were used, with a mean time of images before surgery of 59 org/10.1016/j.annonc.2023.11.008). In patients with stage
days (range: 3-183 days before surgery). For the partial-solid I disease, detection of ctDNA was 42% for adenocarcinoma
tumors, ground-glass components and solid components of (n ¼ 11/26) and 78% for squamous cell carcinoma (n ¼ 7/9;
the tumor were delineated separately by using the lung Supplementary Figure S2A, available at https://doi.org/10.
window on CT. The overall tumor volume is computed as 1016/j.annonc.2023.11.008). Clinical stage and lung cancer
the aggregate of the volumes associated with both the GGN histologies were balanced in this cohort (Supplementary
and solid nodule regions. Figure S2B and C, available at https://doi.org/10.1016/j.
annonc.2023.11.008) and no differences between geno-
Data analysis types on the detection of ctDNA were observed
(Supplementary Figure S3, available at https://doi.org/10.
Data were analyzed using R (v4.2.1). Survival including log-
1016/j.annonc.2023.11.008). Additionally, ctDNA detection
rank test and Cox proportional hazard ratio (HR) was
rates increased with histological grading (G1: 38%; G2: 52%,
calculated using the R packages survival (v3.4) and surv-
and G3þ: 85%; Supplementary Figure S4A, available at
miner (v0.4.9). Receiver operator characteristics was
https://doi.org/10.1016/j.annonc.2023.11.008). Further-
assessed using pROC (v1.18) by associating the presence of
more, lung adenocarcinoma histological subtypes demon-
ctDNA at baseline before surgery with the tumor volume as
strated equal detection rates for acinar versus non-acinar
assessed by radiographic 3D reconstruction. Cut-offs for
subtypes (both 44%; Supplementary Figure S4B, available at
optimal stratification of tumor volume were calculated
https://doi.org/10.1016/j.annonc.2023.11.008). Lastly,
based on Youden’s J and the closest distance to the top left
detection of ctDNA at baseline before surgery did not differ
corner using pROC (Supplementary Table S1, available at
between patients with extrapulmonary versus pulmonary
https://doi.org/10.1016/j.annonc.2023.11.008). Differences
relapse (67% versus 64%; Supplementary Figure S4C,
in RFS and OS from the time of surgery were analyzed using
available at https://doi.org/10.1016/j.annonc.2023.11.008).
Cox proportional hazards models and KaplaneMeier anal-
Importantly, the presence of ctDNA before surgery was
ysis. Patients were further stratified by ctDNA clearance,
associated with a numerically shorter RFS (HR 2.14, 95% CI
which was defined by a pre-surgery (baseline) ctDNA-
0.95-4.81, P ¼ 0.06) and a statistically significant decrease
positive test result followed by a ctDNA-negative test
in OS (HR 3.29, 95% CI 0.95-11.45, P ¼ 0.047) (Figure 1A
result at any subsequent time point after completion of SOC
and B), suggesting detectability of ctDNA before surgery
therapy. For the analysis of cohort 2 in the combination of
may have prognostic value.
ctDNA plus radiological tumor volume, Cox proportional HR
with Firth’s penalized likelihood has been used for estima-
tion of HR and confidence intervals (CIs). ctDNA detected after standard-of-care therapy is
associated with worse outcome
RESULTS We next investigated whether the detection of ctDNA after
definitive SOC therapy, defined as surgery and neoadjuvant
Patient characteristics or adjuvant chemotherapy, and/or post-operative radio-
Patient characteristics are summarized in Table 1. Among therapy (PORT) if administered, was also associated with
the 85 included patients, 58 (68%) had adenocarcinoma, risk of relapse and OS. We observed that after SOC therapy,
and 41 (48%) were diagnosed with stage I disease. Twenty- the presence of detectable ctDNA was associated with
eight percent of patients received neoadjuvant therapies, statistically significant decreases in RFS (HR2.80, 95% CI
and 27% received post-surgical adjuvant therapies. With a 1.23-6.37, P ¼ 0.01) and OS (HR3.99, 95% CI 1.40-11.40,
median follow-up of 4.3 years (range: 0.2-6.1 years) for this P ¼ 0.0053) compared to patients without detectable
cohort, 39% of patients had a relapse of disease. ctDNA after SOC therapy (Figure 1C and D).

Detection of ctDNA before surgery ctDNA clearance defines patient with lower risk or
Before surgery, 80/85 samples were analyzed successfully, recurrence
and ctDNA was detected in 48 (60%) of these samples We observed that some patients ‘cleared’ ctDNA after SOC
overall, including 49%, 58%, and 100% of patients with therapy, defined by a positive ctDNA detection at baseline
stage I, II, and III carcinoma that did not receive followed by a negative ctDNA sample during follow-up after

Volume 35 - Issue 2 - 2024 https://doi.org/10.1016/j.annonc.2023.11.008 185


Annals of Oncology H. T. Tran et al.

A RFS - ctDNA detection before to surgery B OS - ctDNA detection before to surgery


1.00 1.00
Median RFS P = 0.06 + + + + Median OS P = 0.047
+ +++++++++++++ ++
++++++ +
+ + + + HR = 2.14 (0.95-4.81) ++ HR = 3.29 (0.95-11.45)
0.75 + ++++ +++++++ + ++++ + 0.75 +++
+++ +++

Survival probability
++++ +++++++++++++ +
RFS probability

+ +++
+++
++++ +++
++
0.50 +++++++ 0.50

0.25 0.25

0.00
0 20 40
+ no ctDNA
60
+ ctDNA detected
80
0.00
0 20 40
+ no ctDNA
60
+ ctDNA detected
80
ctDNA detected 48 32 28 12 0 48 45 36 14 0
no ctDNA 32 27 23 7 0 32 31 27 9 0
0 20 40 60 80 0 20 40 60 80
Months Months

C RFS - ctDNA detection after SOC D OS - ctDNA detection after SOC


1.00 1.00
Median RFS P = 0.01 + + ++ + Median OS P = 0.0053
+++++
+++++++++++++ + ++++++++ +++ +
+ + HR = 2.80 (1.23-6.37) HR = 3.99 (1.40-11.40)
0.75 + + 0.75

Survival probability
+++++
+++ ++ +++++ + ++++
RFS probability

+++ ++ +
0.50 0.50 ++ + +++ +
++ + + +
0.25 0.25

0.00
0 20 40
+ no ctDNA
60
+ ctDNA detected
80
0.00
0 20 40
+ no ctDNA
60
+ ctDNA detected
80
ctDNA detected 14 7 6 2 0 ctDNA detected 14 14 9 3 0
no ctDNA 50 42 34 11 0 no ctDNA 50 48 41 12 0
0 20 40 60 80 0 20 40 60 80
Months Months

Figure 1. Detection of ctDNA before surgery and/or after completion of standard of care (SOC) therapies are associated with worsen clinical outcomes. (A) RFS and
(B) OS based on ctDNA detection before surgery. (C) RFS and (D) OS based on ctDNA detection after SOC. RFS was calculated from time of surgery to relapse, death
or censoring and OS was calculated from time of surgery to death or censoring. Log-rank tests were used to compute P values, and Cox proportional HRs were
calculated in comparison to patients with detectable ctDNA.
ctDNA, circulating tumor DNA; HR, hazard ratio; OS, overall survival; RFS, relapse-free survival; SOC, standard-of-care.

SOC therapy. We hypothesized that ctDNA clearance at any with risk of RFS and OS, there remained a substantial
point during treatment would be associated with better number of patients with non-detectable ctDNA that
prognosis. Indeed, patients who had detectable ctDNA recurred, as well as ctDNA-positive patients who did not
before treatment and cleared ctDNA after completion of recur. Given the established association between tumor size
SOC treatment had outcomes that were not significantly and recurrence,10 we investigated whether the integration
different than patients with undetectable ctDNA of radiological tumor measurement with ctDNA could
throughout the study for RFS (HR 1.5, 95% CI 0.49-4.7, P ¼ improve prediction of RFS and OS. Radiological tumor vol-
0.478) and OS (HR 4.6, 95% CI 0.48-44, P ¼ 0.186). Patients ume was larger in patients with detectable ctDNA at
with persistent ctDNA, by contrast, had significantly shorter baseline (P ¼ 0.009; Supplementary Figure S7A, available at
RFS and OS compared to patients with undetectable ctDNA https://doi.org/10.1016/j.annonc.2023.11.008) and was
throughout the study [RFS HR persistent compared to never associated with ctDNA detection (area under the receiving
detected ¼ 4.0 (1.46-10.8), P ¼ 0.007; OS HR ¼ 16.5 (2.09- operating characteristic curve ¼ 0.682; Supplementary
131), P ¼ 0.008; Figure 2; Supplementary Figure S5, avail- Figure S7B, available at https://doi.org/10.1016/j.annonc.
able at https://doi.org/10.1016/j.annonc.2023.11.008]. We 2023.11.008). We used Youden’s J to calculate the optimal
furthermore observed a trend toward shorter RFS (HR 2.50, cut-off of tumor volume for the detection of ctDNA in our
95% CI 0.91-6.87, P ¼ 0.075) and OS (HR 3.51, 95% CI 0.95- cohort (Supplementary Table S1, available at https://doi.
13.0, P ¼ 0.061) for persistent ctDNA compared to patients org/10.1016/j.annonc.2023.11.008) to further stratify pa-
who cleared ctDNA (Supplementary Figure S5, available at tients into small-tumor-volume and large-tumor-volume
https://doi.org/10.1016/j.annonc.2023.11.008). Clearance groups and created three risk groups: low risk for patients
for patients with neoadjuvant and those without neo- with both small tumor volume and undetectable ctDNA at
adjuvant therapies are highlighted in Supplementary baseline before surgery; medium risk for patients who
Figure S6, available at https://doi.org/10.1016/j.annonc. either have larger tumor volume or detectable ctDNA at
2023.11.008. Median time to clearance was 20 days baseline before surgery; and high risk for patients with
(range: 2-181 days). both, larger tumor volume and detectable ctDNA at base-
line before surgery (Supplementary Figure S8, available at
https://doi.org/10.1016/j.annonc.2023.11.008). In order to
Combination of ctDNA detection and radiological tumor confirm the results in an independent cohort, we included a
volume assessment predicts clinical outcome second cohort (cohort 2) of 32 patients that have been
While these data provided evidence that ctDNA levels, analyzed with an earlier iteration of the assay. Patients in
particularly after definitive SOC therapy, were associated the high-risk group had a significantly higher risk of relapse

186 https://doi.org/10.1016/j.annonc.2023.11.008 Volume 35 - Issue 2 - 2024


H. T. Tran et al. Annals of Oncology

A RFS - ctDNA clearance B OS - ctDNA clearance


1.00 1.00 + + ++
+ +++++++++ +++++
++ +
Median RFS P = 0.0094 ++ Median OS P = 0.00076
+ +
++ ++
+ + ++ + + +++
0.75 0.75

Survival probability
+ ++ +
++ ++ ++++ + ++++++ +
RFS probability

0.50
+++ 0.50 +
+++ ++ +
+
+++ +
0.25 0.25

0.00 + Persistent + Cleared + Never Detected 0.00 + Persistent + Cleared + Never Detected
0 20 40 60 80 0 20 40 60 80
Persistent 16 8 6 1 0 Persistent 16 16 9 2 0
Cleared 17 13 11 5 0 Cleared 17 15 13 4 0
Never Detected 23 20 16 6 0 Never Detected 23 23 20 8 0
0 20 40 60 80 0 20 40 60 80
Months Months

Figure 2. Failure to clear ctDNA after completion of SOC therapies are associated with greater risks for disease recurrence and shortened overall survival. (A) RFS
and (B) OS in patients without detectable ctDNA throughout the study (Never Detected), patients who cleared ctDNA during treatment (Cleared), and patients
with persistent ctDNA detectable (Persistent). Log-rank tests were used to compute P values.
ctDNA, circulating tumor DNA; OS, overall survival; RFS, relapse-free survival.

compared to patients in the low-risk group (Nlow risk ¼ 19, 1016/j.annonc.2023.11.008), highlighting a robust predic-
Nhigh risk ¼ 32; HR 3.0, 95% CI 1.0-9.1, P ¼ 0.049) but not tion of relapse and survival in two independent cohorts.
patients in the medium-risk group who demonstrated Prediction of relapse was maintained after refining the
comparable outcome (Nmedium risk ¼ 20; HR 1.5, 95% CI radiological tumor analysis to the solid component only
0.43-5.4, P ¼ 0.518) in cohort 1, as confirmed in cohort 2 (Supplementary Figure S9, available at https://doi.org/10.
(Nlow risk ¼ 7, Nmedium risk ¼ 18, Nhigh risk ¼ 7; Figure 3A and 1016/j.annonc.2023.11.008; Supplementary Table S3,
B; Supplementary Table S2, available at https://doi.org/10. available at https://doi.org/10.1016/j.annonc.2023.11.008).
1016/j.annonc.2023.11.008). Likewise, combining ctDNA
and radiology demonstrated statistically significant differ- DISCUSSION
ences in OS in both cohorts (Figure 3C and D; While detection of ctDNA in longitudinal analysis is clearly
Supplementary Table S2, available at https://doi.org/10. able to track tumor evolution and recurrence,12 uncertainty

A RFS - combining ctDNA with radiology (cohort 1) B RFS - combining ctDNA with radiology (cohort 2)
1.00 1.00 ++++ ++
+ + Median RFS P = 0.072 Median RFS P = 0.00067

0.75 + + ++ ++++ ++ ++++ + 0.75


++ ++ + +
RFS probability
RFS probability

+ +++
++
0.50 + ++++ 0.50
+ +++++ ++

0.25 0.25

+
0.00 + high + medium + low 0.00
0 20 40 60 0 20 40 60 80
high 32 20 17 8 high 7 1 1 1 0
medium 20 16 14 4 medium 18 11 11 10 0
low 19 17 15 4 low 7 7 7 7 0
0 20 40 60 0 20 40 60 80
Months Months

C OS - combining ctDNA with radiology (cohort 1) D OS - combining ctDNA with radiology (cohort 2)
1.00 + + 1.00 ++++ ++
++ + ++++++++++ + +++
+ +++++++ ++ ++++Median OS P = 0.033 Median OS P = 0.0015

0.75
++ 0.75
Survival probability

+++
++ ++
RFS probability

+ +++++ ++
++ + +++++ ++++
0.50 0.50
++

0.25 0.25
++

0.00 + high + medium + low 0.00


0 20 40 60 80 0 20 40 60 80
high 32 30 22 9 0 high 7 2 2 2 0
medium 20 19 18 6 0 medium 18 17 15 14 0
low 19 19 17 5 0 low 7 7 7 7 0
0 20 40 60 80 0 20 40 60 80
Months Months

Figure 3. Validation of a risk model with combination of ctDNA and radiologic tumor volume. (A) RFS in patients with detectable ctDNA and radiological tumor
volume above the computed threshold (high risk), patients with undetectable ctDNA and radiological tumor volume below the computed threshold (low risk),
patients with detectable ctDNA but tumor volume below the calculated threshold, or patients with undetectable ctDNA but tumor volume above the calculated
threshold (medium risk) as well as in (B) an independent cohort (cohort 2) of 32 patients. (C) OS for cohort 1 as well as (D) in the independent cohort (cohort 2).
Log-rank tests were used to compute P values.
ctDNA, circulating tumor DNA; OS, overall survival; RFS, relapse-free survival.

Volume 35 - Issue 2 - 2024 https://doi.org/10.1016/j.annonc.2023.11.008 187


Annals of Oncology H. T. Tran et al.

persists in the appropriate time point for ctDNA detec- ctDNA has been proven useful to predict worse clinical
tion.13 In line with previously published data,14,15 detection outcomes7,16,24 while the upcoming stage 2 of the Br.36
of ctDNA before surgery was associated with worse prog- clinical trial17 will address how ctDNA detection can guide
nosis which might enable stratification to treatment inten- intensification in patients receiving adjuvant pem-
sification. However, additional time points within the first 1- brolizumab to further establish ctDNA detection as a critical
4 months of treatment initiation with immune checkpoint biomarker in early-stage NSCLC.
blockers have been proposed.12,16,17 Here we further
demonstrated that clearance of ctDNA has comparable Conclusions
prognosis to patients with a baseline negative ctDNA, in line In this study, we demonstrated that ctDNA persistence,
with recently published results from both early-stage16,17 detected using an integrated genomic and methylation
and advanced-stage NSCLC.18 Using clearance as an assay from plasma ctDNA, is highly predictive of clinical
endpoint for future clinical studies is of high interest since it outcome in early-stage NSCLC and that the integration of
would allow inclusion of patients where a baseline sample radiographic tumor volume can improve prediction. Such
could not be obtained and thus would facilitate treatment biomarkers may help tailor-adjuvant therapies or surveil-
stratification based on MRD detection. lance strategies in resected NSCLC patients.
Interestingly, in our study detection of ctDNA after
definitive SOC therapy was highly associated with worse ACKNOWLEDGEMENTS
clinical outcome and this pragmatic endpoint might there-
We thank Ashli Nguyen-Villarreal, MS, Associate Scientific
fore facilitate comparison across treatment regimens and
Editor, and Sarah Bronson, Scientific Editor, in the Research
would allow a more personalized ctDNA-guided therapy.
Medical Library at The University of Texas MD Anderson
We furthermore confirmed that tumor volume was
Cancer Center, for editing this article. The authors would like
associated with ctDNA detectability.8 However, both ctDNA
to acknowledge the Thoracic medical and surgical clinical
absence and smaller radiological tumor volume were
team for their research support. We would also like to
independently associated with prolonged RFS. This high-
acknowledge the generous philanthropic contributions to
lights that both measurements are at least partially inde-
the University of Texas MD Anderson Cancer Center Moon
pendent predictors of relapse. Thus, the integration of
Shots Program, the Mugnaini fund, and Guardant Health for
ctDNA detection with radiological volumetric tumor mea-
their support and collaboration on this project.
surement might further enhance the stratification of
patients.
FUNDING
Importantly, the assay used in this study is a laboratory-
developed assay that detects ctDNA for MRD assessment This work was supported by The University of Texas MD
and does not require baseline tissue profiling, which is a Anderson Lung Moon Shot Initiative, UT MD Anderson
clear logistical and practical advantage compared to assays Cancer CenterdNational Institute of Health [grant numbers
with similar performance that require customization for CCSG CA016672, R01 CA262425, R01 CA276178], Guardant
each patient.17 The availability of tumor-agnostic ap- Health, Inc.
proaches should facilitate access to MRD testing for pa-
tients who do not have sufficient tumor tissue available for DISCLOSURE
tissue-informed assays and those who require a short test HTT reports consulting for Abion Bio. SH reports consulting/
turnaround time following definitive treatment.14,19 How- speaker fees from Guardant Health, Qiagen, Boehringer-
ever, use of such an assay might be more prone to influence Ingelheim, and AstraZeneca. NV reports consulting fees
by clonal hematopoeisis,20 and additional studies are from Sanofi Genzyme, and Oncocyte. JZ reports personal fees
required to further investigate specificity of ctDNA detec- from AstraZeneca, Geneplus, Hengrui, Innovent, Merck, and
tion in this setting. However, detection rates at baseline Roche as well as grants and personal fees from Johnson and
were comparable to other published assays21 with higher Johnson and Novartis, and grants from OrigMed. VKL reports
ctDNA detection in squamous cell carcinoma. Furthermore, advisory roles and/or consulting fees from Seagen, Bristol
tumor staging is another important factor to be considered Myers Squibb, AstraZeneca, Guardant Health, Anheart
for treatment selection in early-stage NSCLC, independent Therapeutics, and research funding from Seagen, Bristol
of ctDNA status, and prospective studies are needed to Myers Squibb, Merck, and AstraZeneca. AV reports fees from
better understand how tumor staging, ctDNA, and radio- AstraZeneca. MCBG reports research funding from Siemens
logical analysis can be integrated. Healthcare. TC reports speaker fees and honoraria from the
The SOC at the time of sample collection (before 2021) Society for Immunotherapy of Cancer, Bristol-Myers Squibb,
was adjuvant or neoadjuvant chemotherapy with platinum- Roche, Medscape, and PeerView; travel, food, and beverage
based doublet therapy for resected NSCLC (stage IB-IIIA). expenses from Dava Oncology and Bristol Myers Squibb;
Recently, however, neoadjuvant chemoimmunotherapy7 advisory roles and/or consulting fees from MedImmune/
and adjuvant immunotherapy22 have both been approved AstraZeneca, Bristol Myers Squibb, EMD Serono, Merck &
by the Food and Drug Administration in this setting, with Co., Genentech, Arrowhead Pharmaceuticals, and Regen-
recently published data supporting the efficacy of a regimen eron; and institutional research funding from MedImmune/
combining the two approaches.23 In this setting, analysis of AstraZeneca, Bristol Myers Squibb, Boehringer-Ingelheim,

188 https://doi.org/10.1016/j.annonc.2023.11.008 Volume 35 - Issue 2 - 2024


H. T. Tran et al. Annals of Oncology

and EMD Serono. BS reports personal fees from AstraZeneca, 10. Wu J, Li C, Gensheimer M, et al. Radiological tumour classification
Medscape, and PeerView. DLG reports consulting or advisory across imaging modality and histology. Nat Mach Intell. 2021;3(9):787-
798.
role: Sanofi, GlaxoSmithKline, Janssen Research & Develop- 11. Saad MB, Hong L, Aminu M, et al. Predicting benefit from immune
ment, Eli Lilly, and Menarini Ricerche; research funding: checkpoint inhibitors in patients with non-small-cell lung cancer by
Janssen Research & Development, Takeda, AstraZeneca, CT-based ensemble deep learning: a retrospective study. Lancet Digit
Astellas, Ribon Therapeutics, and NGM Biopharmaceuticals; Health. 2023;5(7):e404-e420.
travel, accommodations, expenses: AstraZeneca/MedI- 12. Abbosh C, Frankell AM, Harrison T, et al. Tracking early lung cancer
metastatic dissemination in TRACERx using ctDNA. Nature.
mmune, BerGenBio, and Takeda. JW and JVH report advisory 2023;616(7957):553-562.
roles and consulting fees from AstraZeneca, EMD Serono, 13. Pellini B, Chaudhuri AA. Circulating tumor DNA minimal residual dis-
Boehringer-Ingelheim, Catalyst, Genentech, GlaxoSmithK- ease detection of nonesmall-cell lung cancer treated with curative
line, Hengrui Therapeutics, Eli Lilly, Spectrum, Sanofi, Takeda, intent. J Clin Oncol. 2022;40(6):567-575.
Mirati Therapeutics, BMS, BrightPath Biotherapeutics, Jans- 14. Gale D, Heider K, Ruiz-Valdepenas A, et al. Residual ctDNA after
treatment predicts early relapse in patients with early-stage non-small
sen Global Services, Nexus Health Systems, Pneuma Respi- cell lung cancer. Ann Oncol. 2022;33(5):500-510.
ratory, RefleXion, and Chugai Pharmaceuticals; institutional 15. Zhou C, Das Thakur M, Srivastava MK, et al. 2O IMpower010: bio-
research funding from AstraZeneca, Boehringer-Ingelheim, markers of disease-free survival (DFS) in a phase III study of atezoli-
Spectrum, and Takeda; and royalties and licensing fees zumab (atezo) vs best supportive care (BSC) after adjuvant
from Spectrum. All other authors have declared no conflicts chemotherapy in stage IB-IIIA NSCLC. Ann Oncol. 2021;32:S1374.
16. Felip E, Srivastava M, Reck M, et al. 1O - IMpower010: ctDNA status in
of interest. patients (pts) with resected NSCLC who received adjuvant chemo-
therapy (chemo) followed by atezolizumab (atezo) or best supportive
REFERENCES care (BSC). Ann Oncol. 2022;16:100100.
1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA 17. Anagnostou V, Ho C, Nicholas G, et al. ctDNA response after pem-
Cancer J Clin. 2022;72(1):7-33. brolizumab in non-small cell lung cancer: phase 2 adaptive trial results.
2. Pantel K, Alix-Panabières C. Liquid biopsy and minimal residual disease Nat Med. 2023;29(10):2559-2569.
d latest advances and implications for cure. Nat Rev Clin Oncol. 18. Pellini B, Madison RW, Childress MA, et al. Circulating tumor DNA
2019;16(7):409-424. monitoring on chemo-immunotherapy for risk stratification in
3. Tarazona N, Gimeno-Valiente F, Gambardella V, et al. Targeted next- advanced nonesmall cell lung cancer. Clin Cancer Res. 2023;29(22):
generation sequencing of circulating-tumor DNA for tracking minimal 4596-4605.
residual disease in localized colon cancer. Ann Oncol. 2019;30(11): 19. Newman AM, Bratman SV, To J, et al. An ultrasensitive method for
1804-1812. quantitating circulating tumor DNA with broad patient coverage. Nat
4. Tie J, Wang Y, Tomasetti C, et al. Circulating tumor DNA analysis detects Med. 2014;20(5):548-554.
minimal residual disease and predicts recurrence in patients with stage 20. Hu Y, Ulrich BC, Supplee J, et al. False-positive plasma genotyping
II colon cancer. Sci Transl Med. 2016;8(346):346ra92. due to clonal hematopoiesis. Clin Cancer Res. 2018;24(18):4437-
5. Chaudhuri AA, Chabon JJ, Lovejoy AF, et al. Early detection of mo- 4443.
lecular residual disease in localized lung cancer by circulating tumor 21. Chabon JJ, Hamilton EG, Kurtz DM, et al. Integrating genomic features
DNA profiling. Cancer Discov. 2017;7(12):1394-1403. for non-invasive early lung cancer detection. Nature. 2020;580(7802):
6. Almodovar K, Iams WT, Meador CB, et al. Longitudinal cell-free DNA 245-251.
analysis in patients with small cell lung cancer reveals dynamic insights 22. Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant
into treatment efficacy and disease relapse. J Thorac Oncol. chemotherapy in resected stage IB-IIIA non-small-cell lung cancer
2018;13(1):112-123. (IMpower010): a randomised, multicentre, open-label, phase 3 trial.
7. Forde PM, Spicer J, Lu S, et al. Neoadjuvant nivolumab plus chemo- Lancet. 2021;398(10308):1344-1357.
therapy in resectable lung cancer. N Engl J Med. 2022;386(21):1973- 23. Heymach JV, Harpole D, Mitsudomi T, et al. Perioperative durvalumab
1985. for resectable nonesmall-cell lung cancer. N Engl J Med. 2023;389(18):
8. Avanzini S, Kurtz DM, Chabon JJ, et al. A mathematical model of ctDNA 1672-1684.
shedding predicts tumor detection size. Sci Adv. 2020;6(50):eabc4308. 24. Reck M, Gale D, Harpole D, et al. LBA59 - Associations of ctDNA
9. Lam VK, Zhang J, Wu CC, et al. Genotype-specific differences in clearance and pathological response with neoadjuvant treatment in
circulating tumor DNA levels in advanced NSCLC. J Thorac Oncol. patients with resectable NSCLC from the phase III AEGEAN trial. Ann
2021;16(4):601-609. Oncol. 2023;34:S1254-S1335.

Volume 35 - Issue 2 - 2024 https://doi.org/10.1016/j.annonc.2023.11.008 189

You might also like