You are on page 1of 21

Journal Pre-proof

Immune and circulating tumor DNA profiling following radiation treatment for
oligometastatic NSCLC; translational correlatives from a mature randomized phase II
trial

Chad Tang, MD, Won-Chul Lee, PhD, Alexandre Reuben, PhD, Lianpeng Chang,
PhD, Hai Tran, PhD, Latasha Little, BS, Curtis Gumbs, BS, Jennifer Wargo, MD,
Andrew Futreal, PhD, Zhongxing Liao, MD PhD, Xuefeng Xia, PhD, Xin Yi, PhD,
Steven G. Swisher, MD, John V. Heymach, MD PhD, Daniel Gomez, MD, Jianjun
Zhang, MD PhD
PII: S0360-3016(19)33961-6
DOI: https://doi.org/10.1016/j.ijrobp.2019.10.038
Reference: ROB 26015

To appear in: International Journal of Radiation Oncology • Biology • Physics

Received Date: 9 August 2019


Revised Date: 2 October 2019
Accepted Date: 10 October 2019

Please cite this article as: Tang C, Lee W-C, Reuben A, Chang L, Tran H, Little L, Gumbs C, Wargo J,
Futreal A, Liao Z, Xia X, Yi X, Swisher SG, Heymach JV, Gomez D, Zhang J, Immune and circulating
tumor DNA profiling following radiation treatment for oligometastatic NSCLC; translational correlatives
from a mature randomized phase II trial, International Journal of Radiation Oncology • Biology • Physics
(2019), doi: https://doi.org/10.1016/j.ijrobp.2019.10.038.

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.

© 2019 Published by Elsevier Inc.


Immune and circulating tumor DNA profiling following radiation treatment for oligometastatic NSCLC;

translational correlatives from a mature randomized phase II trial

Chad Tang MD1,7*†, Won-Chul Lee PhD2,3*†, Alexandre Reuben PhD 3*, Lianpeng Chang PhD 4, Hai Tran

PhD 3, Latasha Little BS2, Curtis Gumbs BS2, Jennifer Wargo MD2,5, Andrew Futreal PhD2, Zhongxing Liao

MD PhD1, Xuefeng Xia PhD4, Xin Yi PhD4, Steven G Swisher MD6, John V Heymach MD PhD3#, and Daniel

Gomez MD1#, Jianjun Zhang MD PhD2,3#

Departments of Radiation Oncology1, Genomic Medicine2, Thoracic/Head and Neck Medical Oncology3,

Surgical Oncology5, Thoracic Surgery6, and Investigational Cancer Therapeutics7, MD Anderson Cancer

Center, Houston TX USA.

Geneplus institute-Beijing4, Beijing China.

*,#These authors contributed equally to this work. †CT and WL were responsible for statistical analysis.

Correspondence to: John Heymach at Department of Thoracic/Head & Neck Medical Oncology, Unit

0432, 1515 Holcombe Blvd., Houston TX 77030-4009, phone 713-792-6363, fax 713-792-1220, email

jheymach@mdanderson.org, Daniel Gomez, Unit 1422, 1400 Pressler St., Houston TX 77030-4008,

phone 713-563-8446, Fax 713-563-2366, Email dgomez@mdanderson.org, or Jianjun Zhang at

Department of Thoracic/Head & Neck Medical Oncology, Unit 0432, 1515 Holcombe Blvd., Houston TX

77030-4009, phone 713-563-6096, Fax 713-563-6096, Email jzhang20@mdanderson.org

Short title: Correlatives oligomet NSCLC phase II trial

Conflict of interest: CT has received consulting fees from Reflexion for work not related to this

manuscript. LC, XX, and XY work for the Geneplus which is a for-profit company and JZ has received

consulting fees from Geneplus. JVH has received personal fees from Guardant Health, Boehringer

Ingelheim, Exelixis, Genentech, GSK, Lilly, Novartis, Spectrum, EMD Serono, SyntaÐ, Hengrui, Bayer,

Takeda, Biotree, BMS, and Astrazeneca outside the submitted work not related to this manuscript.
Funding: Supported in part by Cancer Center Support (Core) Grant NCI CA016672 to The University of

Texas MD Anderson Cancer Center and the Lung Cancer Moonshot Initiative.
ABSTRACT

Purpose: NCTXXXXXX was a phase II prospective trial in which non-small lung cancer patients were

randomized to local consolidative therapy (LCT) versus maintenance therapy or observation (MT/O).

Methods and Materials: Peripheral blood from patients enrolled on NCTXXXXXX were labelled as 1)

baseline, 2) early follow-up (FU) if obtained in the 1st or 2nd FU evaluation (6-18 weeks), and 3) late FU if

obtained in the 3rd to 6th FU evaluations (22-50 weeks). All LCT patients included in this analysis received

radiation. Among 49 randomized patients, 21 underwent T-cell CDR3 variable region sequencing utilizing

immunoSEQ, 31 patients underwent ctDNA analysis via next generation sequencing with a 1,021 cancer

gene panel, and cytokine concentration was assayed in 19 patients utilizing ELISA. All analyses were

exploratory and not corrected for multiple testing.

Results: No associations were identified between baseline T-cell repertoire and ctDNA metrics with

patient outcomes. Among baseline cytokines, IL 1α was the only cytokine associated with both OS

(HR=0.02, 95% CI: 0.1-0.5, P=0.0006) and PFS (HR=0.5, 95% CI: 0.2-0.9, P=0.03).

At early FU, LCT was associated with decreased ctDNA burden including lower number of

detected mutations (median: 2 [interquartile range, IQR: 1-6] vs. 6 [IQR: 4-18]) and decreased average

variable allele frequency (VAF; median: 0.006 [IQR: 0.003-0.010] vs. 0.011 [IQR: 0.007-0.014]) compared

with MT/O. Among 6 patients with serial ctDNA analysis, a rise in ctDNA detected mutation burden

preceded clinical progression by 6.7 months. At early FU, LCT was associated with changes in T-cell

clonality that suggested oligoclonal expansion, specifically increased T-cell clonality (median: 0.15 [IQR:

0.12-0.24] vs. 0.10 [IQR: 0.05-0.13])) and frequency of top 10 clones (median: 0.14 [IQR: 0.06-0.18] vs

0.21[IQR: 0.19-0.28])).

Conclusion: LCT was associated with decreased ctDNA burden and oligoclonal expansion at early FU

timepoints. Baseline IL 1α was associated with improved patient outcomes.

1
INTRODUCTION:

In 1995 Wiechselbaum and Hellman posited the existence of an intermediate metastatic state between

localized and widespread metastatic disease, “oligometastatic” disease state, which was believed to

represent disease with limited dissemination potential and thus it was hypothesized that these patients

may derive benefit from definitive local therapy[11]. Recently two randomized phase II trials have

indicated a PFS and OS benefit in oligometastatic NSCLC patients who received definitive local

therapy[7,8,13]. Explanations for this benefit include decreased bulk of cancer clonogens with

metastatic potential, abrogation of pro-metastatic cytokine secretion from bulky tumors, and induction

of systemic anti-tumor immunity from irradiated lesions[9,15,21]. However, translational studies

investigating the mechanisms underlying the observed benefit with local therapy in this disease state

has been lacking.

We have reported a PFS benefit (median: PFS 4 vs 12 months, P=0.005) from local consolidation

therapy (LCT) in a phase II randomized trial investigating local consolidative radiation compared to

maintenance therapy or observation (MT/O) in oligometastatic NSCLC patients[7,8]. Subsequent follow

up demonstrated an overall survival (OS) benefit with LCT (median OS: 17 vs 41 months, P=0.017)[8]. In

the current study, we analyzed peripheral blood samples from this trial for immunogenomic profiles

including cytokine profile and T cell receptor (TCR) repertoire in addition to mutations detected in

circulating tumor DNA (ctDNA). The aim of the current study is to investigate changes in peripheral

biomarkers induced by LCT and the association of these markers with patient outcomes.

METHODS AND MATERIALS

Study Design

2
The details of the clinical study design and clinical results have been previously published[7,8]. Briefly,

49 NSCLC patients with up to 3 metastatic lesions after induction systemic therapy were randomized to

definitive local therapy to all sites of disease (LCT arm) versus maintenance therapy/observation (MT/O

arm). Peripheral blood was obtained from enrolled patients and categorized as 1) baseline if obtained

immediately prior to randomization up to LCT administration, 2) early follow-up (FU) if obtained in the

1st or 2nd FU visit (6-18 weeks after randomization), and 3) late FU if obtained in the 3rd to 6th FU visits

(22-50 weeks after randomization). In the event that patients had multiple correlative blood draws

within a given time range the earlier timepoint was utilized. Assays selection was post-hoc and

conducted blinded to clinical outcomes and randomization arm. Correlatives were optional for study

patients and collected until progression. Depending on the amount and quality of collected material,

assays were prioritized to as follows: 1) TCR sequencing, 2) ctDNA analysis, and 3) cytokine analysis. All

LCT arm patients with correlative blood draws received definitive radiation therapy.

Circulating Tumor DNA (ctDNA) Sequencing

Collected ctDNA was subjected to next generation sequencing of a 1,021 cancer gene panel using DNA

from leukocytes as germline control (Geneplus) as previously described[16]. Briefly, peripheral blood

was collected in EDTA Vacutainer tubes (BD Diagnostics, Franklin Lakes, NJ, USA) and processed within 2

h to separate plasma and buffy coat (as a source samples of germline DNA). Circulating DNA was

isolated from plasma using a QIAamp Circulating Nucleic Acid Kit (Qiagen, Hilden, Germany). Buffy coat

DNA was extracted using the DNeasy Blood & Tissue Kit (Qiagen). DNA concentration was measured

using a Qubit fluorimeter and the Qubit dsDNA HS (High Sensitivity) Assay Kit (Invitrogen, Carlsbad, CA,

USA). The size distribution of the ctDNA was assessed using an Agilent 2100 BioAnalyzer and a DNA HS

kit (Agilent Technologies, Santa Clara, CA, USA).

3
Before library construction, 1 μg each of buffy coat DNA was sheared to 300-bp fragments with

a Covaris S2 ultrasonicator. Indexed Illumina NGS libraries were prepared from tissue, and germ line and

circulating DNA libraries were prepared using the KAPA Library Preparation Kit (Kapa Biosystems,

Wilmington, MA, USA). Libraries were hybridized to custom-designed biotinylated oligonucleotide

probes (Integrated DNA Technologies, Iowa, IA, USA). Capture probe was designed to cover coding

sequencing or hot exons of 1,021 genes frequently mutated in solid tumors (Supplemental Table

1)[4,22]. Genes and coordinates of selected regions of each version are provided in Supplemental Table

1. Following hybrid selection, the captured DNA fragments were amplified and then pooled to generate

several multiplex libraries. Sequencing was conducting using Illumina 2×100 bp paired-end reads on an

Illumina HiSeq 3000 instrument according to the manufacturer's recommendations using a TruSeq PE

Cluster Generation Kit v3 and a TruSeq SBS Kit v3 (Illumina, San Diego, CA, USA).

Identification of somatic mutation in ctDNA

Terminal adaptor sequences and low-quality reads were removed from raw data of paired samples.

Burrows-Wheeler Aligner (BWA; version 0.7.12-r1039) was employed to align clean reads to the

reference human genome (hg19). Picard (version 1.98) was used to mark polymerase chain reaction

(PCR) duplicates. Realignment and recalibration was performed using GATK (version 3.4-46-gbc02625).

Single nucleotide variants (SNV) were called using MuTect2 (3.4–46-gbc02625) and NChot, a software

developed in-house to review hotspot variants[24]. Small insertions and deletions (InDels) were

determined by GATK. Candidate variants were manually verified in the Integrative Genomics Viewer

(IGV).

Mutations were considered as a candidate somatic mutation only when (i) the mutation had at

least five high-quality reads (Phred score ≥20, mapping quality ≥20, and without paired-end reads bias)

containing the particular base; (ii) variable allele frequency (VAF) ≥0.5 %; (iii) the mutation was not

4
present in ≥1% of population in the 1,000 Genomes Project or dbSNP databases; (iv) the mutation was

not presented in ≥0.1% of population in the ESP6500 and ExAC databases; and (v) the mutation was not

present in a local database of normal samples. All somatic mutations annotated by ANNOVAR were used

in clonal structure reconstruction PyClone. From the PyClone analysis, we determined a major clone to

be one in which the average cancer cell fraction was the highest among all distinct clones identified.

Mutations belonging to the major clone were defined as clonal mutations. Metrics analyzed include

mutation burden and variable allele frequency (VAF).

T-cell Receptor (TCR) Sequencing

Sequencing of the CDR3 regions of human TCR-β chains was performed using the immunoSEQ® Assay

(Adaptive Biotechnologies, Seattle, WA) as previously described[19]. Clonality was defined as 1-Peilou’s

evenness [14] and was calculated on productive rearrangements by:

∑ p log ( p )
N

i 2 i
i

( )
1+
log 2 N

Where pi is the proportional abundance of rearrangement i and N is the total number of

rearrangements. T cell density was calculated by normalizing TCR-β template counts to the total amount

of DNA usable for TCR sequencing, where the amount of usable DNA was determined by PCR-

amplification and sequencing of housekeeping genes expected to be present in all nucleated cells.

Richness was calculated using the preseqR package.

Cytokine Analysis

Peripheral blood samples were collected and plasma was stored at -80C until analysis. For cytokines,

chemokines and angiogenic factors (CAF) analysis, plasma samples were processed and analyzed using

multiplexed magnetic bead-based assays (EMD Bioscience Research Reagents, Temecula, CA, USA) as

5
previously published[10,12]. For all CAF analysis, duplicate samples were analyzed and the mean

reported. In the event that the detected cytokine concentration was below the limit of detection, for

the purposes of calculations, the threshold of detection value was inputted.

Statistical Analysis

Continuous variables were compared between randomization arms via T-test. Spearman correlation

coefficients were utilized to assess association of continuous variables. Time-to-event analyses were

conducted starting at the date of randomization and comparisons made via Cox regression analyses.

Cytokine concentrations were subject to log base 10 transformation and were analyzed as continuous

variables, with confirmatory analysis utilizing log-rank test stratified by the median value. We did not

control for multiple testing as all analyses were exploratory. Analyses were performed utilizing SAS ver.

9.4 (Cary, NC).

RESULTS

Dynamic change of ctDNA mutation landscape with or without LCT

ctDNA analysis were conducted on 21 patients (LCT arm=10, MT/O arm=11) who had next-generation

sequencing (NGS) of 1,021 cancer genes from ctDNA. Baseline ctDNA metrics were not associated with

overall survival (OS) or progression free survival (PFS). At early FU timepoints, patients randomized to

the LCT arm exhibited lower detected mutation burden (median: 2 [IQR: 1-6] vs. 6 [IQR: 4-18]), average

VAF (median: 0.006 [IQR: 0.003-0.010] vs. 0.011 [IQR: 0.007-0.015]), highest VAF (0.008 [IQR: 0.004-

0.014] vs. 0.03 [IQR: 0.010-0.048]), and average VAF of clonal mutations (0.007 [IQR: 0.002-0.023] vs.

0.01 [IQR: 0.008-0.026]) than patients randomized to the MT/O LCT arm (Fig. 1a) that may have

reflected the reduction of tumor burden by LCT. At late FU timepoints, no difference in ctDNA metrics

was observed between randomization groups (Fig. 1a).

6
Six patients underwent serial ctDNA analysis with at least one baseline measurement, 5 of which

were noted to progress radiographically at last follow-up. Among the 5 patients who progressed

radiographically, all exhibited an increase in the mutation burden above what was measured at baseline.

The first detection of mutation burden increase was noted at a median of 6.7 months (range: 2.9 to 17.9

months) prior to radiographic progression (Fig. 1b). In the patient who did not progress at last FU, there

was no rise in the mutation burden. In this patient the number of detected mutations decreased from

31 measured at baseline to 2 at last follow up. These results are consistent with previous reports

suggesting that ctDNA metrics may reflect the disease burden over the treatment course and molecular

progression can precede radiological progression[2,5].

TCR Analysis

Peripheral blood TCR sequencing analysis was conducted in 31 patients (LCT arm=15 and MT/O arm=16).

Baseline and changes in TCR metrics were not associated with overall survival (OS) or progression free

survival (PFS). Assessing early FU timepoints, patients in the LCT arm exhibited increased clonality

(median: 0.15 [IQR: 0.12-0.24] vs. 0.10 [IQR: 0.05-0.13]) compared with the MT/O arm (Fig. 2a),

suggesting expansion of specific T cell clones. Correspondingly, in the LCT arm the top 10 clones

(median: 0.14 [IQR: 0.06-0.18] vs 0.21 [IQR: 0.19-0.28]) and top 100 clones (median: 0.20 [IQR: 0.11-

0.27] vs 0.30 [IQR: 0.26-0.42]) were higher among patients who received LCT although the difference did

not reach statistical significance. In contrast, the frequency of the top clone was similar between both

arms (median: 0.05 [IQR: 0.03-0.09] vs 0.07 [IQR: 0.04-0.09]; Fig. 2a). Representative plots of T cell clone

frequency at baseline versus early FU from a patient treated in the MT/O arm demonstrated no

substantial changes in the observed frequencies between timepoints (Fig. 2b). A representative plot

from a patient treated in the LCT arm shows significant increase in existing clones and the emergence of

new clones at early FU (Fig. 2b).

7
Peripheral Cytokine Analysis

Plasma samples were available from 19 patients for analysis of 36 CAFs. This analysis demonstrated

significant association between high levels of IFNγ (HR=0.4, 95% CI: 0.1-0.96, P=0.04), IL12 (p70)

(HR=0.3, 95% CI: 0.1-1.0, P=0.0498), IL 1α (HR=0.02, 95% CI: 0.1-0.5, P=0.0006), and IP 10 (HR=0.1, 95%

CI: 0.0-0.95, P=0.04) at baseline with improved OS (Table 1). Of these cytokines that exhibited an

association with OS, only IL 1α also exhibited an association with PFS (HR=0.5, 95% CI: 0.2-0.9, P=0.03)

as well. When assessing IL 1α as a binary variable dichotomized by the median value an association was

observed with OS (P=0.0009) and PFS (P=0.04) (Fig. 3a). No clear temporal trends in IL 1α concentration

were identified.

IL 1α is produced mainly by activated macrophages and neutrophils, among others, to promote

a pro-inflammatory state[3]. Further analysis showed that baseline IL 1α concentration was found to be

associated with baseline white blood cell count (WBC; R=0.65, P=0.02) and absolute neutrophil count

(ANC; R=0.61, P=0.03) but not absolute lymphocyte count (ALC) in 12 patients with concordant clinical

blood draws at baseline (Fig. 3b). Of note, baseline IL 1α concentration was not associated with ANC and

WBC count measured at other time points including at the first follow up or prior to induction systemic

therapy.

DISCUSSION

Clinical trials from our group and others have demonstrated that patients with oligometastatic NSCLC

may benefit from upfront LCT[7,8,13]. The underlying mechanisms of the observed survival benefit are

still under debate and there are currently no biomarkers to identify patients who will benefit most from

LCT. To our knowledge, this is the first correlative study on a completed randomized phase II study

assessing the role of LCT for oligometastatic NSCLC. Given the small sample size, the goal of this study

8
was to identify trends in peripheral blood biomarkers and was thus exploratory in nature. The following

observations were made: 1) ctDNA measures of tumor burden at early FU were numerically lower in

patients in the LCT arm compared with the MT/O arm but at late FU these differences resolved. ctDNA

levels increased prior to radiographic evidence of disease progression in all patients who exhibited

clinical progression. 2) Radiation may promote an oligoclonal peripheral T-cell expansion at early FU as

indicated by increase in clonality and frequency of top clones. 3) Numerous baseline peripheral

cytokines were associated with OS and PFS, of which only IL 1α was associated with both OS and PFS.

The current gold standard to identify the oligometastatic state is counting the number of

metastatic sites evident on conventional imaging. However, this definition is considered to be crude and

investigators have advocated for the incorporation of biomarkers that quantify systemic disease burden

into the definition of oligometastatic disease[11,23]. Mutation burden as detected via ctDNA have been

reported to be associated with overall tumor burden and thus may facilitate defining patients with

“true” oligometastatic disease[1]. There therefore exists the potential for ctDNA to predict those who

may benefit from LCT, to identify patients who may benefit from continued systemic therapy vs.

observation after LCT, and to expedite detection of progression or recurrence[17]. However, the current

study did not identify an association between baseline ctDNA mutations and outcomes, due to small

sample size and intragroup heterogeneity. Patients enrolled on the LCT arm exhibited numerically lower

ctDNA burden at early follow up, although these differences did not reach statistical significance once

again due to small sample size. This finding is consistent with the hypothesis that the ctDNA

concentration may have reflected tumor burden that is reduced with LCT. Finally, ctDNA concentration

increased preceding clinical progression in all patients tested. This occurred at a median of 6.7 months

prior to radiographic progression, which is similar to the median 5.2 months identified by Chaudhuri et

al. who analyzed 40 stage I-IIII NSCLC patients treated with curative intent[5].

9
Radiation-induced cancer cell apoptosis remains the primary mechanism of radiotherapy-

induced effect. However, a plethora of evidence has suggested that immunogenic cell death may play an

important role in cancer cell killing[18,21]. This is of particular interest in the context of oligometastatic

state, whereby abscopal effect is warranted to control the micrometastases and drive the overall

therapeutic benefit from radiation. In the current study, radiation was associated with changes in the

peripheral T cell repertoire, specifically decreased diversity of peripheral T cell receptors concordant

with clonal expansion of a subset of T cell clones as evidenced by lower richness and increased

productive clonality. Furthermore, the frequency of the top 10 and 100 clones increased after LCT while

the frequency of the top clone was similar between arms. However these changes were only detected

transiently at early FU while at late FU TCR metrics were similar between LCT and MT/O patients. We

interpret these findings to suggest that radiation induces an early oligoclonal expansion, in which T cell

clones reactive towards released antigens disproportionally proliferated. These findings are consistent

with Tran et al. who recently demonstrated radiation-induced T cell clonal expansion from a partially

completed randomized study of oligometastatic prostate cancer[20].

Cytokines play indispensable roles in inflammation and anti-tumor immune response[6]. In the

current study, multiple cytokines were found to be associated with either PFS or OS, of which, only IL-1α

exhibited a significant association with both PFS and OS. IL-1α is secreted as an “alarm cytokine” from

macrophages and initiates an inflammatory cascade that facilitates neutrophil mobilization and anti-

tumor activity[3]. These findings are consistent with the observed association between IL-1α

concentration and baseline peripheral ANC and WBC. A potential mechanism to explain this association

is that a systemic anti-tumor inflammatory state, either exhibited by some patients at baseline or

induced by induction chemotherapy, aids in metastatic disease control.

As an exploratory analysis, our analyses had many limitations including small sample size,

diverse patient population, heterogeneous blood collection time-points, etc. that have made rigorous

10
statistical analysis impossible. Despite these limitations, this is the first study to our knowledge to assess

ctDNA, cytokine and TCR biology in oligometastatic NSCLC with or without LCT and one of the first

studies to assess these markers in the oligometastatic state in any cancer types. These findings provide

interesting preliminary data for the design of correlative endpoints for future trials assessing therapies

for the oligometastatic state. To this end, the kinetics of biomarker changes observed in this study have

informed biomarker collection timepoints in our ongoing XXXXX basket trial (NCTXXXXXX) and XXXXX

(NCTXXXXXXX).

REFERENCES

[1] Abbosh C, et al. Phylogenetic ctdna analysis depicts early-stage lung cancer evolution. Nature

2017;545:446-451.

[2] Anagnostou V, et al. Dynamics of tumor and immune responses during immune checkpoint

blockade in non-small cell lung cancer. Cancer Res 2019;79:1214-1225.

[3] Apte RN, et al. The involvement of il-1 in tumorigenesis, tumor invasiveness, metastasis and

tumor-host interactions. Cancer Metastasis Rev 2006;25:387-408.

[4] Cao W, et al. Genotyping of circulating tumor DNA reveals the clinically actionable mutation

landscape of advanced colorectal cancer. Mol Cancer Ther 2019;18:1158-1167.

[5] Chaudhuri AA, et al. Early detection of molecular residual disease in localized lung cancer by

circulating tumor DNA profiling. Cancer Discov 2017;7:1394-1403.

[6] Coussens LM Werb Z. Inflammation and cancer. Nature 2002;420:860-867.

[7] XXXXXXXXXX

[8] XXXXXXXXXX

[9] Gundem G, et al. The evolutionary history of lethal metastatic prostate cancer. Nature

2015;520:353-357.

11
[10] Hanrahan EO, et al. Distinct patterns of cytokine and angiogenic factor modulation and markers

of benefit for vandetanib and/or chemotherapy in patients with non-small-cell lung cancer. J Clin

Oncol 2010;28:193-201.

[11] Hellman S Weichselbaum RR. Oligometastases. J Clin Oncol 1995;13:8-10.

[12] Heymach JV, et al. Effect of low-fat diets on plasma levels of nf-kappab-regulated inflammatory

cytokines and angiogenic factors in men with prostate cancer. Cancer Prev Res (Phila)

2011;4:1590-1598.

[13] Iyengar P, et al. Consolidative radiotherapy for limited metastatic non-small-cell lung cancer: A

phase 2 randomized clinical trial. JAMA Oncol 2018;4:e173501.

[14] Kirsch I, Vignali M Robins H. T-cell receptor profiling in cancer. Molecular oncology 2015;9:2063-

2070.

[15] McAllister SS, et al. Systemic endocrine instigation of indolent tumor growth requires

osteopontin. Cell 2008;133:994-1005.

[16] Nong J, et al. Circulating tumor DNA analysis depicts subclonal architecture and genomic

evolution of small cell lung cancer. Nat Commun 2018;9:3114.

[17] Pitroda SP Weichselbaum RR. Integrated molecular and clinical staging defines the spectrum of

metastatic cancer. Nat Rev Clin Oncol 2019.

[18] Reits EA, et al. Radiation modulates the peptide repertoire, enhances mhc class i expression, and

induces successful antitumor immunotherapy. J Exp Med 2006;203:1259-1271.

[19] Reuben A, et al. Tcr repertoire intratumor heterogeneity in localized lung adenocarcinomas: An

association with predicted neoantigen heterogeneity and postsurgical recurrence. Cancer Discov

2017;7:1088-1097.

12
[20] Tran PT, et al. Sabr produces systemic adaptive immune responses in castration-sensitive

oligometastatic prostate cancer patients. International Journal of Radiation Oncology • Biology •

Physics 2018;102:S24-S25.

[21] Twyman-Saint Victor C, et al. Radiation and dual checkpoint blockade activate non-redundant

immune mechanisms in cancer. Nature 2015;520:373-377.

[22] Wang Y, et al. Circulating tumor DNA analyses predict progressive disease and indicate

trastuzumab-resistant mechanism in advanced gastric cancer. EBioMedicine 2019.

[23] Weichselbaum RR Hellman S. Oligometastases revisited. Nat Rev Clin Oncol 2011;8:378-382.

[24] XXXXXXXXXXX

FIGURE LEGENDS

Figure 1: (A) peripheral circulating tumor (ctDNA) metrics at specified timepoints stratified by

randomization arm. No significant statistically significant associations were observed (all P>0.05). (B)

Swimmer plot of all patients with baseline ctDNA measurement and at least 2 additional ctDNA

measurements obtained during FU. The number of detected mutations is listed below each timepoint.

*Indicates that the specified timepoints occurred on the same day.

Figure 2: (A) Peripheral T cell CDR3 metrics at various timepoints stratified by randomization arm. P-

values listed above each comparison between arms. (B) Representative plots showing T cell clone

frequency at baseline and at first follow up in a patient enrolled on the no LCT arm and a patient

enrolled on MT/O arm. Blue circles indicate new clones detected at first follow up and red circles

indicate clones that are no longer detected at first follow up.

13
Figure 3: (A) Association of baseline IL-1α and patient outcomes when IL-1α is dichotomized by the

median baseline IL-1α value. (B) Correlation between peripheral lab values and baseline IL-1α

concentration, Spearman correlation coefficient and associated p-value is shown.

14
Table 1: Association of baseline cytokine factors and patient outcomes
PFS OS
Log10(Cytokine) HR 95% CI P HR 95% CI P
Angiopoietin 2 2.1 0.2-18.9 0.53 1.1 0.1-13.5 0.95
HGF 0.8 0.2-3.0 0.85 0.6 0.1-2.5 0.46
HBEGF 1.1 0.5-2.8 0.79 0.9 0.3-2.4 0.85
MIF 1.7 0.4-7.4 0.47 1.8 0.4-8.9 0.49
TRAIL 1.1 0.3-4.6 0.91 0.2 0.0-1.2 0.08
OPN 0.9 0.1-5.1 0.86 0.8 0.1-5.2 0.79
MMP 1 1.8 0.4-8.1 0.45 2.9 0.6-14.5 0.20
MMP 2 2.1 0.0-189 0.75 0.1 0.0-10.1 0.37
MMP 9 0.4 0.1-2.6 0.33 0.9 0.1-11.4 0.94
sICAM 1 1.3 0.4-4.3 0.65 0.7 0.1-3.2 0.63
RANTES 2.5 0.7-9.0 0.16 3.4 0.6-18.5 0.15
PDGF AB/BB 1.6 0.5-4.6 0.39 2.1 0.6-7.4 0.23
sVCAM 1 2.3 0.4-13.8 0.38 1.3 0.1-14.3 0.83
EGF 0.3 0.1-0.9 0.04 0.4 0.1-1.4 0.16
EOTAXIN 0.3 0.0-1.7 0.16 1.3 0.1-16.3 0.84
G CSF 0.8 0.4-1.6 0.44 0.6 0.3-1.3 0.18
FRACTALKINE 0.7 0.3-1.5 0.32 0.7 0.3-1.7 0.40
IFN α2 0.6 0.2-1.7 0.35 0.5 0.2-1.4 0.19
IFNγ 0.6 0.3-1.1 0.12 0.4 0.1-0.96 0.04
GRO 2.5 0.5-14.0 0.28 3.3 0.4-28.4 0.28
IL 10 0.6 0.2-1.6 0.34 0.3 0.1-1.2 0.08
MDC 0.04 0.0-1.4 0.08 1.5 0.0-54.0 0.82
IL 12 p70 0.6 0.3-1.5 0.29 0.3 0.1-1.0 0.0498
IL 15 0.7 0.4-1.5 0.38 0.5 0.2-1.2 0.10
SCD 40L 1.0 0.4-2.6 0.97 2.0 0.6-6.8 0.26
IL 1RA 0.6 0.4-1.0 0.07 0.6 0.3-1.1 0.10
IL 1α 0.5 0.2-0.9 0.03 0.2 0.1-0.5 0.0006
IL 4 0.7 0.4-1.4 0.36 0.4 0.2-1.1 0.08
IL 6 0.9 0.4-2.0 0.87 0.5 0.2-1.4 0.21
IL 7 0.7 0.3-1.6 0.36 0.4 0.2-1.1 0.09
IP 10 0.7 0.2-2.9 0.62 0.1 0.0-0.95 0.04
MCP 1 1.2 0.1-12.5 0.87 1.8 0.1-26.7 0.68
MIP 1α 0.5 0.2-1.3 0.49 0.4 0.1-1.4 0.16
MIP 1β 0.6 0.3-1.1 0.10 0.5 0.2-1.1 0.07
TNFα 0.3 0.0-1.7 0.17 0.2 0.0-1.4 0.11
VEGF 0.8 0.3-1.8 0.52 0.6 0.2-1.4 0.20

You might also like