ArticlePDF Available

Lymphopenia and Radiation Dose to Circulating Lymphocytes With Neoadjuvant Chemoradiation in Esophageal Squamous Cell Carcinoma

Authors:

Abstract

Background We hypothesized that radiation-induced lymphopenia could be predicted by the effective dose to the circulating immune cells (EDIC) in advanced esophageal squamous cell carcinoma (ESCC) treated with trimodality therapy according to the Dutch CROSS trial regimen. To test this hypothesis, we examined the effect of EDIC on the degree of lymphocyte drop (lymphocyte nadir). Methods Patients with advanced non-metastatic ESCC treated in a single tertiary cancer centre from 2012–2018 were eligible for this study. All patients must have a radiotherapy plan available for EDIC computation and received neoadjuvant chemoradiation according to the Dutch CROSS trial regimen before radical esophagectomy. The EDIC was calculated as a function of integral doses to the lung, heart and total body with a verified mathematical model. The association between EDIC and lymphocyte nadir was studied, and the relationships of overall survival (OS) with lymphocyte nadir and EDIC were assessed using multivariable Cox regression model. Results This analysis included 92 eligible consecutive patients with 77 males and 15 females. The mean EDIC was 2.8 Gy (range 0.6–4.4). EDIC was significantly correlated with lymphocyte nadir (Spearman coefficient = -0.505; P < 0.01) and lymphocyte nadir was a significant independent factor for shorter OS (Hazard ratio [HR] = 0.63; P < 0.001). Lymphocyte nadir was also the most significant factor in determining OS among other clinical parameters. Exploratory analysis showed significant OS differences between EDIC groups (< 2 Gy, 2–4 Gy and > 4 Gy). The 2–year OS rates were 66.7%, 42.7% and 16.7% for EDIC < 2 Gy, 2–4 Gy and > 4 Gy, respectively. Conclusions There was significant correlation between radiation dose to circulating immune cells and lymphocyte nadir which in turn impacted overall survival in patients with advanced non–metastatic ESCC treated by trimodality therapy.
Journal Pre-proof
Lymphopenia and radiation dose to circulating lymphocyte with neoadjuvant
chemoradiation in esophageal squamous cell carcinoma
Tsz Him So, FRCR, Sik Kwan Chan, BSC, Wing Lok Chan, FRCR, Horace Choi,
PhD, Chi Leung Chiang, FRCR, Victor Lee, MD, Tai Chung Lam, FRCR, Ian Wong,
FRCS, Simon Law, MS, Dora Kwong, MD, Feng Ming (Spring) Kong, FASTRO,
JianYue Jin, PhD, Ka On Lam, FRCR
PII: S2452-1094(20)30079-8
DOI: https://doi.org/10.1016/j.adro.2020.03.021
Reference: ADRO 444
To appear in: Advances in Radiation Oncology
Received Date: 19 January 2020
Revised Date: 25 March 2020
Accepted Date: 31 March 2020
Please cite this article as: So TH, Chan SK, Chan WL, Choi H, Chiang CL, Lee V, Lam TC, Wong I,
Law S, Kwong D, Ming (Spring) Kong F, Jin J, Lam KO, Lymphopenia and radiation dose to circulating
lymphocyte with neoadjuvant chemoradiation in esophageal squamous cell carcinoma, Advances in
Radiation Oncology (2020), doi: https://doi.org/10.1016/j.adro.2020.03.021.
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.
© 2020 The Author(s). Published by Elsevier Inc. on behalf of American Society for Radiation Oncology.
Title page
Lymphopenia and radiation dose to circulating lymphocyte with neoadjuvant
chemoradiation in esophageal squamous cell carcinoma
Short Title: Radiotherapy and Lymphopenia in CA Esophagus
Tsz Him So,
#
FRCR, Sik Kwan Chan
#
, BSC, Wing Lok Chan,
#
FRCR, Horace Choi,
#
PhD, Chi
Leung Chiang
#
, FRCR, Victor Lee,
#
MD, Tai Chung Lam
#
, FRCR, Ian Wong, * FRCS ,Simon
Law,* MS, Dora Kwong,
#
MD, Feng Ming (Spring) Kong,
#
FASTRO, JianYue Jin, PhD, Ka On
Lam,
#
FRCR
Department of
#
Clinical Oncology and * Surgery, the University of Hong Kong, Hong Kong
&
Department of Radiation Oncology, University Hospitals/Seidman Cancer Centre and Case
Comprehensive Cancer Centre, the United States
Declarations of interest: none
Source of Financial Support/Funding Statement: No external funding received
*Corresponding author
Dr. Ka-on Lam MBBS, FRCR, FHKCR, FHKAM
Specialist in Clinical Oncology and Clinical Assistant Professor
Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong
Kong; 1/F, Professorial Block, Queen Mary Hospital, Hong Kong; Email: lamkaon@hku.hk; Tel:
+852 22554352; Fax: +852 28726426
Summary
In advanced squamous cell oesophageal cancer (ESCC) treated with trimodality therapy according to the
Dutch CROSS trial regimen, we found that radiation induced lymphopenia can be predicted by the
effective dose to the circulating immune cells (EDIC). EDIC was found to be highly correlated with
lymphocyte nadir (Spearman coefficient = -0.505; P < 0.01) which was found to significantly
independently associated with shorter OS (Hazard ratio [HR] = 0.63; P < 0.001).
1
Title
Lymphopenia and radiation dose to circulating lymphocyte with neoadjuvant
chemoradiation in esophageal squamous cell carcinoma
Abstract
Background: We hypothesized that radiation-induced lymphopenia could be predicted
by the effective dose to the circulating immune cells (EDIC) in advanced esophageal
squamous cell carcinoma (ESCC) treated with trimodality therapy according to the
Dutch CROSS trial regimen. To test this hypothesis, we examined the effect of EDIC
on the degree of lymphocyte drop (lymphocyte nadir).
Methods: Patients with advanced non-metastatic ESCC treated in a single tertiary
cancer centre from 2012–2018 were eligible for this study. All patients must have a
radiotherapy plan available for EDIC computation and received neoadjuvant
chemoradiation according to the Dutch CROSS trial regimen before radical
esophagectomy. The EDIC was calculated as a function of integral doses to the lung,
heart and total body with a verified mathematical model. The association between
EDIC and lymphocyte nadir was studied, and the relationships of overall survival (OS)
2
with lymphocyte nadir and EDIC were assessed using multivariable Cox regression
model.
Results: This analysis included 92 eligible consecutive patients with 77 males and 15
females. The mean EDIC was 2.8 Gy (range 0.6–4.4). EDIC was significantly
correlated with lymphocyte nadir (Spearman coefficient = -0.505; P < 0.01) and
lymphocyte nadir was a significant independent factor for shorter OS (Hazard ratio
[HR] = 0.63; P < 0.001). Lymphocyte nadir was also the most significant factor in
determining OS among other clinical parameters. Exploratory analysis showed
significant OS differences between EDIC groups (< 2 Gy, 2–4 Gy and > 4 Gy). The 2–
year OS rates were 66.7%, 42.7% and 16.7% for EDIC < 2 Gy, 2–4 Gy and > 4 Gy,
respectively.
Conclusions: There was significant correlation between radiation dose to circulating
immune cells and lymphocyte nadir which in turn impacted overall survival in patients
with advanced non–metastatic ESCC treated by trimodality therapy.
Key words: Esophageal Squamous Cell Carcinoma (ESCC), lymphopenia, radiation
dose, lymphocyte, CROSS Regimen
3
4
Background
Radiation is well-known to have a potent lymphocyte-killing effect. Radiation can
destroy mature circulating lymphocyte at as low as 1 Gy [1].
The importance of this
has been overlooked for decades until recently when immunotherapy emerges as a
standard treatment in many cancers. Since lymphocytes play a major role in exerting
anti-tumour immunity, many clinical studies have demonstrated the correlation
between absolute lymphocyte counts (ALCs) and survival [2-4].
Radiation–induced lymphopenia has been reported to be adversely associated with
overall survival and recurrence–free survival in various cancers including
glioblastoma, non–small cell lung cancer (NSCLC), pancreatic cancer, and head and
neck cancer [5-9]. One of the possible mechanisms of lymphopenia in radiotherapy is
the large volume of low dose bath that kills a vast number of circulating lymphocytes
in both systemic and pulmonary circulation [7]. Yovino et al [10]
first reported that a
single fraction of typical RT plan (60Gy in 30 fractions) for glioblastoma patients
resulted in 0.5 Gy exposure to 5% of circulating cells. Ninety-nine percent of
circulating lymphocytes would be exposed to at least 0.5 Gy after the whole course of
5
30 fractions.
Tang et al [7] further reported significant correlation between low dose
lung dosimetry V1–V5 with the degree of lymphopenia in NSCLC patients. They
hypothesised that as all cardiac output must go through pulmonary circulation, the
large volume low dose bath radiation to pulmonary vasculature would kill most
circulating lymphocytes, contributing to worse outcomes in patients with higher V5
value. XXXX et al [11] further studied the contributing factors and calculated the
dose to circulating lymphocyte in lung cancer patients. They developed a model to
estimate the effective dose to circulating immune cells (EDIC) and found that higher
EDIC was correlated with poorer survival in lung cancer patients in the RTOG 0617
trial.
Esophagus is an organ mainly confined in the thorax. The majority of entry and exit
radiation beams have to pass through lung tissue during treatment of esophageal
cancer with either conformal 3D or intensity modulated radiotherapy (IMRT) photon
technique. Recently published studies [12-14] in the US suggested higher lymphocyte
nadir predicts better pathological and survival outcomes for esophageal cancer
patients. However, these studies included a majority of adenocarcinoma patients
treated with radical chemoradiation and there was heterogeneity regarding
6
radiotherapy doses and chemotherapy regimens. Therefore, it is worth studying the
dosimetry and clinical parameters that cause lymphopenia in esophageal squamous
cell cancer (ESCC) during neoadjuvant chemoradiation with the CROSS regimen [15],
a standard of care employed in Europe and Asia.
As immunotherapy is increasingly incorporated to the systemic treatment of various
cancer, it is important to devise a better lymphocyte–sparing radiation technique. For
example, stereotactic body radiation therapy (SBRT) is associated with significantly
less radiation–induced lymphopenia than fractionated radiotherapy in locally
advanced pancreatic cancer [16]. Lymphocyte–sparing radiation technique may be
crucial for subsequent immunotherapy if there were metastases later. For instance,
there are reported phases II and III studies on the use of immunotherapy including
checkpoint inhibitors in advanced ESCC. The landmark phase III Keynote 181
study[17] demonstrated improved clinical outcomes with second line pembrolizumab
in advanced esophageal cancer with PDL1 combined proportional score 10. Thus,
studying factors in preserving lymphocytes during initial radiotherapy for esophageal
cancer may have far-reaching importance in subsequent metastatic settings.
7
Method
Hypothesis and objectives
We performed two analyses to test two related hypotheses. Firstly, radiation–induced
lymphopenia can be predicted by the EDIC that was computed by modelling the doses
to the total body, lung and heart. Secondly, EDIC had an impact on survival in
patients with ESCC treated with trimodality therapy.
The primary objective of our study was to validate the EDIC model in ESCC and to
study the correlation of EDIC with lymphocyte nadir (lowest absolute lymphocyte
count). Secondary objectives were to explore the relationships between EDIC,
lymphocyte nadir and overall survival.
Study population
This was a retrospective study in patients with ESCC treated with neoadjuvant
chemoradiation therapy according to the CROSS regimen. After approval by our
institutional review board, clinical and dosimetric data of all ESCC patients at our
hospital from June 2012 to February 2018 were retrospectively reviewed. Patient
8
confidentiality was maintained according to our institution regulations.
Included subjects must have a histological diagnosis of ESCC and they must have
received neoadjuvant chemoradiation with the CROSS regime [15]
(Five weekly
carboplatin AUC = 2 and paclitaxel 50 mg/m
2
, radiation dose = 41.4 Gy in 23 daily
fractions) and subsequent surgery. All subjects must have records of weekly blood test
available during chemoradiation and at 2 months after the completion of
chemoradiation, and a complete dosimetry data available for EDIC computation.
Subjects with distant metastasis or dropouts before the end of chemoradiation were
excluded.
Data collection and dosimetric computation
For each eligible patient, results of blood tests including total white blood cell count,
neutrophil count, lymphocyte count and platelet count during neoadjuvant
chemoradiation were retrospectively retrieved. Lymphocyte nadir was defined as the
lowest absolute value of lymphocyte count among the set of lymphocyte values
during chemoradiation and at 2 months after the completion of chemoradiation.
9
Demographic and clinical data were retrieved from a prospectively–maintained
database. Dosimetry data was obtained from our planning system.
The EDIC was computed as a function of mean heart dose, mean lung dose, integral
dose (ITD), and number of fractions according to the method reported by XXXX et al
[11]. In this model, estimated dose to the immune system was computed by using
the dose to the circulating immune cells as a surrogate. The model assumed that the
radiation dose was uniformly delivered to all rapidly circulating immune cells in the
heart, lung and great vessels but also to the slowly circulating immune cells within the
irradiated volumes. With regard to chemoradiation for ESCC, major organs to
consider include the lung, heart, large vessels and other organs. ITD of the total body
was used to approximate the mean organ dose (MOD) for large vessels and other
organs. The blood dose contributions of the heart (mean heart dose MHD) and lungs
(mean lung dose MLD) were derived from the respective MODs and the estimated
percentage of cardiac output and blood volume they received. Then the equivalent
uniform dose (EUD) was calculated from the DVH. EDIC is the sum of EUDs of all
organs in the irradiated volume. EUD could be estimated by a simple function of
MOD of the above 4 organs.
10
In summary, EDIC was calculated using the following equation[11]:
=.+.+.+..
/


where n is the fraction number (23), k =45.
Statistical consideration
For primary endpoint of lymphocyte nadir, study variables included patient
demographics, tumour factors and treatment factors such as radiation dosimetry to the
major organs at risk (OAR). Spearman’s correlation coefficient between EDIC and
lymphocyte nadir was analysed.
Further univariable and multivariable linear regression analyses were used to correlate
other variables with lymphocyte nadirs. PTV was converted into log(PTV) to obtain
normal distribution.[7] Parameters with p-values <0.1 in univariable model were
considered for multivariable analysis. Normal distribution assumption was tested
11
using using Shapiro-Wilk normality test. The significance of low dose lung dosimetry
with lymphocyte nadirs was similarly analysed.
For secondary endpoint of survival, Cox regression was used to study the prognostic
factors (including lymphocyte nadir) of overall survival (OS) and recurrence–free
survival (RFS). Similar to linear regression, the multivariable analysis included
parameters with p-values <0.1 in univariable models. We also performed survival
analysis in different lymphocyte nadir and EDIC subgroups. Statistical analyses were
conducted by Statistical Package for the Social Sciences (SPSS) version 25.0 (IBM)
and R version 3.5.1. P less than 0.05 was considered significant.
Results
Patient characteristics
Ninety–two patients were eligible for final analysis. Baseline demographics, tumour
and treatment characteristics of these patients are listed in Table 1. All patients were
ethnic Chinese. Around half of the patients (54.3%) were of age 65 or above, and 84%
were male. All 92 patients had squamous cell carcinoma, which reflected the disease
pattern in XXX as opposed to the Western population where adenocarcinoma
12
predominates. Seventy-nine patients (86%) had normal baseline lymphocyte counts
before chemoradiation. All patients but one (99%) had T3 disease and all of them had
at least one regional lymph node involvement (N1: 50%, N2 42.4% and N3 7.6%).
Only 60.8% of patients finished the planned 5 cycles of weekly chemotherapy (4
cycles = 30.4%; 3 cycles = 7.6%; 2 cycles = 0%; 1 cycle = 1.1%) and the commonest
reason for non-compliance was suboptimal marrow tolerance. More patients were
treated with 3D conformal technique than with IMRT technique (56.5% vs 40%).
EDIC and lymphocyte nadir
Univariable and multivariable linear regressions demonstrated that larger PTV volume
was significantly associated with lower lymphocyte nadirs (Table 2), in which
normality was not rejected for the Studentized residuals of the multiple linear
regression with p=0.44 using Shapiro-Wilk normality test. In addition, baseline
lymphocyte count was also associated with lower lymphocyte nadir. No other
demographic and clinical parameter correlated significantly with lymphocyte nadir.
Both radiotherapy technique and the number of courses of chemotherapy did not
affect lymphocyte nadirs.
13
The result of primary outcome was shown in Table 3. The estimated EDIC, which is a
function of mean heart dose, mean lung dose and mean doses to large vessels and
other thoracic organs (approximated by mean total body dose), had a significantly
negative correlation with the lymphocyte nadir (Spearman Coefficient = -0.505; P <
0.01). Individual mean doses to the lung, heart and total body were also highly
significant in predicting lymphocyte nadirs (Spearman coefficients = -0.34, -0.502
and -0.36 respectively; P < 0.01).
Lymphocyte nadir and low dose bath in lung
Previous study has established the correlation between low dose bath to lung and
lymphocyte nadir in non-small cell lung cancer [11]. We performed a similar analysis
to see if this relationship also exists in our patients with ESCC. Spearman’s
correlation coefficients (R) between lymphocyte nadir and different lung dose-volume
histogram (DVH) parameters, namely, V1 to V40, were shown in Figure 1. The
coefficients were highly significant at low dose lung DVH zone from V1 to V25
(Coefficient from -0.47 to -0.27; P < 0.05). The coefficients lost statistical
significance at V30 level onward (P > 0.05). The absolute values of correlation
coefficients decreased together with incremental increase in P value from low dose
14
zone level (V1) to high dose zone level (V40).
Lymphocyte nadir, EDIC and survivals
Our secondary outcome was the survival impact of lymphocyte nadir and EDIC.
Median overall survival (OS) for the whole population was 17.5 months (95% CI
9.30–25.77 months) after a median follow up of 16.9 months (range 1.3–79.3).
Univariable and multivariable Cox regression results of clinical variables on OS and
recurrence–free survival (RFS) are shown in Table 4. Multicollinearity was not found
with variance inflation factor (VIF) 1.18 in either OS and PFS. Multivariable models
suggested that lymphocyte nadir was the only variable with statistical significance
(p<0.05) in both OS and RFS. Higher lymphocyte nadir was found to have lower risk
on both OS and RFS (hazard ratio [HR] = 0.75 per 10
8
cells/L; P =0.003, and 0.78 per
10
8
cells/L; P =0.022 respectively) after adjustment in the multivariable Cox
regression models. log PTV, in contrast to common belief, did not reach statistically
significant effect on both OS and RFS in both univariable and multivariable models.
Nadirs of total white cell count, age, Sex, radiotherapy techniques, number of courses
of chemotherapy given, and N stage did not have impact on survival.
15
We then performed further analysis to explore relationships between overall survival
with different groups of lymphocyte nadir and EDIC. Following previously reported
studies [5]
we used lymphocyte count 0.5 (10
9
cells/L) as cut-off as reported The
Kaplan Meier (KM) curve is shown in Figure 2a. There is a trend of better survival for
the group with lymphocyte nadir 0.5 than the group < 0.5 (P = 0.192).
EDIC of our 92 patients ranged from 0.64 to 4.39 Gy. We divided the patients into
three groups according to EDIC value, namely <2 Gy, 2 – <4 Gy and 4 Gy. There
was significant difference in overall survival with different EDIC groups, with higher
EDIC predicting poorer survival (P = 0.01; Figure 2b). In addition, higher EDIC also
predicted lower probability of 2–year overall survival.
Discussion
Our study demonstrated the importance of lymphocyte count in determining the
outcomes of locally advanced ESCC and advised additional dose constraints in RT
planning. Lower lymphocyte nadir was associated with poorer OS and RFS in this
group of patients. Similar results have been shown in other solid tumours including
lung, pancreatic, head and neck and glioblastoma [5]. In our study, the HR of
lymphocyte nadir on OS is 0.63 per 10
5
lymphocytes/mL (or 10
8
cells/L). In other
16
words, there is 37% relative reduction in death per 10
5
lymphocytes/mL (or 10
8
cells/L) increment in lymphocyte nadir. The size of the effect is comparable to similar
study on NSCLC reported by Tang et al [7], in which the corresponding HR is 0.51
per 10
3
lymphocytes/mL (P = 0.01) on univariate analysis. Contrary to previous
reports, PTV was not a predictor of survivals with statistical significant after adjusting
of lymphocyte nadirs in the multivariable in Table 4. log PTV has large HR (>6) but
not statistically significant (p=0080 / 0.10 in uni-/multi-variable for OS). Similarly in
RFS, log PTV was close to statistically significant with p values =0.086 and 0.15,
respectively. The findings suggested that log PTV might have a trend in clinical
significance with high HR but cannot reach the statistical significance. We believe that
PTV effect on survivals may likely be indirect and PTV might exert its effect on
survivals through its contribution to lymphocyte nadirs.
Interestingly, the large scale study INT–0123 published in 2002 proved that there was
no survival benefit and local regional control of oesophageal cancer by escalating the
radiation dose above 50.4Gy [18]
. Despite controversies on the trial design and RT
techniques adopted in that study, our results may uncover the missing link between
RT dose and survival. Higher radiation dose might have detrimental effect on OS due
to lymphocyte killing. A recent analysis of radiation dose escalation in oesophageal
17
cancer [19] concluded that while local control might benefit from dose escalation,
there was no benefit in OS. Similar result was also shown in the dose escalation study
RTOG 0617 for NSCLC. The study [20]
showed an unexpected detrimental effect on
OS for higher dose of 74Gy compared to the control arm of 60Gy. This was
postulated to be due to lower lymphocyte nadir in higher dose arm by XXXX et al
[11]. As both Gross Tumour Volumes (GTV) and Planning Target Volumes (PTV) of
oesophageal cancer and NSCLC are mainly confined to the thorax, and lymphopenia
was shown to affect the outcomes in both cases, it is reasonable to hypothesize that
the lack of survival benefit with dose escalation in ESCC might be due to higher dose
to immune cells.
Low dose parameters of lung DVH (namely, V1–V25) are relevant to the survival of
circulating lymphocyte as almost all the circulating blood go through the pulmonary
circulation. Lung V5 has been shown to be correlated with survival in NSCLC [7].
The low dose bath could have effectively killed off a large number of circulating
lymphocytes. Similarly, PTV was shown to correlate with lymphocyte nadir. This was
likely caused by a larger radiation field resulting in more circulating immune cells
being exposed to radiation. We postulate that it might impair anti–tumour immunity
18
and decrease the ability to mount immunity response to infection. Thus, it might
explain a poorer survival in subjects with lower lymphocyte nadir.
Besides the lung DVH, dose to other previously undefined organs and tissue might
also contribute to the lymphocyte drop. For instance, the heart, large vessels and the
thoracic duct, which house the return of lymphocytes into systemic circulation, are all
in the thorax. In addition, the atrophic thymus in adult could have retained some
lymphatic function as well. Validated mathematical model is needed to account for
dose received by these organs in the thorax.
Therefore, we adopted the formula of predicting EDIC from our co–authors (JJ and
SK) in their study on lymphopenia and survival in NSCLC in RTOG 0617 [11]. We
found that EDIC strongly correlated with the lymphocyte nadir with Spearman
coefficient of -0.505 (P < 0.01). In our study, EDIC ranged from 0.64 to 4.39Gy
which was significantly lower than that reported in the RTOG 0617 cohort (2.05–
12.20Gy), this can be explained by a much lower dose of 41.4Gy prescribed in our
study according to the Dutch CROSS trial. Despite the difference of EDIC range, both
studies demonstrated the importance of EDIC in determining the lymphocyte nadir.
19
Besides, Ladbury et al[21] published their results in adopting the EDIC formula on
stage III non-small cell lung cancer patients in the International Journal of Radiation
Oncology·Biology·Physics. They retrospectively reviewed 117 patients and calculated
their EDIC. They found that EDIC was independently associated with overall survival
(HR 1.17, P = 0.03), local progression-free survival (HR 1.17, P = 0.02), and
disease-free survival (HR 1.15, P = 0.04). As both lung cancers and esophageal
cancers are mainly located within the thoracic, we believe that EDIC formula would
also be a valid model in esophageal cancer too. On the other hand, Saito et al[22]
found that splenic dose-volumes but not bone marrow dose-volumes were predictive
in treatment-related lymphopenia during chemoradiotherapy for esophageal cancer.
We believe future studies can incorporate dose to the spleen as a better estimate of
dose to EDIC.
While lymphocyte nadir has been proven to affect OS in multivariable Cox regression,
the Kaplan Meir survival curve only showed a trend of better survival in lymphocyte
nadir 0.5 compared with that of < 0.5. We think that given the 0.5 cut-off as
reported by other studies is arbitrary, a significant KM curve may be shown with other
20
lymphocyte cut-offs or with larger sample size. Furthermore, we were able to show
significant difference in 2–year survival probability in different EDIC subgroups. This
was in line with the RTOG0617 cohort.
Similar studies have been reported by Fang et al [13]
from the MD Anderson Cancer
Centre. In their cohort of 313 oesophageal cancer patients who received neoadjuvant
chemoradiotherapy, a higher lymphocyte nadir correlated with a higher pathological
complete response rate (OR = 1.82, P <0.003). They also found that mean body dose
was inversely related to high ALC nadir (OR = 0.77 per Gy, P <0.001). Although this
is consistent with our finding, there are substantial differences between the two
studies. Ninety-five percent of patients were adenocarcinoma in their study, and there
was significant heterogeneity in terms of radiotherapy dose, use of neoadjuvant
chemotherapy and chemotherapy regimens in their concurrent chemoradiotherapy
phase. In our study, all patients had squamous cell carcinoma, and all received the
same chemotherapy and radiotherapy dose.
While lower lymphocyte count predicts worse survival outcomes in other solid
tumours, there have been contradicting reports in the literature for oesophageal cancer.
21
One study [2] in the United States showed that in stage I–III oesophageal cancer
patients (85% adenocarcinoma), the lymphopenia group (defined as lymphocyte < 0.5
x 10
9
/L has a higher risk of death (HR for death 1.6; P = 0.027) compared with the
non–lymphopenia group. This is supported by another study [23] comparing proton
beam therapy with IMRT in 448 stage I-IVA oesophageal cancer patients, which
showed improved survival with higher lymphocyte count (HR for survival = 1.551, P
= 0.01 per 1 unit of lymphocyte). However, another study [24] showed that in 395
stage I–III oesophageal cancer patients, 5–year OS was not significantly different
between patients with grade 4 and non–grade 4 lymphopenia (34% vs 41%; P = 0.47).
As from the Cox regression model in our study, lymphocyte nadir strongly predicts
survivals. We believe the inconsistencies were mostly due to the arbitrary cut-off
value of lymphocyte count and statistical power of respective sample sizes [24].
Some other groups have proposed special radiation techniques to preserve lymphocyte
in other solid tumours. It involves the use of stereotactic body irradiation (SBRT) [16]
to minimize the number of fractions and the PTV, in order to reduce the low dose bath
effect to circulating lymphocyte. Use of proton therapy instead of photon has also
be suggested [23]. As there is minimal exit dose beyond the Bragg Peak, the total
22
integral dose is reduced in proton therapy. This has been shown in the MD Anderson
patient cohort in which proton therapy was a predictor of higher lymphocyte nadir
(compared with IMRT, OR = 4.18; P <0.001). However, neither of these was standard
for radiotherapy in esophageal cancer. Drug therapy in preserving circulating
lymphocyte is also an attractive option. However, there is no well-established therapy
at the time of writing.
Our study was unique that the tumour histology of ESCC and treatment regimens of
the Dutch CROSS were homogenous, in contrast to many other studies in the US in
which adenocarcinoma predominated. Our hospital is the major tertiary referral centre
for esophageal cancer patients in XXX which has a population of over 7 million and
we have included all locally advanced ESCC treated during the period. We believe the
results could be generalized to Asian patients in whom ESCC predominates.
Our study shared the intrinsic weakness of all retrospective studies. We minimized the
selection bias by including only patients with ESCC treated by the Dutch CROSS
regimen. Another limitation was that the lymphocyte count obtained was total
lymphocyte count. Lymphocyte subsets such as CD4+ve and CD8+ve cell counts
23
were not available. Future prospective trial shall address this issue by collecting
weekly cell counts of lymphocyte subsets.
Conclusion
Higher EDIC is associated with lower lymphocyte nadir and lymphocyte nadir
predicts OS. These findings shall be confirmed with larger scale prospective data in
future studies. Perhaps, the retrospective analysis of dosimetry data and lymphocyte
nadirs in INT–0123 trial [18] and CROSS trial [15] might be able to validate our
finding.
List of abbreviations
ALC - absolute lymphocyte counts
DVH - dose volume histogram
EDIC - effective dose to circulating immune cells
ESCC - oesophageal squamous cell carcinoma
24
EUD - equivalent uniform dose
GTV - Gross Tumour Volume
IMRT - Intensity modulated radiotherapy
ITD - Integral dose
NSCLC - non–small cell lung cancer
OAR - organs at risk
OS - overall survival
PTV - Planning Target Volume
RFS - recurrence–free survival
25
SBRT - stereotactic body radiation therapy
Declarations
Ethics approval and consent to participate
Approval was obtained by the Institutional Review Board of the University
of XXX
Consent was obtained from each subject
References
[1] Sellins KS, Cohen JJ. Gene induction by gamma-irradiation leads to DNA
fragmentation in lymphocytes. J Immunol. 1987;139:3199-206.
[2] Davuluri R, Jiang W, Fang P, Xu C, Komaki RU, Hsu CC, et al. Absolute Lymphocyte
Count Nadir During Chemoradiation as a Prognostic Indicator of Esophageal Cancer
Survival Outcomes. International Journal of Radiation Oncology • Biology • Physics.
2016;96:E177.
[3] Karantanos T, Karanika S, Seth B, Gignac G. The absolute lymphocyte count can
predict the overall survival of patients with non-small cell lung cancer on nivolumab:
26
a clinical study. Clin Transl Oncol. 2019;21:206-12.
[4] Pike LRG, Bang A, Mahal BA, Taylor A, Krishnan M, Spektor A, et al. The Impact of
Radiation Therapy on Lymphocyte Count and Survival in Metastatic Cancer Patients
Receiving PD-1 Immune Checkpoint Inhibitors. Int J Radiat Oncol Biol Phys.
2019;103:142-51.
[5] Grossman SA, Ellsworth S, Campian J, Wild AT, Herman JM, Laheru D, et al.
Survival in Patients With Severe Lymphopenia Following Treatment With Radiation
and Chemotherapy for Newly Diagnosed Solid Tumors. J Natl Compr Canc Netw.
2015;13:1225-31.
[6] Mendez JS, Govindan A, Leong J, Gao F, Huang J, Campian JL. Association between
treatment-related lymphopenia and overall survival in elderly patients with newly
diagnosed glioblastoma. J Neurooncol. 2016;127:329-35.
[7] Tang C, Liao Z, Gomez D, Levy L, Zhuang Y, Gebremichael RA, et al. Lymphopenia
association with gross tumor volume and lung V5 and its effects on non-small cell
lung cancer patient outcomes. Int J Radiat Oncol Biol Phys. 2014;89:1084-91.
[8] Chadha AS, Liu G, Chen HC, Das P, Minsky BD, Mahmood U, et al. Does
Unintentional Splenic Radiation Predict Outcomes After Pancreatic Cancer Radiation
Therapy? Int J Radiat Oncol Biol Phys. 2017;97:323-32.
27
[9] Campian JL, Sarai G, Ye X, Marur S, Grossman SA. Association between severe
treatment-related lymphopenia and progression-free survival in patients with newly
diagnosed squamous cell head and neck cancer. Head Neck. 2014;36:1747-53.
[10] Yovino S, Kleinberg L, Grossman SA, Narayanan M, Ford E. The etiology of
treatment-related lymphopenia in patients with malignant gliomas: modeling
radiation dose to circulating lymphocytes explains clinical observations and suggests
methods of modifying the impact of radiation on immune cells. Cancer Invest.
2013;31:140-4.
[11] XXX
[12] Davuluri R, Jiang W, Fang P, Xu C, Komaki R, Gomez DR, et al. Lymphocyte Nadir
and Esophageal Cancer Survival Outcomes After Chemoradiation Therapy. Int J Radiat
Oncol Biol Phys. 2017;99:128-35.
[13] Fang P, Jiang W, Davuluri R, Xu C, Krishnan S, Mohan R, et al. High lymphocyte
count during neoadjuvant chemoradiotherapy is associated with improved
pathologic complete response in esophageal cancer. Radiother Oncol.
2018;128:584-90.
[14] Shiraishi Y, Fang P, Xu C, Song J, Krishnan S, Koay EJ, et al. Severe lymphopenia
during neoadjuvant chemoradiation for esophageal cancer: A propensity matched
28
analysis of the relative risk of proton versus photon-based radiation therapy.
Radiother Oncol. 2018;128:154-60.
[15] Shapiro J, van Lanschot JJB, Hulshof M, van Hagen P, van Berge Henegouwen MI,
Wijnhoven BPL, et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery
alone for oesophageal or junctional cancer (CROSS): long-term results of a
randomised controlled trial. Lancet Oncol. 2015;16:1090-8.
[16] Crocenzi T, Cottam B, Newell P, Wolf RF, Hansen PD, Hammill C, et al. A
hypofractionated radiation regimen avoids the lymphopenia associated with
neoadjuvant chemoradiation therapy of borderline resectable and locally advanced
pancreatic adenocarcinoma. J Immunother Cancer. 2016;4:45-.
[17] Doi T, Bennouna J, Shen L, Enzinger PC, Wang R, Csiki I, et al. KEYNOTE-181:
Phase 3, open-label study of second-line pembrolizumab vs single-agent
chemotherapy in patients with advanced/metastatic esophageal adenocarcinoma.
Journal of Clinical Oncology. 2016;34:TPS4140-TPS.
[18] Minsky BD, Pajak TF, Ginsberg RJ, Pisansky TM, Martenson J, Komaki R, et al. INT
0123 (Radiation Therapy Oncology Group 94-05) phase III trial of combined-modality
therapy for esophageal cancer: high-dose versus standard-dose radiation therapy. J
Clin Oncol. 2002;20:1167-74.
29
[19] Brower JV, Chen S, Bassetti MF, Yu M, Harari PM, Ritter MA, et al. Radiation Dose
Escalation in Esophageal Cancer Revisited: A Contemporary Analysis of the National
Cancer Data Base, 2004 to 2012. Int J Radiat Oncol Biol Phys. 2016;96:985-93.
[20] Bradley JD, Paulus R, Komaki R, Masters G, Blumenschein G, Schild S, et al.
Standard-dose versus high-dose conformal radiotherapy with concurrent and
consolidation carboplatin plus paclitaxel with or without cetuximab for patients with
stage IIIA or IIIB non-small-cell lung cancer (RTOG 0617): a randomised, two-by-two
factorial phase 3 study. Lancet Oncol. 2015;16:187-99.
[21] Ladbury CJ, Rusthoven CG, Camidge DR, Kavanagh BD, Nath SK. Impact of
Radiation Dose to the Host Immune System on Tumor Control and Survival for Stage
III Non-Small Cell Lung Cancer Treated with Definitive Radiation Therapy. Int J Radiat
Oncol Biol Phys. 2019;105:346-55.
[22] Saito T, Toya R, Yoshida N, Shono T, Matsuyama T, Ninomura S, et al. Spleen
Dose-Volume Parameters as a Predictor of Treatment-related Lymphopenia During
Definitive Chemoradiotherapy for Esophageal Cancer. In vivo (Athens, Greece).
2018;32:1519-25.
[23] Fang P, Shiraishi Y, Verma V, Jiang W, Song J, Hobbs BP, et al. Lymphocyte-Sparing
Effect of Proton Therapy in Patients with Esophageal Cancer Treated with Definitive
30
Chemoradiation. Int J Part Ther. 2018;4:23-32.
[24] Zhang M, Oh P, Brady P, Ilson DH, Janjigian Y, Ku G, et al. Lack of validation of
lymphopenia as a prognostic factor in esophageal cancer chemoradiation.
International Journal of Radiation Oncology • Biology • Physics. 2018;102:e46.
31
Figure legends.
Figure 1. Lung Dose-Volume Histogram and correlation with lymphocyte nadir
Lymphocyte nadir (primary outcome) was correlated negatively with low dose lung
dosimetry only reflecting dose to circulating lymphocyte. Significance lost above
V25.
Figure 2 Overall survival relationship with (a) lymphocyte nadir and (b) EDIC
groups
32
Table 1 – Baseline characteristics
Table 2
Univariable and multivariable linear regression associating baseline variables
with lymphocyte nadirs (10
9
cells/L) during radiation treatment
Table 3: OAR Dosimetry and EDIC versus lymphocyte nadir
Table 4 Cox regression association with outcomes during radiation treatment
Table 1 – Baseline characteristics
Characteristics N = 92 (%)
Age at diagnosis
Under 65 42 (45.7%)
65 and above 50 (54.3%)
Sex
Male 77 (83.7%)
Female 15 (16.3%)
Baseline lymphocyte level
Normal 79 (85.9%)
Low 13 (14.1%)
RT technique
3DCRT 52 (56.5%)
IMRT 40 (43.5%)
T-staging
T1 0 (0%)
T2 1(1.1%)
T3 91 (98.9%)
T4 0 (0%)
N-staging
N0 0 (0)
N1 46 (50%)
N2 39(42.4%)
N3 7 (7.6%)
RT, radiation therapy; 3DCRT, 3-dimensional conformal radiation
therapy; IMRT, intensity-modulated radiation therapy
Number of chemotherapy courses
5 56 (60.8%)
4 28 (30.4%)
3 7 (12.5%)
2 0 (0%)
1 1 (1.1%)
EDIC group
<2 Gy 18 (19.6%)
2 - <4 Gy 68 (73.9%)
>- 4Gy 6 (6.5%)
Table 3: OAR Dosimetry and EDIC versus Lymphocyte nadir
EDIC= effective dose to the circulating immune cells
Spearman coefficient P
mean total body dose -0.36 < 0.001
mean lung dose -0.34 0.01
mean heart dose -0.502 < 0.001
EDIC -0.505 < 0.01
Table 2
Univariable and multivariable linear regression associating baseline variables with lymphocyte nadirs (10
9
cells/L)
during radiation treatment
Univariable analysis Multivariable analysis
Characteristic
Regression
coefficient 95% CI P
Regression
coefficient 95% CI P
Male (vs female) 0.048 -0.039 to 0.136 0.278
NI
Age (1 y) -0.001 -0.005 to 0.002 0.537
NI
Stage
N3 (vs N2 and N1) -0.085 -0.206 to 0.037 0.168
NI
Baseline lymphocyte
(10
9
cells/L) 0.112 0.068 to 0.156 <0.001 0.098 0.060 to 0.135 <0.001
Chemotherapy
5 courses
(vs fewer than 5 courses) 0.060 -0.005 to -0.126 0.071 0.044 –0.005 to 0.093 0.081
Radiation therapy
log PTV -0.393 -0.629 to -0.158 0.001
–0.205
–0.399 to –0.011
0.038
EDIC -0.085 -0.115 to -0.055 <0.001 –0.061
–0.089 to –0.034
<0.001
IMRT (vs 3DCRT) 0.019 -0.047 to 0.084 0.567
NI
Abbreviations: 3DCRT, 3-dimensional conformal radiotherapy; CI, confidence interval; IMRT, intensity-modulated radiotherapy; NI, not
included; PTV, planning target volume.
Table 4
Cox regression association baseline variables with outcomes during radiation treatment
Overall survival Univariable Multivariable
Variables HR 95% CI P HR 95% CI P
Lymphocyte nadir (10
cells/L) 0.72 0.58 to 0.89 0.003 0.75 0.58 to 0.96 0.022
Age (1 y) 0.99 0.97 to 1.02 0.68 NI
WCC nadir (10
cells/L) 1.02 0.83 to 1.27 0.84 NI
Male (vs female) 1.002 0.51 to 1.98 0.995 NI
Stage
N3 (vs N2 and N1) 0.66 0.24 to 1.83 0.43 NI
Baseline lymphocyte 0.56 0.36 to 0.87 0.010 0.67 0.41 to 1.11 0.12
Chemotherapy
5 courses (vs <5 courses) 0.66 0.40 to 1.10 0.11 NI
Radiation therapy
log
10
PTV 6.41 0.80 to 51.16 0.080 5.32 0.56 to 50.51 0.15
EDIC 1.18 0.89 to 1.57 0.25 NI
IMRT (vs 3DCRT) 0.74 0.44 to 1.24 0.25 NI
Recurrence-free survival Univariable Multivariable
Variables HR 95% CI P HR 95% CI P
Lymphocyte nadir (10
8
cells/L) 0.74 0.60 to 0.91 0.004 0.78 0.62 to 0.99 0.043
Age (1 y) 0.99 0.96 to 1.01 0.30 NI
WCC nadir (10
9
cells/L) 1.01 0.82 to 1.23 0.96 NI
Male (vs female) 1.14 0.58 to 2.24 0.71 NI
Stage
N3 (vs N2 and N1) 0.60 0.22 to 1.64 0.32 NI
Baseline lymphocyte 0.52 0.33 to 0.82 0.005 0.63 0.37 to 1.06 0.079
Chemotherapy
5 courses (vs <5 courses) 0.72 0.44 to 1.19 0.20 NI
Radiation therapy
log
10
PTV 6.07 0.76 to 47.45 0.086 4.79 0.52 to 44.47 0.17
EDIC 1.19 0.91 to 1.56 0.21 NI
IMRT (vs 3DCRT) 0.70 0.43 to 1.18 0.19 NI
Abbreviations: 3DCRT, three-dimensional conformal radiation therapy; CI, confidence interval; EDIC, effective dose to the circulating immune
cells; IMRT, intensity-modulated radiation therapy; NI, not included; PTV, planning target volume; WCC, white cell count.
Figure 1 Lung Dose Volume Histogram and correlation with lymphocyte nadir
Lymphocyte nadir(primary outcome) is correlated negatively with low dose lung
dosimetry only reflecting dose to circulating lymphocyte. Significance lost above
V25
† P < 0.001, ** P < 0.01, * P < 0.05
This is the correlation coefficient plot showing decreasing coefficient value at higher dose
lung DVH and lost statistical significance after 25 Gy.
**
** **
**
-0.5
-0.45
-0.4
-0.35
-0.3
-0.25
-0.2
-0.15
-0.1
-0.05
0
0 10 20 30 40 50
Spearman's Correlation coefficient
Percent Lung Dose (Gy)
Figure 2 Exploratory Analysis
To determine overall survival relationship with lymphocyte nadir and EDIC groups
Figure 2a
OS difference between two groups (lymphocyte nadir < 0.5 or 0.5)
Log rank P = 0.192
Figure 2b
OS difference between high and lower EDIC
Log rank P = 0.010
(EDIC range 0.64 to 4.39 Gy)
EDIC= effective dose to the circulating immune cells
... In the same time, despite all the measures taken to protect normal tissues from exposure to ionizing radiation, there is still a risk of developing complications during RT, including complications caused by the destruction of radiosensitive cells of the immune system (IS) circulating through the radiation portal. Many researchers have reported the development of radiation-induced lymphopenia (RIL) as one of the main side effects of remote RT for oncological diseases, including BC [3][4][5][6][7][8]. ...
... IS dysfunction not only plays an important role in the pathogenesis of the initial response of human tissues to irradiation, but it is also an unfavorable prognostic factor for the overall survival of patients with solid tumors, which may result from altered antitumor immunity [7][8][9][10]. ...
Article
Full-text available
Background: Radiation therapy (RT) is an important step in the treatment of primary breast cancer as it is one of the leading contributors to cancer incidence among women. Most patients with this disease acquire radiation-induced lymphopenia in the early post-radiation period; however, little is known about the effect of RT on the composition of lymphocyte populations in such patients. This study was aimed at investigating the effect of adjuvant remote RT-performed in the classical mode for patients with primary breast cancer-on the main components of cell-mediated immunity (major lymphocyte populations), including those in patients receiving chemotherapy. Methods: Between 2020 and 2022, 96 patients with stage I-III breast cancer were included in this study. All patients in the final stage of complex treatment received RT via a 3D conformal technique (3DCRT). The clinical target volume of this RT included the breast or chest wall and locoregional lymphatics. Flow cytometry was used to assess the levels and phenotypes of circulating lymphocytes before and after RT (no more than 7 days before and after RT). The evaluation of the impact of polychemotherapy (PCT) was conducted to determine whether it was a risk factor for the onset of radio-induced lymphopenia (RIL) in the context of RT. Results: When assessing the immune status in the general group of patients (n = 96), before the start of adjuvant external beam radiotherapy (EBRT), the average number of lymphocytes was 1.68 ± 0.064 × 109/L; after the course of adjuvant EBRT, it decreased to 1.01 ± 0.044 × 109/L (p < 0.001). When assessing the absolute indicators of cellular immunity in the general group of patients with BC after a course of adjuvant EBRT, significant dynamics were revealed by the changes in all cell populations of lymphocytes (paired t-test, p < 0.05). Conclusion: The adaptive immune system in breast cancer patients changed in the early post-radiation period. The absolute levels of B-, T- and natural killer cells significantly reduced after RT regardless of whether the patients previously underwent chemotherapy courses. RT for patients with primary breast cancer should be considered in clinical management because it significantly alters lymphocyte levels and should be considered when assessing antitumor immunity, as significant changes in T-cell immunity have been observed. In addition, the identified changes are critical if specific targeted therapy or immunotherapy is needed.
... For lung cancer, SLP during CCRT was significantly associated with dismal survival outcomes, particularly diminishing the survival benefits of maintenance immune checkpoint inhibitors [17][18][19][20][21][22]. Also, SLP is related to poor RT response in gastrointestinal cancers (esophagus, rectum, pancreas, etc.) [23][24][25][26]. Several studies have highlighted the negative impact of SLP in patients with GBM [4,6,9]. ...
... However, most evidence is derived from clinical trials, especially data on the combination of chemotherapy and immunotherapy in ESCC, which is still in the experimental stage, lacking data from real-world clinical application. Additionally, the absolute lymphocyte count (ALC) has been identi ed as a crucial prognostic factor for malignant tumor patients, with clinical validation [10,11] . However, its predictive value for the prognosis of ESCC patients receiving ICIs in combination with CRT has been rarely reported. ...
Preprint
Full-text available
Backgroud: This study aimed to assess the effectiveness, safety, and recurrence patterns of first-line immunotherapy combined with chemoradiotherapy in esophageal squamous cell carcinoma (ESCC) patients. Methods:A retrospective analysis of 79 eligible ESCC patients was conducted. Primary outcomes included overall survival (OS) and progression-free survival (PFS), with secondary outcomes being objective response rate (ORR), disease control rate (DOR), treatment-related adverse events (trAEs), and treatment failure patterns. Results: The median follow-up was 29.4 months, with median OS unreached and median PFS of 14.6 months (95% CI: 10.7-18.5). ORR was 82.3%, and DOR was 96.2%. Factors affecting OS were clinical stage, immunotherapy cycles, immunotherapy and chemoradiotherapy sequence, radiation coverage, mid-treatment lymphocyte count, and short-term efficacy (HR=2.254, 0.374, 2.653, 2.957, 2.309, 2.789; P=0.030, 0.019, 0.009, 0.004, 0.001, 0.014). Factors impacting PFS were clinical stage, immunotherapy and chemoradiotherapy, and post-treatment lymphocyte count (HR=2.135, 2.048, 1.911; P=0.007, 0.010, 0.001). Among the cohort, 46.8% experienced treatment failure, with 33 receiving second-line treatment, resulting in a median OS of 14.17 months (95% CI: 7.303-21.037) and 1- and 2-year OS rates of 56.7% and 24.3%. Notably, 36.7% experienced grade ≥2 trAEs, bone marrow suppression most commonly happened, and 5.1% developed esophageal fistulas. Conclusion: Immunotherapy combined with chemoradiotherapy demonstrates strong anti-tumor activity and tolerability in ESCC patients, The radiation for all leisions, sequential immunotherapy combined with chemoradiotherapy, and higher levels of lymph node cell counts are associated with a better prognosis. Large-scale randomized controlled trials are needed for further validation.
Article
Lymphocytes, which are highly sensitive to radiation, play a crucial role in the body’s defense against tumors. Radiation-induced lymphopenia has been associated with poorer outcomes in different cancer types. Despite being the largest secondary lymphoid organ, the spleen has not been officially designated as an organ at risk. This study hypothesizes a connection between spleen irradiation and lymphopenia and seeks to establish evidence-based dosage limits for the spleen. We retrospectively analyzed data from 96 patients with locally advanced gastric cancer who received postoperative chemoradiotherapy (CRT) between May 2010 and May 2017. Complete blood counts were collected before, during and after CRT. We established a model for predicting the minimum absolute lymphocyte count (Min ALC) and to investigate potential associations between spleen dosimetric variables and Min ALC. The median follow-up was 60 months. The 5-year overall survival (OS) and disease-free survival (DFS) were 65.2% and 56.8%, respectively. The median values of pre-treatment ALC, Min ALC and post-treatment ALC were 1.40 × 109, 0.23 × 109 and 0.28 × 109/L, respectively. Regression analysis confirmed that the primary tumor location, number of fractions and spleen V5 were significant predictors of Min ALC during radiation therapy. Changes in ALC (ΔALC) were identified as an independent predictor of both OS and DFS. Spleen V5 is an independent predictor for Min ALC, and the maximum dose of the spleen is associated with an increased risk of severe lymphopenia. Therefore, these doses should be restricted in clinical practice. Additionally, ΔALC can serve as a prognostic indicator for adjuvant radiotherapy in gastric cancer.
Article
Purpose Emerging data have illuminated the impact of effective radiation dose to immune cells (EDIC) on outcomes in patients with locally advanced, unresectable non‐small cell lung cancer (NSCLC) treated with intensity-modulated radiotherapy (IMRT). Hypothesizing that intensity-modulated proton therapy (IMPT) may reduce EDIC versus IMRT, we conducted a dosimetric analysis of patients treated at our institution. Materials and Methods Data were retrospectively collected for 12 patients with locally advanced, unresectable NSCLC diagnosed between 2019 and 2021 who had physician-approved IMRT and IMPT plans. Data to calculate EDIC from both Jin et al (PMID: 34944813) and Ladbury et al’s (PMID: 31175902) models were abstracted. Paired t tests were utilized to compare the difference in mean EDIC between IMPT and IMRT plans. Results IMPT decreased EDIC for 11 of 12 patients (91.7%). The mean EDIC per the Jin model was significantly lower with IMPT than IMRT (3.04 GyE vs 4.99 Gy, P < .001). Similarly, the mean EDIC per the Ladbury model was significantly lower with IMPT than IMRT (4.50 GyE vs 7.60 Gy, P < .002). Modeled 2-year overall survival was significantly longer with IMPT than IMRT (median 71% vs 63%; P = .03). Conclusion IMPT offers a statistically significant reduction in EDIC compared to IMRT. Given the emergence of EDIC as a modifiable prognostic factor in treatment planning, our dosimetric study highlights a potential role for IMPT to address an unmet need in improving oncologic outcomes in patients with locoregionally advanced NSCLC.
Chapter
Recent studies have emphasized the importance of protecting thoracic duct during radiation therapy (RT), as dose distributions in thoracic duct may be associated with the development radiation-induced lymphopenia. Because of its thin/slim size, curved geometry and extremely poor (intensity) contrast of thoracic duct, manual delineation of thoracic duct in RT planning CT is time-consuming and with large inter-observer variations. In this work, we aim to automatically and accurately segment thoracic duct in RT planning CT, as the first attempt to tackle this clinically critical yet under-studied task. A two-stage coarse-to-fine segmentation approach is proposed. At the first stage, we automatically segment six chest organs and combine these organ predictions with the input planning CT to better infer and localize the thoracic duct. Given the coarse initial segmentation from first stage, we subsequently extract the topology-corrected centerline of initial thoracic duct segmentation at stage two where curved planar reformation (CPR) is applied to transform the planning CT into a new 3D volume representation that provides a spatially smoother reformation of thoracic duct in its elongated medial axis direction. Thus the CPR-transformed CT is employed as input to the second stage deep segmentation network, and the output segmentation mask is transformed back to the original image space, as the final segmentation. We evaluate our approach on 117 lung cancer patients with RT planning CT scans. Our approach significantly outperforms a strong baseline model based on nnUNet, by reducing 57% relative Hausdorff distance error (from 49.9 mm to 21.2 mm) and improving 1.8% absolute Jaccard Index.
Article
Full-text available
It is well known that radiation therapy causes lymphopenia in patients and that this is correlated with a negative outcome. The mechanism is not well understood because radiation can have both immunostimulatory and immunosuppressive effects. How tumor dose conformation, dose fractionation, and selective lymph node irradiation in radiation therapy does affect lymphopenia and immune response is an active area of research. In addition, understanding the impact of radiation on the immune system is important for the design and interpretation of clinical trials combining radiation with immune checkpoint inhibitors, both in terms of radiation dose and treatment schedules. Although only a few percent of the total lymphocyte population are circulating, it has been speculated that their increased radiosensitivity may contribute to, or even be the primary cause of, lymphopenia. This review summarizes published data on lymphocyte radiosensitivity based on human, small animal, and in vitro studies. The data indicate differences in radiosensitivity among lymphocyte subpopulations that affect their relative contribution and thus the dynamics of the immune response. In general, B cells appear to be more radiosensitive than T cells and NK cells appear to be the most resistant. However, the reported dose-response data suggest that in the context of lymphopenia in patients, aspects other than cell death must also be considered. Not only absolute lymphocyte counts, but also lymphocyte diversity and activity are likely to be affected by radiation. Taken together, the reviewed data suggest that it is unlikely that radiation-induced cell death in lymphocytes is the sole factor in radiation-induced lymphopenia.
Article
Background: The immune system may influence prognosis, and lymphopenia is a frequent side effect of concurrent chemoradiotherapy (CCRT). Radical irradiation for locally advanced esophageal cancer (LA-EC) exposes significant vascular and heart volumes. In this study, we hypothesized that lymphopenia is linked to cardiac and pericardial doses and affects patient prognosis. Methods and materials: We identified 190 LA-EC patients who received radical CCRT. Multivariate analysis (MVA) was performed to correlate clinical factors and dosimetric parameters with overall survival (OS). We collected lymphocyte-related variables and ratios before and during CCRT. MVA was performed to correlate hematologic toxicity with OS. The relationship between dosimetric parameters and G4 lymphopenia was determined using logistic stepwise regression. Finally, a nomogram of G4 lymphopenia was developed and validated externally. Results: Median follow-up time for all patients was 27.5 months. On MVA for OS, higher pericardial V30 (PV30 ) was linked to worse survival (HR: 1.013, 95% CI: 1.001-1.026, p = 0.039). The median OS stratified by PV30 > 55.3% and PV30 ≤ 55.3% was 24.0 months and 54.0 months, respectively (p = 0.004). G4 lymphopenia was shown to be linked with worse OS in the MVA of hematological toxicity with OS (HR: 2.042, 95% CI: 1.335-3.126, p = 0.001). Thirty of the 100 patients in the training set had G4 lymphopenia. Logistic stepwise regression was used to identify variables associated with G4 lymphopenia, and the final model consisted of stage-IVA (p = 0.017), platelet-to-lymphocyte ratio during CCRT (p = 0.008), Heart V50 (p = 0.046), and PV30 (p = 0.048). Finally, a nomogram predicting G4 lymphocytopenia were constructed and externally validated. The ROC curve showed an AUC for internal validation of 0.775 and external validation of 0.843. Conclusion: Higher doses of pericardial radiation might affect LA-EC patients' prognosis by inducing G4 lymphopenia during CCRT. Further prospective studies are warranted to confirm these findings, especially in the era of immune-checkpoint inhibitor treatment.
Article
Full-text available
Aim: Our study sought to identify dosimetric predictors of treatment-related lymphopenia during chemoradiotherapy for esophageal cancer. Materials and methods: Patients with esophageal cancer who had received definitive chemoradiotherapy at our Institution were retrospectively assessed. The absolute volume of the spleen, body, and bone marrow that had received 5, 10, 20, and 30 Gy and the mean splenic dose were recorded. Results: Multivariate linear regression analysis revealed that docetaxel use and spleen dose-volume parameters (V5, V10, V20, V30, and mean splenic dose) were significant independent factors negatively influencing the absolute lymphocyte count at nadir. An increase of 1 Gy in mean splenic dose predicted a 2.9% decrease in nadir absolute lymphocyte count. Univariable logistic regression analysis showed that the mean splenic dose was a significant predictor of grade 4 lymphopenia. None of the body or bone marrow dose-volume parameters significantly predicted lymphopenia. Conclusion: Higher spleen dose-volume parameters were associated with severe lymphopenia during chemoradiotherapy.
Article
Full-text available
Purpose: To assess whether radiation treatment modality with proton beam therapy (PBT) or intensity-modulated radiation therapy (IMRT) is associated with lymphopenia in patients treated with definitive chemoradiation for esophageal cancer. Methods and materials: Patients with esophageal cancer treated with bimodality therapy (n = 448) between 2004 and 2016 were retrospectively reviewed. Patients treated with PBT were matched by propensity score with those treated with IMRT, based on key patient and disease factors, and stratified by clinical disease stage. Patients who developed early, distant metastatic disease within 1 month of completing radiation were excluded. Univariable and multivariable logistic regression were used to identify variables associated with increased risk of grade 4 lymphopenia. Multivariable Cox proportional hazards regression was used to assess factors associated with overall survival, disease-free survival, and locoregional relapse-free survival. Results: Patients who had IMRT and PBT matched by propensity score (n = 220) were not different with respect to age, sex, stage, performance status, tumor location, histology, tumor target volume, or induction chemotherapy. Treatment with IMRT, compared with PBT (odds ratio [OR], 2.13; 95% confidence interval [95% CI], 1.19-3.81; P = .01), increased age (OR, 1.039/y increase; 95% CI, 1.003-1.076; P = .03), and greater planning target volume (OR, 3.47 per 1-unit increase in log (planning target volume); 95% CI, 1.67-7.21; P < .001), was associated with increased risk of grade 4 lymphopenia. Radiation modality was associated with lymphocyte reduction in patients with tumors in the lower esophagus (P = .005) but not for those with tumors in the upper or middle esophagus (P = 0.32). Conclusions: In patients with esophageal cancer treated with definitive chemoradiation, PBT reduces the risk of severe, treatment-related lymphopenia, particularly in tumors of the lower esophagus.
Article
6068 Background: Severe treatment-related lymphopenia (TRL) occurs in 50% of glioblastoma and pancreatic cancer patients and is associated with early death from tumor progression. We sought to determine if similar findings were seen in head and neck squamous cell carcinoma (HNSCC). Methods: Eligible patients for this retrospective study had: 1) HNSCC cancer diagnosed in 2007-2009, 2) good performance status, 3) platinum-based chemoradiation, and 4) follow-up at Johns Hopkins. Serial total lymphocyte counts (TLC), overall survival (OS) and progression-free survival (PFS) were analyzed accounting for known prognostic factors. Results: Fifty-six adults met eligibility criteria: median age: 57 years, female: 21%, HPV+: 61%, surgery prior to chemoradiation: 16%, stage IVA-IVB: 77%, T stage 3-4: 40% and N stage 2b-3: 56%. Changes in TLC are shown below (Table). 14/56 patients (25%) had tumor recurrence and 9 (16%) died of their tumor. HPV+ patients had longer PFS (p=0.01) and OS (p=0.006) than HPV- patients. 10/22 HPV- patients had disease progression and 7 died. HPV- patients who developed grade III-IV TRL two months after beginning chemoradiation had a strikingly higher hazard rate for disease progression than those whose TLC remained higher (multivariate analysis HR 6.2, 95%CI: 1.1-35.2; p=0.039). Too few events occurred in the HPV+ cohort for analysis. Conclusions: TLCs were normal before chemoradiation. However, two months after chemoradiation ~60% of patients had severe TLC regardless of HPV status. HPV- patients with severe TRL were much more likely to have disease progression than those without TRL. Prospective studies are needed to confirm these findings which are similar to those reported in other cancers. Preservation of the immune system during chemoradiation may be important to improving PFS and OS. [Table: see text]
Article
Purpose: The significance of radiation dose to the host immune system during the treatment of stage III non-small cell lung cancer (NSCLC) is unknown, but higher doses were associated with worse tumor control and overall survival (OS) in a secondary analysis of RTOG 0617. In this study, we sought to assess the impact of the estimated dose of radiation to immune cells (EDRIC) on cancer-specific outcomes in an independent cohort of patients treated at our institution. Methods and materials: We retrospectively identified 117 patients with stage III NSCLC treated with definitive fractionated radiation from 2004 to 2017 at a single academic center (median dose of 60 Gy; 60% underwent intensity modulated radiation therapy and 92% received concurrent platinum-based chemotherapy). EDRIC was calculated as a function of the number of radiation fractions and mean doses to the lung, heart, and remaining body based on a model developed by Jin et al. Results: Median follow-up was 16 months with 77% of patients followed until death. In the entire population, 5-year OS was 11.2% with a median survival of 17.3 months. Median EDRIC for the entire cohort was 6.1 Gy (range, 2.5-10.0 Gy). A higher EDRIC was correlated with greater risk of grade ≥3 lymphopenia (P = .004). On multivariate analysis including total prescription radiation dose, planning target volume, and chemotherapy utilization, EDRIC was independently associated with OS (hazard ratio [HR] 1.17, P = .03), local progression-free survival (HR 1.17, P = .02), and disease-free survival (HR 1.15, P = .04). The median OS for patients with an EDRIC above 7.3 Gy (fourth quartile) and below 5.1 Gy (first quartile) was 14.3 and 28.2 months, respectively. Conclusions: Higher doses of radiation to the immune system were associated with tumor progression and death after the definitive treatment of stage III NSCLC. Tailoring radiation therapy to spare the immune system may be an important future direction to improve outcomes in this population.
Article
Purpose: Therapeutic radiation has conflicting immune effects: radiation (RT)-induced immunogenic cell death can contribute to immune response, but lymphocytes are also sensitive to RT. It is unknown whether palliative RT leads to lymphopenia in patients treated with immune checkpoint inhibitors (ICI) and whether this impacts outcomes. As such, we sought to assess the impact of palliative RT on circulating lymphocyte count and neutrophil-lymphocyte-ratio (NTL) in patients being treated with PD-1 directed ICI and associations with survival. Materials and methods: We identified patients from five radiation oncology centers, treated with palliative RT and either pembrolizumab or nivolumab with non-small cell lung cancer (NSCLC), metastatic melanoma (MM), and renal cell carcinoma (RCC). Patients who received intervening cytotoxic chemotherapy were excluded. We recorded absolute lymphocyte count (ALC) and neutrophil to lymphocyte ratio (NTL) before and after palliative RT, and at the start of ICI. Survival was analyzed using the Kaplan-Meier method and Cox proportional hazard models. Results: One hundred ten patients received 225 courses of palliative RT. Median change in ALC (dALC) after RT was -161 cells/mL. Decreases in ALC were greater with RT to the spine, lung/mediastinum, and chest wall compared with the brain, extremity, or abdomen/pelvis (p=0.002), and after courses >5 fractions (p=0.003). Extracranial and >5 fraction radiation was associated with increased odds of severe lymphopenia (ALC<500) at the end of RT (OR 3.7, p=0.001, and OR 3.9 p=0.001, respectively). Patients who developed RT-induced severe lymphopenia were more likely to have severe lymphopenia when ICI was initiated (OR 6.4, p=0.0001), particularly when RT was administered in the previous 3 months (OR 189, p<0.0001). Severe lymphopenia at onset of ICI therapy was associated with increased mortality on multivariable analysis (HR 2.1, p=0.03). Conclusions: Extracranial or prolonged courses of RT increase the risk of severe lymphopenia, which is associated with poorer survival in patients treated with ICI.
Article
Introduction: The neutrophil-to-lymphocyte (ANC/ALC) ratio is associated with worse prognosis in patients with NSCLC on immunotherapies, but the role of ALC remains unclear. The previous radiation therapy causes lymphopenia, and given approaches of combining radiation with immunotherapies, it is critical to better understand the impact of peripheral lymphocytes. Patients and methods: We evaluated retrospectively 22 patients with advanced NSCLC treated with nivolumab at Boston Medical Center from January 2014 to September 2016 and correlated the peripheral blood counts with the overall survival (OS) and overall time on treatment. We assessed the effect of the previous radiation on peripheral blood counts and clinical outcomes. Results: Baseline ALC and ANC/ALC ratios are positively and negatively correlated, respectively, with the OS on nivolumab. The ALC and ALC/WBC ratios at 6 weeks on treatment are positively associated with the OS. Kaplan-Meier analysis at baseline and at 6 weeks showed significantly increased OS in the group of patients with the highest ALC. The previous radiation therapy was positively correlated with the ANC and negatively correlated with the ALC/WBC ratio at 8 weeks after the initiation of nivolumab. Conclusion: Our finding that ALC at baseline and at 6 weeks on treatment is positively correlated with the OS provides an easily obtained predictive marker. Our result that the previous radiation is associated with higher ANC and lower ALC during treatment supports that the combination of radiation therapy with immunotherapy should be carefully applied and potentially peripheral blood counts can be utilized to stratify patients for this approach.
Article
Background and purpose: Neoadjuvant chemoradiation (nCRT) can reduce tumor infiltrating lymphocytes. We examined absolute lymphocyte count (ALC) nadir during nCRT for esophageal cancer (EC) and pathologic complete response (pCR). Materials and methods: Patients with stage I-IVA EC (n = 313) treated 2007-2013 with nCRT followed by surgery were analyzed. ALC was obtained before, during/weekly, and one month after CRT. pCR was defined as no viable tumor cells at surgery. High ALC was defined as nadir of ≥0.35 × 103/μL (highest tertile). Comparison of continuous and categorical variables by pCR was assessed by ANOVA and Pearson's chi-square. Univariate/multivariate logistic regression was used to assess predictors of pCR and high ALC nadir. Results: Eighty-nine patients (27.8%) achieved a complete pathological response (pCR). For patients with pCR, median ALC nadir was significantly higher than those without (0.35 × 103/μL vs 0.29 × 103/μL, p = 0.007). Patients maintaining high ALC nadir had a higher pCR rate (OR1.82, 95%CI 1.08-3.05, p = 0.024). Predictors of high ALC included treatment with proton therapy vs. IMRT (OR4.18, 95%CI 2.34-7.47, p < 0.001), smoking at diagnosis (OR2.80, 95%CI 1.49-5.25, p = 0.001), early stage I-II disease (OR2.33, 95%CI 1.32-4.17, p = 0.005), and SCC histology (OR3.70, 95%CI 1.01-14.29, p = 0.048). Mean body dose (MBD) was inversely related to high ALC nadir (OR0.77 per Gy, 95%CI 0.70-0.84, p < 0.001). Conclusion: A higher ALC level during nCRT is associated with a higher rate of pCR for esophageal cancer patients undergoing trimodality therapy.
Article
Background and purpose: Circulating lymphocytes are exquisitely sensitive to radiation exposure, even to low scattered doses which can vary drastically between radiation modalities. We compared the relative risk of radiation-induced lymphopenia between intensity modulated radiation therapy (IMRT) or proton beam therapy (PBT) in esophageal cancer (EC) patients undergoing neoadjuvant chemoradiation therapy (nCRT). Material and methods: EC patients treated with IMRT and PBT were propensity matched based on key clinical variables. Treatment-associated lymphopenia was graded using CTCAE v.4.0. Using matched cohorts, univariate and multivariable multiple logistic regression was used to identify factors associated with increased risk of grade 4 lymphopenia as well as characterize their relative contributions. Results: Among the 480 patients treated with nCRT, 136 IMRT patients were propensity score matched with 136 PBT patients. In the matched groups, a greater proportion of the IMRT patients (55/136, 40.4%) developed grade 4 lymphopenia during nCRT compared with the PBT patients (24/136, 17.6%, P < 0.0001). On multivariable analysis, PBT was significantly associated with a reduction in grade 4 lymphopenia risk (odds ratio, 0.29; 95% confidence interval, 0.16-0.52; P < 0.0001). Conclusion: PBT is associated with significant risk reduction in grade 4 lymphopenia during nCRT in esophageal cancer.