| Home | E-Submission | Sitemap | Contact Us |  
top_img
Radiation Oncology Journal > Volume 42(4); 2024 > Article
Koukourakis, Georgakopoulos, Desse, Tiniakos, Kouloulias, and Zygogianni: Lymphopenia is an adverse prognostic factor in rectal adenocarcinoma patients receiving long-course chemoradiotherapy

Abstract

Purpose

Neoadjuvant radiotherapy (RT) or chemoradiotherapy (CRT) is the standard treatment for locally advanced rectal adenocarcinoma. The recent emerging data on preoperative immunotherapy as an effective therapeutic modality for mismatch repair deficient rectal carcinomas suggests that the immune system plays a significant role in tumor eradication. Although RT has been shown to stimulate anti-tumor immunity, it also leads to substantial lymphopenia, hindering the effect of immune response.

Materials and Methods

We retrospectively analyzed 33 rectal adenocarcinoma patients who underwent CRT in our department, aiming to identify the effects of CRT on the peripheral blood lymphocyte counts (LC) and the potential impact of CRT-induced lymphopenia on tumor response and prognosis of patients.

Results

A statistically significant decrease in the LC of patients was observed after CRT (median values of 2,184/μL and 517/μL before and after treatment, respectively; p < 0.001). While no correlation between ypT-stage, ypN status, and LC was found, poor tumor regression grade was significantly associated with lower LC (p = 0.036). Moreover, lymphopenia was associated with poorer distant metastasis-free survival (p = 0.003). Distant metastases were documented in 0% of patients with post-CRT LC above 518/μL vs. 44.5% of patients with lower LC values.

Conclusion

Although further investigation is demanded, given the limited number of patients analyzed in the study, lymphopenia emerges as a significant adverse event that rectal adenocarcinoma patients face during treatment with neoadjuvant CRT, with subsequent implications on tumor response and prognosis. Protection of the immune system during CRT emerges as an important target for clinical research.

Introduction

Neoadjuvant radiotherapy (RT) with or without chemotherapy (chemoradiotherapy [CRT]) has been the gold standard treatment for locally advanced rectal adenocarcinoma for approximately 30 years. Ever since the publication of the results of the Swedish Rectal Cancer trial [1], short-course RT or long-course CRT has been utilized in the neoadjuvant setting with the goal of enhancing patients' prognosis and facilitating optimal surgical procedures. During the past 5 years, total neoadjuvant therapy—which incorporates a full preoperative chemotherapy course that is followed or preceded by RT or CRT—has emerged as the new standard treatment approach [2]. More recently, anti-PD1 immunotherapy is considered a first-line therapy for the treatment of mismatch-repair deficient locally advanced rectal adenocarcinoma [3], suggesting an important role of immunity in the control of this malignancy. Nevertheless, even in mismatch-repair proficient tumors preliminary results from clinical studies provide encouraging enhancement of pathological complete responses after combination of RT with immunotherapy [4].
Since a robust immune response is crucial for the outcome of CRT or immuno-CRT, immunocompromisation of any reason could potentially counteract treatment efficacy [5]. Strong experimental and clinical data support the concept of radio-vaccination, thus, the stimulation of the immune system through several pathways, including the type-I interferon response and HLA-class-I molecules up-regulation [6-8]. However, numerous studies reveal an additional ominous effect of RT, as irradiated patients experience profound lymphopenia depending upon the extent of field, fractionation, and dose levels [9-11]. Several clinical studies have documented the prognostic relevance of pre-treatment and treatment-related lymphopenia in solid tumors [12]. Regarding rectal cancer, Liu et al. reported that lower lymphocyte counts post-CRT define poor disease-free survival (DFS) [13]. Although a study by Lutsyk et al. [14] found no effect of lymphopenia on DFS, a significant association with poorer response to therapy was noted.
This retrospective study aimed to assess the effect of preoperative CRT on the peripheral blood lymphocytes of rectal adenocarcinoma patients and evaluate the impact of lymphopenia on treatment efficacy and patient prognosis.

Materials and Methods

Thirty-three rectal adenocarcinoma patients who were treated with preoperative CRT between 2014 and 2022 at the Radiation Oncology Unit of the Aretaieion University Hospital were included in this retrospective study. Table 1 summarizes patient demographics and imaging and histopathological disease characteristics. Patients who did not complete the treatment schedule or interrupted therapy for any reason for more than 1 week were excluded from the analysis.

1. Radiotherapy

RT was delivered via a 6-MV linear accelerator (Siemens, Erlangen, Germany). An intensity-modulated radiation therapy (IMRT) technique was applied using the Oncentra RT planning system (Elekta, Stockholm, Sweden). A two-phase irradiation schedule was followed. During the first phase, clinical target volume (CTV) included the rectum and perirectal area, the regional lymphatics (obturator, internal and common iliac nodes), and the presacral space. One patient with T4 stage also received RT to the external iliac nodes. A margin of 0.5–0.7cm beyond CTV was applied to define planning target volume (PTV). The total administered dose of radiation was 45 Gy using 25 fractions of 1.8 Gy, 5 fractions per week. During the second RT phase, a booster dose was given to the radiologically detectable tumor (gross tumor volume [GTV]). CTV was defined as GTV plus a margin of 2 cm, while a margin of 0.5 cm beyond CTV was considered for PTV with manual adjustment. The total RT dose was 5.4 Gy in 3 daily fractions of 1.8 Gy. Overall, the treatment planning was based on the relative guidelines of the Radiation Therapy Oncology Group [15].

2. Concurrent chemotherapy

Patients also received oral capecitabine at a dose of 825 mg/m2 twice daily, 5 days per week, throughout the RT course (approximately 5 weeks). Blood and biochemical tests were performed weekly in order to assess chemotherapy-related toxicity.

3. Lymphocyte assessment

Pre- and post-treatment lymphocyte counts (LC; in number of lymphocytes/μL) of patients were recorded one day before CRT initiation and on the day of the last fraction of CRT, respectively. Lymphopenia was graded using the Common Terminology Criteria for Adverse Events (CTCAE) version 5.0 [16]. LC are generally considered normal in the range of 1,500/μL and 4,000/μL. Lymphopenia was graded as 1 for values 800–1.500/μL, 2 for values 500–800/μL, 3 for values 200–500/μL, and 4 for values <200/μL. We further assessed the neutrophil to lymphocyte ratio (NLR; assessed by dividing the neutrophil counts with the lymphocyte counts obtained in same blood sample) after CRT.

4. Assessment of response

The pre-CRT radiological (c) and post-CRT pathologic (yp) T (tumor) and N (lymph nodal) stage were based on the 8th edition of the American Joint Committee on Cancer (AJCC) staging manual [17]. Tumor regression grade (TRG) was assessed based on the AJCC-College of American Pathologists (AJCC-CAP) system [18,19]. Archival histopathological material of post-CRT rectal resection specimens was centrally reviewed according to the World Health Organization 2019 Classification of Digestive System Tumours [20] by an expert digestive tract pathologist to define T & N stage, histological grade, blood and lymph vessel invasion (VI), and TRG.

5. Statistical analysis

Statistical analysis and graph presentation were performed using the GraphPad Prism 8.0.2 statistical package (GraphPad Software, Boston, MA, USA). Graphs presented in box and whisker plots display the range, median values, and 25th and 75th percentiles. The chi-square test and Fisher exact test were applied to investigate statistically significant differences between groups of categorical variables. The Wilcoxon matched-pairs signed rank test was used to test significant differences between paired groups of continuous variables. The Mann-Whitney test was utilized to compare unpaired groups of continuous variables. The Kruskal-Wallis test was used to compare multiple groups of continuous variables. Linear regression was applied to assess associations between groups of continuous variables. Kaplan-Meier curves were plotted to show locoregional relapse-free survival (LRFS), distant metastasis-free survival (DMFS) and disease-specific overall survival (OS). The log-rank (Mantel-Cox) test was used to assess differences in survival rates and hazard ratios. Cox-regression multivariate analysis and receiver operating characteristic (ROC) curve analysis were performed using the IBM SPSS 26.0 statistical package (version 26; IBM, Armonk, NY, USA). A p-value <0.05 was considered significant.

Results

1. Effects of CRT on lymphocyte counts

The LC before CRT ranged from 1,345/μL to 6,239/μL (median, 2,184/μL). All but two patients had normal LC; one patient had grade 1 lymphopenia, and one patient had abnormally high LC. The LC after CRT ranged from 256/μL to 2,500/μL (median, 517/μL). Grade 0, 1, 2 and 3 lymphopenia were observed in 1, 6, 12, and 14 patients, respectively. LC drop was noted in all 33 rectal adenocarcinoma patients after CRT (Fig. 1A). The difference between LC counts before and after treatment was strongly significant (p < 0.001) (Fig. 1B). We performed a linear regression analysis to assess whether the reduction in LC was affected by the initial LC levels. Excluding one patient with very high LC before CRT (outlier), there was no statistically significant association (p = 0.254, r = 0.20) (Fig. 1C).

2. Analysis of response to CRT

Out of four patients and 28 patients with cT2 and cT3-stage, respectively, 3 (75%) and 10 (35.7%) patients displayed disease downstaging (Fig. 2A). Out of 26 patients with radiologically involved lymph nodes (cN+), 16 patients (61.5%) showed a lack of nodal involvement after CRT (ypN0) (Fig. 2B).
Histopathology examination showed that out of 33 cases, three specimens (9.1%) demonstrated no viable cancer cells (TRG0), while 5 (15.2%) exhibited isolated small cancer cell groups (TRG1). Fifteen specimens (45.4%) had features of tumor regression, with groups of viable cancer cells outgrown by fibrosis/necrosis (TRG2). Extensive residual tumor was noted in 10/33 (30.3%) cases (TRG3) (Fig. 2C).

3. Pre-treatment LC and response to CRT

Although almost all patients had normal LC before CRT, we performed an analysis to test whether the levels of LC had an impact on response to CRT. Analysis of the pre-CRT LC demonstrated no difference between the ypT-stage groups of patients (p = 0.219). Regarding ypN status of cN+ cases, we found no statistically significant differences between the ypN+ or ypN– subgroups of patients (p = 0.310). As far as TRG is concerned, analysis of LC before CRT displayed no statistically significant differences between the TRG subgroups of patients (p = 0.298).

4. Post-treatment LC and response to CRT

Analysis of "LC" and "lymphopenia grade" after CRT according to the ypT stage did not demonstrate any statistically significant differences (p = 0.985 and p = 0.900, respectively). Similar analysis regarding ypN status of cN+ patients showed no significant association (p = 0.508 and p = 0.285 for LC and lymphopenia grade, respectively). Regarding the histopathological grade after CRT, only four out of 30 patients with viable tumor in the surgical specimens were of high grade. This hinders any subsequent analysis. VI, noted in 6/30 patients, was not related with post-CRT LC, or lymphopenia grade (p = 0.187 and p = 0.402, respectively).
Analysis of post-CRT LC according to TRG showed a progressive drop of the median values from the TRG0 to the TRG3 subgroup of patients (median values of 645, 676, 504, and 445 for TRG0, TRG1, TRG2, and TRG3, respectively; p = 0.135). However, grouping patients into two TRG categories (TRG0,1 vs. TRG2,3) displayed a statistically significant association of intense lymphopenia with poorer histopathological response (median values of 660 and 494, respectively; p = 0.036). No significant correlation between lymphopenia grade and TRG was noted (p = 0.113).
After CRT, the NLR ratio ranged from 2.23 to 15.02, with a median value of 6.00. Analysis of NLR according to the ypT stage, ypN status of cN+ patients and TRG did not reveal any significant correlation (p = 0.481, p = 0.124, and p = 0.236, respectively), although a significant association of higher NLR with VI was shown (p = 0.018).

5. LPFS analysis

Within a median follow-up of 32 months (range, 9 to 102 months), none of the patients relapsed locally or within the irradiated pelvic lymph node area. Therefore, any analysis regarding the role of ypT stage, ypN status, VI, TRG, and LC after CRT is not feasible.

6. DMFS analysis

Within a median follow-up of 32 months (range, 9 to 102 months), eight out 33 patients (24.2%) developed distant metastasis. Liver metastases were recorded in six out of 8 patients, while three patients developed lung metastases. Kaplan-Meier survival analysis showed that the median DMFS was not reached. The projected 5-year DMFS was 68%. Analysis according to the initial cT (T2 vs. T3,4) and cN stage (negative vs. positive) showed no significant impact on DMFS (p = 0.838 and p = 0.372, respectively).
Kaplan-Meier analysis of DMFS according to ypT stage, ypN status, VI after CRT, TRG and distance from anorectal ring is shown in Table 2. The first three parameters were significantly related to prognosis (p = 0.043, p = 0.033, and p = 0.002, respectively), while TRG displayed marginal significance (p = 0.050). As mentioned earlier, only four patients had residual tumors of high histopathological grade, rendering any survival analysis unreliable.
We further examined the impact of post-CRT lymphopenia on DMFS. By grouping our cases into two categories according to the CTCAE criteria (lymphopenia grade 0,1 vs. 2,3), it was shown that patients with grade 2,3 lymphopenia had worse, although not statistically significant, DMFS (p = 0.125). When categorizing LC into three groups based on percentiles: 0%–33%, 33%–67%, and 67%–100%, it was observed that patients with the highest LC (624–2,500/μL) after CRT exhibited significantly better DMFS compared to other groups (p = 0.034) (Fig. 3A). As the number of LC after CRT appears of prognostic relevance, we performed a ROC curve to identify the best LC value that predicts DMFS. Analysis showed an area under the ROC curve value of 0.71 and suggested a LC cutoff point of 538/μL. Therefore, we used the existing 518 value to split cases into two groups (≤518 vs. >518). Kaplan-Meier DMFS curves were plotted according to this value and showed a strong significant association (p = 0.003) (Fig. 3B). None of the patients (0%) with post-CRT LC above 518/μL presented with metastases versus 8/18 patients (44.5%) with LC ≤518/μL. The median DMFS was 48 months for patients with LC ≤518/μL and was not reached (more than half of the patients had no metastasis at the time of analysis) for patients with LC >518/μL. NLR was not related to DMFS (p = 0.304).
Multivariate analysis of parameters with prognostic significance in univariate analysis (log-rank test) did not identify any independent prognostic variable. In specific, we examined in multivariate Cox-regression analysis a model based on the five parameters that were significantly related to DMFS (ypT stage, ypN status, VI, TRG, and LC after CRT considering the cutoff point as defined by ROC analysis). None of the parameters showed statistical significance (p = 0.966, p = 0.426, p = 0.222, p = 0.996, and p = 0.956 for ypT stage, ypN status, VI, TRG, and LC, respectively).

7. Disease-specific OS analysis

Out of 33 patients, three were deceased at the time of last follow-up. One more patient had died from other causes. The median OS was not reached. It is stressed that the number of death events is too low to extract conclusions regarding the impact of different parameters on OS. Nevertheless, we performed an analysis to present potential trends. None of the examined parameters showed a significant association with OS.

Discussion and Conclusion

Preoperative CRT is the standard treatment approach for locoregionally advanced rectal adenocarcinoma, while immunotherapy with the anti-PD1 monoclonal antibody dostarlimab has recently emerged as a potential active agent to further enhance prognosis for a subgroup of patients with mismatch repair deficiency [21]. RT has also been shown to activate in situ immune responses that may stimulate abscopal and in-field anti-tumor immunity which can eventually contribute to tumor eradication [22]. We recently summarized the effects of CRT on immune-checkpoint molecules up- or down-regulation on the cancer cell surface of rectal carcinomas, as well as its impact on the quantity and quality of tumor-infiltrating lymphocytes [23]. However, it is essential to underline that an intact systemic immune response is required in order to facilitate any kind of intratumoral immune response [24].
In this context, RT or CRT-related lymphopenia rises as a significant barrier to a potentially effective immune-mediated local or even distant—through the abscopal effects—disease eradication [25]. Multiple studies have reported on the severe lymphopenia documented post-treatment with RT or CRT for various tumor sites [26,27]. In fact, this adverse event becomes more prominent shortly after treatment completion [27], with a gradual restoration of LC being noted in the following months. Grossman et al. [26] further highlighted that the median LC count of patients 1 year after CRT was below 1,000/μL. As far as rectal adenocarcinoma is concerned, Campian et al. [28] demonstrated that severe lymphopenia (grade 3 or 4) was evident in 35% of 57 rectal adenocarcinoma patients 2 months after treatment with RT of 45 Gy and concurrent capecitabine. A more recent study by Lutsyk et al. [14] also confirmed significant CRT-induced lymphopenia in 198 patients with locally advanced rectal adenocarcinoma. In addition, a profound decrease in LC has been displayed in patients with rectal adenocarcinoma post-adjuvant CRT [29].
In the current study, we observed a statistically significant drop in the LC of 33 rectal adenocarcinoma patients who received 50.4 Gy of IMRT in combination with capecitabine. The median LC before CRT and at treatment completion were 2,184/μL and 517/μL, respectively, suggesting an incidence of grade 3 lymphopenia in 38% of patients, similar to the study by Campian et al. [28]. In the study by Li et al. [29], grade 3 lymphopenia was noted in 58% of patients, while Lutsyk et al. [14] reported lymphopenia—defined as LC less than 1,000/μL—in 75% of patients. The high intrinsic radiosensitivity of lymphocytic populations, the large radiation portals applied to cover pelvic lymph nodes, and the relatively high dose delivered to the bone marrow of the pelvic bones are the main contributing factors to RT-induced lymphopenia [25]. Fractionation of RT also appears to impact the severity of lymphopenia. Hypofractionated RT has been associated with less significant lymphopenia in two pancreatic and lung cancer studies, potentially due to the lower normalized total dose delivered to the lymphocytes and the shorter overall treatment time [30,31]. To our knowledge, there are no relevant data in rectal cancer patients comparing the long courses of CRT with short-course hypofractionated regimens. In the era of immune-RT, the potential incorporation of monoclonal antibodies in the preoperative setting brings forward the eventual value of reduced-length RT-fields to treat involved-only lymph nodes, while hypofractionation would further contribute to the protection of LCs and enhancement of immunotherapy’s efficacy [32].
In the next step, we assessed whether the graveness of treatment-related lymphopenia could affect response to CRT. Several studies have showcased a significant correlation between high LC after preoperative CRT for rectal adenocarcinoma patients and achieving a pathological CR [13,14,33,34]. Kitayama et al. [33] examined the lymphocyte ratio in the total white cell counts and found that this was increased in complete responders. In addition, Heo et al. [34] reported that patients who sustained LC above 35% of the pre-RT values experienced higher pathological CR rates, while Liu et al. [13] observed better tumor responses in patients with higher LC during CRT. On the other hand, Wu et al. [35] suggested that lymphopenia after neoadjuvant treatment could be linked with tumor regression, citing deprivation of regulatory T-cells as a potential explanation.
Although our study found no association between post-CRT LC and ypT stage or ypN status, a significant correlation was revealed between higher blood LC and better TRG. A larger cohort of patients could potentially allow the extraction of safer conclusions regarding the effect of CRT-related lymphopenia on treatment response. Moreover, the complexity of T-downstaging further hinders drawing out clear correlations; for example, downstaging of a cT3 stage to ypT2 and ypT1 stages are two different scenarios, possibly concealing a different effect of patients’ systemic immunity status and microenvironmental conditions that define the access of tumor-infiltrating lymphocytes. Hypoxia and low pH may suppress cytotoxic T-cell survival and anti-tumor activity in the tumor stroma, and this can counteract even strong systemic anti-tumor responses [36,37]. Poor lymphocytic presence within rectal carcinomas strongly affects the response to CRT and the outcome of therapy [23].
Complete response to CRT is associated with better prognosis for patients with rectal adenocarcinoma, as suggested by the majority of published data. While older data of a randomized phase III trial comparing preoperative CRT with adjuvant CRT for rectal adenocarcinoma support no correlation between response and survival [38], more recent retrospective studies and a meta-analysis suggest improved OS and DFS of patients with complete tumor regression [39-41]. Specifically, Ulusoy et al. reported that patients with better TRG(0,1) had significantly better OS [40]. In addition, a National Cancer Database analysis of 56,812 rectal adenocarcinoma patients who received neoadjuvant CRT demonstrated that patients with pathologic complete responses survived longer when compared to non-complete responders [42].
In our study, a higher ypT stage was significantly linked with shorter DMFS. This was also the case for patients with TRG2,3, although the statistical association was of marginal significance. VI and ypN+ status were also shown to define poor DMFS. Due to the relatively low number of patients and short-follow up in the current study, no significant correlations were found between some prognostic factors such as ypT stage, ypN status, VI, and TRG and survival outcomes including LPFS/OS. These findings are in accordance with previously published studies. Tang et al. [43] recently reported a significant association of ypN stage with distant metastasis in a series of 146 patients with rectal cancer. Similarly, VI has been suggested to adversely affect progression-free and overall survival [44,45].
Aside from tumor regression-related features, multiple studies have showcased the negative effect of RT- or CRT-induced lymphopenia on the prognosis of patients with carcinomas of various origins [11,46-48]. In a report by Liu et al. [13], it was demonstrated that lower LC post-CRT were associated with worse OS and DFS of rectal adenocarcinoma patients. A more recent study, however, found no effect of lymphopenia on DFS [14]. In the current retrospective analysis, a statistically significant impact of LC on DMFS was evident, since patients with more pronounced lymphopenia (LC ≤518/μL) developed distant metastases more frequently (44.5% vs. 0% of patients with LC >518/μL). Of note, Liu et al. [13] used a cutoff point of 440/μL for LC, as defined by ROC curve analysis, similar to the one proposed in our study. In contrast, a cutoff point of 1,000/μL was applied in the analysis by Lutsyk et al. [14].
Furthermore, Sung et al. [49] showed a significant association between an elevated NLR after preoperative CRT and worse prognosis of patients with locally advanced rectal adenocarcinoma. Ιn our study, this was not confirmed. It should be taken into account that the NLR incorporates neutrophil counts that are primarily affected by chemotherapy. Moreover, many neutropenic patients during CRT also receive granulocyte-colony stimulating factors; thus, the full blood count at treatment completion does not necessarily represent the actual effect of CRT. Thus, post-treatment NLR is suggested to harbor substantial bias related to chemotherapy.
Certain limitations of the study have already been addressed. The retrospective nature of our investigation, combined with the limited number of cases, does not allow the extraction of safe conclusions. Longer follow-up could also reveal associations of the examined parameters with OS. In addition, a comparative analysis of the current data with patients treated with hypofractionated short-course RT could further shed light on the impact of different fractionation on lymphopenia. However, we believe that our patient group represents a unique set with detailed pre- and post-CRT data. Moreover, central review of histopathology significantly contributes to the quality of the study.
Overall, lymphopenia has emerged as a significant adverse event that rectal adenocarcinoma patients face during treatment with neoadjuvant CRT, with subsequent implications on tumor response and prognosis. As suggested in the current study, LC reduction to levels in the range of profound grade 2 and grade 3 lymphopenia defines poorer pathological tumor responses and higher incidence of distant metastases. Further investigation on the effects of different RT schedules on lymphopenia (normal fractionation vs. hypofractionation or even ultra-hypofractionation), as well as the reduction of the RT fields that would spare functional lymph nodes in combination with targeted immunotherapy is required to understand better and overcome this prognosis-affecting toxicity.

Statement of Ethics

This study was performed in line with the principles of the Declaration of Helsinki. Approval was granted by the local Ethics and Research Committee (No. 316/26-03-2021). Written informed consent was obtained from all individual participants included in the study. The authors affirm that human research participants provided informed consent for anonymous publication of their clinical and laboratory data.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Acknowledgments

The study has been conducted in the context of the ongoing PhD thesis by IMK. AZ, DT and VK are members of the PhD supervising committee.

Funding

None.

Author Contributions

Conceptualization, IMK, IG, DD, DT, VK, AZ; Investigation and methodology, IMK, IG, DD; Writing of the original draft, IMK; Writing of the review and editing, IG, DD, DT, VK, AZ; Formal analysis, AZ, DT, VK, IMK.

Data Availability Statement

All data are kept in the archives of our department and can be made available upon reasonable request.

Fig. 1.
Lymphocyte count (LC) changes after chemoradiotherapy (CRT): (a) individual changes of LC after CRT; (b) box and whisker plot of LC before and after CRT showing the range, median value and 25th and 75th percentiles; (c) linear regression analysis of LC before vs. after CRT.
roj-2024-00052f1.jpg
Fig. 2.
Response after chemoradiotherapy: (a) T-downstaging of cT2 and cT3 cases, (b) N-downstaging of cN+ cases and (c) tumor regression grade (TRG). pts, patients; pos, positive; neg, negative.
roj-2024-00052f2.jpg
Fig. 3.
Distant metastasis-free survival (DMFS) analysis according to post chemoradiotherapy lymphopenia: (a) Kaplan-Meier survival curves according to 33th and 67th LC percentiles and (b) Kaplan-Meier survival curves according to the LC cutoff point defined by ROC analysis (≤518/μL vs. >518/μL) LC, lymphocyte count; ROC, receiver operating characteristic.
roj-2024-00052f3.jpg
Table 1.
Patient and disease characteristics (n=33)
Characteristic Value
Sex
 Male 23
 Female 10
Age (year) 72 (46–87)
PS
 0 33
Histological type
 Adenocarcinoma NOS 33
Histological grade (post-CRT)
 Low 26
 High 4
 Not assessable/complete response 3
Vascular invasion (post-CRT)
 No 24
 Yes 6
 Not assessable/complete response 3
Distance from anorectal ring (cm) 5 (0–10)
 >5 15
 ≤5 18
cT stage (MRI)
 T2a) 4
 T3 28
 T4 1
ypT stage
 T0 3
 T1 5
 T2 6
 T3 19
cN status (MRI)
 Positive 26
 Negative 7
ypN statusb)
 Positive 10
 Negative 16
Tumor regression grade AJCC/CAP
 0 3
 1 5
 2 15
 3 10

Values are presented as number or median (range).

PS, performance status; NOS, not otherwise specified; CRT, chemoradiotherapy; MRI, magnetic resonance imaging; AJCC/CAP, American Joint Committee on Cancer-College of American Pathologists.

a)Node positive, b)cN positive status patients.

Table 2.
Histopathological and lymphocyte-related prognostic factors for DMFS by univariate analysis using the log-rank test
Parameter 2-yr DMFS (%) p-value HR (95% CI)
Distance from anorectal ring (≤5 vs. >5 cm) 70 vs. 93 0.134 2.95 (0.71–12.16)
ypT-stage (ypT2,3 vs. ypT0,1) 74 vs. 100 0.043 4.85 (1.04–22.57)
ypN status (ypN+ vs. ypN0) 60 vs. 84 0.033 5.65 (1.14–27.94)
VI (Yes vs. No) 33 vs. 91 0.002 25.19 (3.17–200.20)
TRG (TRG 2,3 vs. TRG 0,1) 74 vs. 100 0.050 4.70 (0.99–22.18)
Lymphopenia grade (2,3 vs. 0,1) 76 vs. 100 0.125 3.73 (0.69–20.15)
Lymphopenia grade (3 vs. 0–2) 78 vs. 83 0.481 1.68 (0.39–7.25)
LC 33rd and 67th percentile (A <450 vs. B 450–623 vs. C ≥624/μL) 81 vs. 59 vs. 100 0.067 A vs. B: 0.56 (0.13–2.36)
A vs. C: 7.25 (0.72–72.91)
B vs. C: 8.52 (1.45–49.96)
LC 67th percentile (<624 vs. ≥624/μL) 71 vs. 100 0.032 4.87 (1.12–21.20)
LC ROC-defined (≤518 vs. >518/μL) 65 vs. 100 0.003 8.42 (2.04–34.70)
NLR ROC-defined (>6.82 vs. ≤6.82) 77 vs. 83 0.304 2.09 (0.50–8.65)

DMFS, distance metastasis-free survival; VI, vascular/lymphatic invasion; TRG, tumor regression grade; LC, lymphocyte counts; NLR, neutrophil to lymphocyte ratio; ROC, receiver operating characteristic; HR, hazard ratio; CI, confidence interval.

References

1. Swedish Rectal Cancer Trial, Cedermark B, Dahlberg M, et al. Improved survival with preoperative radiotherapy in resectable rectal cancer. N Engl J Med 1997;336:980–7.
crossref pmid
2. Bahadoer RR, Dijkstra EA, van Etten B, et al. Short-course radiotherapy followed by chemotherapy before total mesorectal excision (TME) versus preoperative chemoradiotherapy, TME, and optional adjuvant chemotherapy in locally advanced rectal cancer (RAPIDO): a randomised, open-label, phase 3 trial. Lancet Oncol 2021;22:29–42.
crossref pmid
3. National Comprehensive Cancer Network. NCCN Guidelines: rectal cancer [Internet]. Plymouth Meeting, PA: National Comprehensive Cancer Network; 2024 [cited 2024 Oct 1]. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=146.

4. Wang Y, Shen L, Wan J, et al. Short-course radiotherapy combined with CAPOX and Toripalimab for the total neoadjuvant therapy of locally advanced rectal cancer: a randomized, prospective, multicentre, double-arm, phase II trial (TORCH). BMC Cancer 2022;22:274.
crossref pmid pmc pdf
5. Menetrier-Caux C, Ray-Coquard I, Blay JY, Caux C. Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer 2019;7:85.
crossref pmid pmc
6. Vanpouille-Box C, Alard A, Aryankalayil MJ, et al. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat Commun 2017;8:15618.
crossref pmid pmc pdf
7. Koukourakis IM, Xanthopoulou E, Sgouras TI, et al. Preoperative chemoradiotherapy induces multiple pathways related to anti-tumour immunity in rectal cancer. Br J Cancer 2023;129:1852–62.
crossref pmid pmc pdf
8. Theelen WS, Chen D, Verma V, et al. Pembrolizumab with or without radiotherapy for metastatic non-small-cell lung cancer: a pooled analysis of two randomised trials. Lancet Respir Med 2021;9:467–75.
crossref pmid
9. Ellsworth SG. Field size effects on the risk and severity of treatment-induced lymphopenia in patients undergoing radiation therapy for solid tumors. Adv Radiat Oncol 2018;3:512–9.
crossref pmid pmc
10. Tang C, Liao Z, Gomez D, 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.
crossref pmid
11. Sun GY, Wang SL, Song YW, et al. Radiation-induced lymphopenia predicts poorer prognosis in patients with breast cancer: a post hoc analysis of a randomized controlled trial of postmastectomy hypofractionated radiation therapy. Int J Radiat Oncol Biol Phys 2020;108:277–85.
crossref pmid
12. Venkatesulu BP, Mallick S, Lin SH, Krishnan S. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit Rev Oncol Hematol 2018;123:42–51.
crossref pmid
13. Liu H, Wang H, Wu J, et al. Lymphocyte nadir predicts tumor response and survival in locally advanced rectal cancer after neoadjuvant chemoradiotherapy: Immunologic relevance. Radiother Oncol 2019;131:52–9.
crossref pmid
14. Lutsyk M, Taha T, Billan S. Can lymphocytes serve as a predictor of response to preoperative chemoradiation therapy for locally advanced rectal cancer? Front Oncol 2023;13:1138299.
crossref pmid pmc
15. Myerson RJ, Garofalo MC, El Naqa I, et al. Elective clinical target volumes for conformal therapy in anorectal cancer: a radiation therapy oncology group consensus panel contouring atlas. Int J Radiat Oncol Biol Phys 2009;74:824–30.
crossref pmid pmc
16. US Department of Health and Human Services. Common Terminology Criteria for Adverse Events (CTCAE), Version 5.0 [Internet]. Washington, DC: US Department of Health and Human Services; 2017 [cited 2024 Oct 1]. Available online: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/CTCAE_v5_Quick_Reference_5x7.pdf.

17. Amin MB, Greene FL, Edge SB, et al. The Eighth Edition AJCC Cancer Staging Manual: continuing to build a bridge from a population-based to a more “personalized” approach to cancer staging. CA Cancer J Clin 2017;67:93–9.
crossref pmid
18. Mace AG, Pai RK, Stocchi L, Kalady MF. American Joint Committee on Cancer and College of American Pathologists regression grade: a new prognostic factor in rectal cancer. Dis Colon Rectum 2015;58:32–44.
crossref pmid
19. Pai RK, Pai RK. Pathologic assessment of gastrointestinal tract and pancreatic carcinoma after neoadjuvant therapy. Mod Pathol 2018;31:4–23.
crossref pmid pdf
20. World Health Organization. Digestive system tumours (WHO Classification of Tumours, 5th edition). Geneva, Switzerland: World Health Organization; 2019.

21. Cercek A, Lumish M, Sinopoli J, et al. PD-1 blockade in mismatch repair-deficient, locally advanced rectal cancer. N Engl J Med 2022;386:2363–76.
crossref pmid pmc
22. Koukourakis IM, Tiniakos D, Kouloulias V, Zygogianni A. The molecular basis of immuno-radiotherapy. Int J Radiat Biol 2023;99:715–36.
crossref pmid
23. Koukourakis IM, Platoni K, Tiniakos D, Kouloulias V, Zygogianni A. Immune response and immune checkpoint molecules in patients with rectal cancer undergoing neoadjuvant chemoradiotherapy: a review. Curr Issues Mol Biol 2023;45:4495–517.
crossref pmid pmc
24. Spitzer MH, Carmi Y, Reticker-Flynn NE, et al. Systemic immunity is required for effective cancer immunotherapy. Cell 2017;168:487–502.
crossref pmid pmc
25. Koukourakis MI, Giatromanolaki A. Lymphopenia and intratumoral lymphocytic balance in the era of cancer immuno-radiotherapy. Crit Rev Oncol Hematol 2021;159:103226.
crossref pmid
26. Grossman SA, Ellsworth S, Campian J, 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.
crossref pmid pmc
27. Takeda K, Umezawa R, Yamamoto T, et al. Acute hematologic toxicity of radiation therapy: a comprehensive analysis and predictive nomogram. J Radiat Res 2023;64:954–61.
crossref pmid pmc pdf
28. Campian JL, Ye X, Sarai G, Herman J, Grossman SA. Severe treatment-related lymphopenia in patients with newly diagnosed rectal cancer. Cancer Invest 2018;36:356–61.
crossref pmid
29. Li S, Yao W, Liu R, Lu Y, Zhang H, Liang X. Severe lymphopenia as a prognostic factor in rectal cancer patients receiving adjuvant chemoradiotherapy: a retrospective study. Sci Rep 2023;13:7566.
crossref pmid pmc pdf
30. Crocenzi T, Cottam B, Newell P, 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.
crossref pmid pmc
31. Zhao Q, Chen G, Ye L, et al. Treatment-duration is related to changes in peripheral lymphocyte counts during definitive radiotherapy for unresectable stage III NSCLC. Radiat Oncol 2019;14:86.
crossref pmid pmc pdf
32. Demaria S, Guha C, Schoenfeld J, et al. Radiation dose and fraction in immunotherapy: one-size regimen does not fit all settings, so how does one choose? J Immunother Cancer 2021;9:e002038.
crossref pmid pmc
33. Kitayama J, Yasuda K, Kawai K, Sunami E, Nagawa H. Circulating lymphocyte is an important determinant of the effectiveness of preoperative radiotherapy in advanced rectal cancer. BMC Cancer 2011;11:64.
crossref pmid pmc pdf
34. Heo J, Chun M, Noh OK, et al. Sustaining blood lymphocyte count during preoperative chemoradiotherapy as a predictive marker for pathologic complete response in locally advanced rectal cancer. Cancer Res Treat 2016;48:232–9.
crossref pmid pmc pdf
35. Wu Z, Zhang J, Cai Y, et al. Reduction of circulating lymphocyte count is a predictor of good tumor response after neoadjuvant treatment for rectal cancer. Medicine (Baltimore) 2018;97:e11435.
crossref pmid pmc
36. Byrnes JR, Weeks AM, Shifrut E, et al. Hypoxia is a dominant remodeler of the effector T cell surface proteome relative to activation and regulatory T cell suppression. Mol Cell Proteomics 2022;21:100217.
crossref pmid pmc
37. Verma NK, Wong BH, Poh ZS, et al. Obstacles for T-lymphocytes in the tumour microenvironment: therapeutic challenges, advances and opportunities beyond immune checkpoint. EBioMedicine 2022;83:104216.
crossref pmid pmc
38. Roh MS, Colangelo LH, O’Connell MJ, et al. Preoperative multimodality therapy improves disease-free survival in patients with carcinoma of the rectum: NSABP R-03. J Clin Oncol 2009;27:5124–30.
crossref pmid pmc
39. Sell NM, Qwaider YZ, Goldstone RN, et al. Ten-year survival after pathologic complete response in rectal adenocarcinoma. J Surg Oncol 2021;123:293–8.
crossref pmid pdf
40. Ulusoy C, Kamali GH, Nikolovski A. Analysis of survival in complete pathological response after long-course chemoradiotherapy in patients with advanced rectal cancer. Curr Oncol 2023;30:1054–64.
crossref pmid pmc
41. Li JY, Huang XZ, Gao P, et al. Survival landscape of different tumor regression grades and pathologic complete response in rectal cancer after neoadjuvant therapy based on reconstructed individual patient data. BMC Cancer 2021;21:1214.
crossref pmid pmc pdf
42. Narayanan S, Attwood K, Gabriel E, Nurkin S. Pathologic complete response despite nodal yield has best survival in locally advanced rectal cancer. J Surg Res 2020;251:220–7.
crossref pmid pmc
43. Tang C, Xu J, Lin M, et al. Risk factors for distant metastasis in T3 T4 rectal cancer. Clin Med Insights Oncol 2024;18:11795549241227423.
crossref pmid pmc pdf
44. Betge J, Pollheimer MJ, Lindtner RA, et al. Intramural and extramural vascular invasion in colorectal cancer: prognostic significance and quality of pathology reporting. Cancer 2012;118:628–38.
crossref pmid
45. Nikberg M, Chabok A, Letocha H, Kindler C, Glimelius B, Smedh K. Lymphovascular and perineural invasion in stage II rectal cancer: a report from the Swedish colorectal cancer registry. Acta Oncol 2016;55:1418–24.
crossref pmid
46. Cheng X, Chen B, Wang S, et al. Association between lymphopenia and survival outcomes in esophageal carcinoma patients receiving combined immunotherapy and chemoradiotherapy. Oncologist 2023;28:e606–16.
crossref pmid pmc pdf
47. Abravan A, Faivre-Finn C, Kennedy J, McWilliam A, van Herk M. Radiotherapy-related lymphopenia affects overall survival in patients with lung cancer. J Thorac Oncol 2020;15:1624–35.
crossref pmid
48. Wild AT, Ye X, Ellsworth SG, et al. The association between chemoradiation-related lymphopenia and clinical outcomes in patients with locally advanced pancreatic adenocarcinoma. Am J Clin Oncol 2015;38:259–65.
crossref pmid pmc
49. Sung S, Son SH, Park EY, Kay CS. Prognosis of locally advanced rectal cancer can be predicted more accurately using pre- and post-chemoradiotherapy neutrophil-lymphocyte ratios in patients who received preoperative chemoradiotherapy. PLoS One 2017;12:e0173955.
crossref pmid pmc
Editorial Office
Department of Radiation Oncology, Samsung Medical Center,
Proton Therapy Center, B2, 81, Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
Tel : +82-2-3410-3617
E-mail: rojeditor@gmail.com, roj@kosro.or.kr
Copyright © The Korean Society for Radiation Oncology.                      Developed in M2PI
Close layer
prev next