Abstract
Radiotherapy is a frequently utilized therapeutic modality in the treatment of esophageal cancer (EC). Even though extensive studies are carried out in radiotherapy for EC, the design of the clinical target volume and the radiation dose is not satisfactorily uniform. Radiotherapy acts as a double-edged sword on the immune system; it has both an immunostimulatory effect and an immunosuppressive effect. Radiation-induced lymphopenia and its potential association with tumor control and survival outcomes remain to be understood. The advent of immunotherapy has renewed the focus on preserving a pool of functioning lymphocytes in the circulation. In this review, we summarize the potential impact mechanisms of radiotherapy on peripheral blood lymphocytes and the prognostic role of radiation-induced lymphopenia in patients with EC. We also propose the concept of organs-at-risk of lymphopenia and discuss potential strategies to mitigate its effects on patients with EC. From an immunological perspective, we put forward the hypothesis that optimizing radiation modalities, radiation target volume schemes, and radiation doses could help to reduce radiation-induced lymphopenia risks and maximize the immunomodulatory role of radiotherapy. An optimized radiotherapy plan may further enhance the feasibility and effectiveness of combining immunotherapy with radiotherapy for EC.
Keywords: anti-tumor immunity, esophageal cancer, lymphopenia, organs-at-risk of lymphopenia, radiotherapy
Background
Esophageal cancer (EC) is one of the most aggressive tumors and is the sixth leading cause of cancer-related mortality worldwide.1 EC includes two predominant histological types: esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC).
At present, neoadjuvant chemoradiotherapy (CRT) followed by surgery is the preferred treatment for locally advanced EC patients, but definitive CRT is an alternative treatment for non-operative candidates.2,3 According to the landmark RTOG 94-05 trial, 50.4 Gy is the standard dose of radiation in Europe and North America for patients undergoing definitive CRT.4 Although radiation dose escalations have failed to improve local control or survival, a dose of 60.0 Gy or more is more prevalent in many Asian countries, including China and Japan.5,6
The nodal clinical target volume (CTV) of external beam radiation therapy (RT) has remained a topic of persistent controversy among radiation oncologists. EC commonly shows lymph node metastases, particularly regional lymph node involvement, even in the early phases of the disease.7 The conservative school of thought on this matter promotes the involved-field irradiation (IFI) – that is, irradiation of positive lymph nodes only – because the rationale to include elective nodal irradiation (ENI) is to prevent regional nodal relapse rather than improve survival. The other school of thought favors prophylactically irradiating the regional lymph node area according to different tumor sites because ENI could be of benefit in eliminating potential subclinical nodal disease.8 The design of different radiation fields is shown in Figure 1.
Figure 1.

The design of IFI and ENI for EC. Red: GTV; Blue: IFI field in the coronal direction; Green: ENI field in the coronal direction.
EC, esophageal cancer; ENI, elective nodal irradiation; GTV, gross tumor volume; IFI, involved-field irradiation.
Treatment-related lymphopenia is a common side effect of RT, chemotherapy, and certain drugs, like steroids, that are used in cancer patients. RT may play a primary role in the etiology of treatment-related lymphopenia. Lymphocytes are highly radiosensitive, and exposure to low doses of radiation could lead to a decrease in the number of peripheral blood lymphocytes.9 In a study on non-small-cell lung cancer (NSCLC), Campian et al. revealed that total lymphocyte counts did not change following neoadjuvant chemotherapy, but steep declines were noted after the initiation of thoracic RT.10 In another study, researchers found that RT, with or without concomitant chemotherapy, induced substantial and long-lasting immune suppression in patients with cervical cancer.11 The immune system has multiple mechanisms for identifying tumor cells and removing them from the body, most of which involve lymphocytes.12 Treatment-related lymphopenia is associated with poor prognosis in many types of cancer,13–17 but the mechanics remain to be understood.
Lymphocytes play a vital role in antitumor immunity, but radiosensitivity makes them vulnerable targets during radiation therapy. It seems reasonable to assume that preserving a pool of functioning lymphocytes in circulation might contribute to treatment outcomes. Because there is controversy in the concept of radiation field size and dose, we need an immunological perspective to assist in improving individualized treatment for EC patients. This review, therefore, summarizes current knowledge on RT for EC and attempts to evaluate the potential effect of RT on the immune system, especially the count of peripheral circulating lymphocytes. Topics covered include effects of radiation-induced lymphopenia (RIL) in patients with EC, the organs-at-risk (OARs) of lymphopenia, and the possible risk avoidance of RIL.
Radiation-induced lymphopenia
In general, the immune system, especially cellular immunity, is thought to play a central role in cancer suppression.10 Lymphocytes are the primary carriers of cell-mediated immune mechanisms; they play a critical role in promoting a systemic immune response against tumors.18 T-cells have efforts of endogenous anti-tumor immunity.19 CD8+ and CD4+ T-cells can drastically improve the prognosis of patients with EC due to their ability to directly destroy tumor cells or secrete cytokines that activate effector cells.20,21 The use of immune checkpoint inhibitor (ICPi) blockers modulates the interaction between T-lymphocytes and tumor cells or macrophages, thereby favoring the re-induction of T cell populations in the tumor environment, which leads to a durable clinical response.22 Recently, with the recognition of ICPi as a potent therapeutic agent in the immunotherapy of cancer, the status of the immune system has been deemed an essential biomarker for responses to novel treatments.22 It is, hence, crucial to maintain an intact adaptive immune system during cancer therapy so as to improve cancer control and enhance the effectiveness of treatment.23
A lymphocyte is the most radiosensitive cell of the hematopoietic system and is frequently depleted by RT using a 50% lethal dose of 1–2 Gy.9,24 Currently, the mechanism of RIL is less clear. A systemic effect of localized postmastectomy radiation upon the long-term lymphocyte counts was described in 1970.25 The study showed that the total lymphocyte counts of irradiated patients decreased, whereas those of unirradiated patients remained unchanged. Lymphopenia was also demonstrated to exist persistently in irradiated patients for at least 4–8 years after treatment. The hypothesis at that time was that direct radiation damage to the thymus prevented the restoration of the usual level of lymphocytes because targeting the internal mammary lymph nodes resulted in the delivery of reasonably high doses to the anterior thymus and mediastinum. Stjernsward et al. also analyzed the change in lymphocyte subpopulations in patients receiving irradiation postmastectomy.26 As a comparison, the authors used healthy individuals in some cases or patients receiving irradiation that did not include treatment reaching the thymus gland. Per the results obtained, compared with both control groups, the thymus-derived lymphocyte subpopulation decreased significantly, and the bone marrow-derived lymphocyte subpopulation increased relatively in breast cancer patients receiving irradiation. Raben et al. later questioned the findings of Stjernsward et al.,27 intimating that the volume of irradiated blood concomitant with tumor irradiation should also have been taken into account. In the research conducted by Raben et al., the irradiation of breast cancer resulted not only in the irradiation of the thymus area but also in the irradiation of the large thoracic and cardiac blood vessels. Given the findings they obtained, the authors compared lymphocyte count in peripheral blood in patients receiving pelvic irradiation with patients receiving thoracic irradiation, and found that the irradiation of the pelvic area resulted in lower lymphocyte counts than the irradiation of the mediastinum. Therefore, the authors ruled out irradiation of the thymus as the cause of lymphopenia and suggested that the decrease in lymphocyte count following irradiation results from irradiation impact on the lymphocytes in the blood vessels.
To test the effects of extracorporeal irradiation of the blood, Weeke observed the irradiation of blood via a radioactive source placed within a dialysis unit, and showed that the degree of lymphocyte depletion was directly proportional to the radioactive source strength and amount of blood passing through the dialysis unit.28 MacLennan and Kay later revealed that the degree of long-term lymphopenia caused by a given 24–25 Gy of whole-brain irradiation was dependent upon the number of fractions into which the irradiation was divided.29 Because the brain and skull contain little bone marrow or lymphoid tissue, the main bulk of lymphocytes entering the fields during irradiation are those circulating in the blood.29 The number of fractions varied from 5 to 15 in this study, and the mean lymphocyte counts of patients 3 months after receiving this dose in 5 fractions was 1.84 × 109/l, in 12 fractions it was 1.12 × 109/l, and in 20 fractions it was 0.64 × 109/l.29 The observations by the researchers above strongly suggest that the radiation dose for circulating blood and bone marrow may be associated with the severity of RIL.
Effects of RIL on patients with EC
Peripheral circulating lymphocytes have a significant effect on survival outcomes in various types of solid tumors.30–32 In the past few years, many immune-inflammation parameters, such as absolute lymphocyte count (ALC), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR), have been confirmed to have prognostic values in several solid tumors treated with RT, but relatively few studies exist on this aspect for EC. Here, we summarize the prognostic value of RIL in six retrospective studies of 2374 patients with EC (Table 1).
Table 1.
Summary of studies of RIL in patients with EC.
| Author | Design/period | n | TNM stage | NLR cutoff (low/high) | PLR cutoff (low/high) | ALC (definition) | Blood exam timing | Treatment | Prognostic value analyses | Endpoint |
|---|---|---|---|---|---|---|---|---|---|---|
| Davuluri et al.12 | Retrospective (2007–2013) |
504 | I–III | NR | NR | Grade 4 ALC nadir: ALC < 200 cells/ml | Before, during (weekly), and 1 month after CRT | Neoadjuvant or definitive CRT ± surgery | Grade 4 ALC nadir | OS, PFS, DSS, DMFS |
| Barbetta et al.33 | Retrospective (2007–2013) |
217 | I–III | ΔNLR | NR | NR | Before and after CRT | 61.3%: CRT 38.7%: Neoadjuvant CRT + surgery |
ΔNLR | pCR, recurrence, DFS |
| Fang et al.18 | Retrospective (2007–2013) |
313 | I–IVa | NR | NR | High ALC > 0.35 × 10ˆ3/μl | Before, during (weekly), and one month after CRT | Neoadjuvant CRT + surgery | ALC | pCR |
| Deng et al.34 | Retrospective (2004–2015) |
755 | I–III | NR | NR | Grade 4 ALC nadir: ALC < 200 cells/ml |
Before, during (weekly), and at the first follow-up visit after CRT | Definitive or neoadjuvant CRT | Grade 4 ALC nadir | OS, LRFS, DMFS, DSS |
| Shiraishi et al.23 | Retrospective (2005–2016) |
480 | I–IVa | NR | NR | Grade 4 ALC nadir: ALC < 200 cells/ml |
During CRT | Neoadjuvant CRT + surgery | Grade 4 ALC nadir | OS, PFS, DMFS |
| Wu et al.35 | Retrospective (2013–2017) |
105 | Ia–IIIc | 4.35 (52/53) | 236 (52/53) | ALC% cutoff set at 16% | 1 week before the first and after the last radiotherapy or chemotherapy cycle | Concurrent CRT | PLR, NLR, ALC% | OS, TPD, TM |
ALC, absolute lymphocyte count; CRT, chemoradiation therapy; DFS, disease-free survival; DMFS, distant metastasis-free survival; DSS, disease-specific survival; EAC, esophageal adenocarcinoma; EC, esophageal cancer; ESCC, esophageal squamous cell carcinoma; LRFS, local-regional recurrence-free survival; N, number of patients; NLR, neutrophil-to-lymphocyte ratio; ΔNLR, posttreatment minus pretreatment NLR; NR, no report; OS, overall survival; pCR, pathologic complete response; PFS, progression-free survival; PLR, platelet-to-lymphocyte ratio; RIL, radiation-induced lymphopenia; TM, time to metastasis; TPD, time to progressive disease.
Short-term therapeutic evaluation
Two articles have reported correlations between immune-inflammation parameters and pathologic complete response (pCR), as shown in Table 1. Barbetta et al. assessed the before and after CRT variations of NLR (∆NLR) as a predictor of treatment response in patients with ESCC treated with CRT with or without surgery.33 Among surgical patients, 43% with pCR showed significantly lower median ∆NLR than patients with residual disease. High ∆NLR was a negative predictor of treatment response. On their part, Fang et al. examined patients with stages I-IVa EC (n = 313) treated with neoadjuvant CRT and then surgery.18 For patients with pCR, the median ALC nadir during CRT was significantly higher than that of patients without pCR. These two investigations both reiterate the fact that high levels of peripheral blood lymphocytes have a positive effect on short-term tumor treatment efficacy.
Recurrence and metastasis
Many researchers have reported determining findings on recurrence and metastasis in EC. Barbetta et al. reported ∆NLR as a prognostic factor for recurrence in their study.33 The median follow-up time after CRT was 5.2 years, there were recurrences in 106 patients, and 43 patients died without recurrence. High ∆NLR was significantly associated with an increased risk of recurrence. In a study of 755 patients with stages I–III EC who received concurrent CRT with or without surgery,34 Deng et al. obtained ALC before, during, and at first follow-up after CRT. Distant metastasis-free survival (DMFS) and local-regional recurrence-free survival (LRFS) were analyzed as the two endpoints. The researchers found that a Grade 4 ALC nadir during CRT was associated significantly with LRFS and DMFS. Per Davuluri et al.’s analysis of weekly ALC changes during the treatment of 504 patients with stages I–III EC subjected to neoadjuvant or definitive CRT,15 a Grade 4 ALC nadir during CRT predicted worse DMFS (p < 0.001), suggesting that there could be a role for host immunity in disease control. Shiraishi et al. reported lymphopenia in 480 EC patients undergoing neoadjuvant CRT,23 finding that a Grade 4 ALC nadir was significantly associated with reduced DMFS. Wu et al. evaluated PLR, NLR, and lymphocyte percentage before and after the concurrent CRT of stages IA-IIIC EC patients.35 The authors chose the time to metastasis (TM) as one of the endpoints in their reports, and their results demonstrated that lower lymphocyte percentage, higher PLR, and higher NLR were significantly associated with reduced TM. Through the studies herein discussed, there is an indication of a potential relationship between lymphocyte counts and a patient’s recurrence and metastasis.
Survival outcomes
Among the six studies in Table 1, only one research did not establish the relationship between immune-inflammation parameters and survival outcomes, with four studies utilizing overall survival (OS) as their observation endpoint. Suggestively, high NLR, high PLR, and low ALC tend to be associated with poor survival outcomes in general. Many published reports have indicated that a cancer-associated inflammatory response is linked to worse long-term results in a variety of solid tumors, including EC. Lymphocytes are at the center of these different anti-tumor immune-inflammation parameters. Therefore, per the reports we have listed, the protection and maintenance of a high level of lymphocyte counts during a treatment process may be beneficial to the survival outcomes of EC patients.
Organs-at-risk of lymphopenia
Hematological toxicities caused by RT include acute side effects stemming from the depletion of peripheral blood progenitor cells and chronic injury caused by alterations in vasculature and fibrosis in lymphoid organs, like bone marrow, thymus, and spleen.36,37 As a result, we can separate the OARs of lymphopenia for the RT of patients with EC into two categories. One class of OARs includes the heart, lungs, prominent blood vessels, and body with abundant blood circulation (Figure 2A). The other category of OARs consists of lymphoid and hematopoietic tissues, such as the spleen and bone marrow that may affect the development of lymphocyte hematopoiesis and maturation (Figure 2B).
Figure 2.
OARs of lymphopenia. (A) Red: GTV; blue: heart and blood vessel; yellow: lung; green: body. (B) Red: spinal cord; yellow: spleen. As the thymus gland gradually shrinks with age, it is not shown here.
GTV, gross tumor volume; OARs, organs-at-risk.
For the first category of OARs, lymphocytes receive a significant dose of radiation through large blood vessels that are contained in the radiation field during transit. This process includes systemic and pulmonary circulations. In line with this category of OARs, Tang et al. analyzed the relationships between lung dose-volume histogram (DVH) parameters with lymphocyte nadirs in 771 NSCLC patients and observed that lymphocyte nadirs were associated with the percentage of lung dose receiving 5 Gy (lung V5).38 In addition, the authors pointed out that the entire cardiac output was channeled through the lungs, causing increased circulating lymphocyte exposure. Saito et al. retrospectively assessed patients with various cancers who had undergone palliative RT using OARs based on body surface contour to evaluate their predictive value,39 and found that higher body dose-volume parameters and a more significant number of RT fractions were predictive of Grade 3 lymphopenia (absolute lymphocyte count <500 × 106/l).
The second category of OARs primarily includes organs rich in lymphoid and hemopoietic tissues. Saito et al. assessed the dosimetric predictors of treatment-related lymphopenia during CRT for EC and showed that higher spleen dose-volume parameters were associated with severe lymphopenia during CRT.40 An increase of 1 Gy in mean splenic dose predicted a 2.9% decrease in nadir ALC. Also, a study published in Radiotherapy and Oncology prospectively identified clinical and dosimetric predictors of both acute and late hematologic toxicity in chemo-naïve patients treated with whole-pelvis RT for prostate cancer.41 The results revealed that higher bone marrow V40 was associated with a higher risk of acute Grade 3 (ALC at nadir <500 × 106/l) or late Grade 2 lymphopenia (ALC at 1 year after RT conclusion <800 × 106/l and ⩾500 × 106/l). In addition, Newman et al. reported the dosimetric relationship between vertebral irradiation and the lymphopenia of EC patients treated with CRT.42 Multivariable linear regression showed that lymphopenia was associated with a greater volume of vertebral bodies receiving radiation during CRT.42 We can, therefore, surmise that the irradiation of lymphoid organs, such as the bone marrow, spleen, and regional lymph nodes, is a potential promoter of lymphopenia. According to the discussed studies, it might have a certain protective effect on lymphocytes if the irradiation of these OARs during RT could be avoided.
Risk avoidance of RIL in EC
The identification of the irradiation of lymphocyte OARs as a tool in causing lymphopenia facilitates the advancement of strategies to evaluate and adopt radiation treatment plans. The protection of lymphocyte OARs using approaches, such as the proton beam therapy (PBT) and the reduction of radiation doses, fractions, and field sizes, may help minimize RIL.
Radiation technology–proton beam therapy
The mechanisms of cell-kill for all forms of radiation are similar, but proton particles predictably lose energy as they pass through the body compared with photon beams; this property could be exploited to optimize clinical outcomes.43 Consequently, the depth of the termination of the proton beam is controllable, and the radiation dose beyond the target region can be restricted. Due to the esophagus’s central location near several OARs of lymphopenia, including the heart, lungs, and spine, the ability of PBT to conform high radiation dose to the tumor volume while reducing the unintentional radiation dose to adjacent healthy tissues has the potential to decrease RIL.44
An example of the potency of PBT is illustrated in a study performed at the MD Anderson Cancer Center that retrospectively evaluated 480 patients with EC treated with preoperative concurrent CRT using PBT or intensity modulated radiation therapy (IMRT) with or without induction chemotherapy followed by surgery.23 The purpose of the analysis was to compare the relative risk of RT-induced lymphopenia between PBT and IMRT. Findings from the study revealed that a higher proportion of IMRT patients (40.4%) developed Grade 4 lymphopenia during neoadjuvant CRT, compared with PBT patients (17.6%, p < 0.0001). Per the multivariable analysis of the research, PBT was associated significantly with a reduced risk of Grade 4 lymphopenia. In another study, the assessment of patients treated with definitive CRT and either PBT or IMRT for EC showed that PBT reduced the risk of severe, treatment-related lymphopenia, particularly in tumors of the lower esophagus.45 In Routman et al.’s comparison of Grade 4 lymphopenia in patients who received concurrent CRT using PRT or photon RT with EC,46 the authors found that photon RT was associated with a significantly higher risk of Grade 4 lymphopenia, compared with PRT. These reports demonstrate that PRT might be one such way to decrease RIL in EC patients.
In another study, Hirano et al. retrospectively reviewed 37 patients with Stage III thoracic ESCC who had received PBT with or without concurrent chemotherapy,47 comparing the dose distributions of PBT with those of dummy 3D-CRT and IMRT, primarily focusing on the doses to OARs, such as healthy lungs and heart tissue. The authors’ results indicated that PBT significantly reduced the doses to the lungs and heart, compared with 3D-CRT or IMRT. As the significantly reduced doses exit through the lungs and heart, the physics of PBT is ideally more suited for EC than that of photon RT.46 PBT would, therefore, seem to hold significant clinical advantages over traditional photon RT.
Radiation dosage and fractions
Yovino et al. devised a typical glioblastoma plan (8-cm tumor, 60 Gy/30 fractions) to estimate the radiation dose that lymphocytes receive from the radiation field.48 Per this model, a single radiation fraction was found to deliver 0.5 Gy to 5% of circulating cells, and 99% of circulating blood received at least 0.5 Gy (mean dose of 2.2 Gy) after 30 fractions. Concurrently, a decrease in the fractionation of RT has been confirmed in some studies to reduce the incidence of lymphopenia risk. Crocenzi et al. analyzed the impact of neoadjuvant standard fractionated and hypofractionated CRT on lymphocytes in patients with locally advanced and borderline resectable pancreatic adenocarcinoma,49 and found that standard fractionated CRT (a total dose of 50.4 Gy in 28 fractions over 5.5 weeks) resulted in a significant loss of lymphocytes, and hypofractionated CRT (a total dose of 30 Gy in 3 fractions over 1 week) was associated with reduced systemic loss of T cells. These studies suggest that the reduction of the duration of exposure or short-course radiation with a hypofractionated schedule could mitigate normal tissue exposure and reduce the risk of RIL. For thoracic RT, the RTOG-0617 trial showed that higher doses were associated with worse OS and might be potentially harmful.50 According to Ladbury et al.,51 higher doses of radiation to the immune system (calculated as a function of the number of radiation fractions and mean doses to the lung, heart, and the rest of the body based on a model developed by Jin et al.) were associated with tumor progression and worse OS after the definitive treatment of stage III NSCLC.52 It is, therefore, reasonable to infer that the standard radiation dose of 50.4 Gy for EC has fewer effects on lymphocytes than a dose of over 60 Gy. Using a higher radiation dose for EC does not always guarantee a better outcome since the response rate and survival outcomes do not necessarily improve.
Radiation target volume
Many studies have performed detailed analyses of radiation field size parameters as predictors of lymphopenia risk in recent years. Tang et al. analyzed the relationships between GTVs and lymphocyte nadirs in 771 NSCLC patients who had received definitive RT and found that larger GTVs correlated with fewer lymphocyte nadirs.38 In 2018, further proof of the effects of field size on lymphopenia risk was provided by a study on 210 glioblastoma patients who were treated with standard-field RT (T1 enhancement + surgical cavity + T2 abnormality + 1.3–2.5 cm margin) versus limited-field RT (T1 enhancement + surgical cavity + 1.8–2 cm margin).13 The results revealed that V25 Gy of the brain was an independent predictor of acute severe lymphopenia in glioblastoma patients receiving 60 Gy of RT with concurrent temozolomide and that a reduction in the volume of the brain irradiated may lead to less treatment-induced lymphopenia. Ellsworth et al. also discussed field size effects on RIL,53 finding that RIL risk was associated with field size, dose per fraction, and fraction number.
As mentioned earlier, one of the most controversial points in the radiation target volume of EC is whether to opt for ENI or IFI. ENI involves delivering RT to the primary tumor, as well as the irradiation of clinically uninvolved regional lymph nodes at risk of micrometastases of the treated disease.54 The evidence of ENI was derived from prophylactic lymph node dissection in Japan. At operation, metastasis was found frequently in the supraclavicular nodes.55 Therefore, the researchers used a three-field lymph node dissection approach to eliminate subclinical metastases, which resulted in better survival following a reduction in the regional lymph node recurrence rates and elimination of micrometastases.56,57 In the RTOG 85-01 randomized trial,2 the irradiation of the entire esophagus, from the supraclavicular fossae to the esophagogastric junction for EC with the initial tumor length plus a 5 cm margin, was recommended. In the RTOG 94-05 trial,4 an area with a margin of 2–5 cm surrounding the gross tumor volume (GTV) was defined as the CTV; the primary tumor and regional lymph nodes were included. For tumors of the cervical esophagus, supraclavicular lymph nodes were covered in the RT field. However, the results of these two trials did not improve regional control or survival significantly. A three-dimensional conformal radiation therapy (3D-CRT) technology was subsequently applied in the RTOG 01-13 trial58; CTV included GTV, a 3 cm cephalad and caudad margin beyond GTV, and locoregional lymph nodes. Planning target volume (PTV) in this trial was defined as having 2 cm margins around CTV and 2 cm superior and inferior margins beyond CTV.
Similarly, many studies have investigated the recurrence pattern and survival outcome of ENI and have found that ENI serves to prevent localized regional failure rather than improve the long-term survival of patients with EC.59–62 In summary, there is no universal recommendation for the radiation field size selection for EC patients. Because the target volume of EC covers many large blood vessels and lymphoid tissues in the cervicothoracic region, it is reasonable to infer that if we reduce the target volume, the exposure of peripheral circulating lymphocytes to radiation will decrease. Large target volumes of ENI can increase treatment-related toxicities, such as hematological adverse events, acute radiation esophagitis, and late cardiopulmonary toxicities.63 In addition, greater PTV is associated with an increased risk of grade 4 lymphopenia in EC.45 Currently, there is not much evidence on the effects of field size on lymphocytes subjected to RT in patients with EC. Nevertheless, a small target volume could miss some subclinical disease, but with the help of modern imaging technology, IFI does not reduce OS compared with ENI. Many studies have demonstrated that the predominant failure pattern after IFI is still the in-field and distant disease rather than the out-field regional failure.64–67
Prospects
Combining radiotherapy with immunotherapy in EC
Since the approval of anti-CTLA4 therapy (ipilimumab) for unresectable or metastatic melanoma on 25 March 2011,68 the development of anti-cancer immunotherapy has accelerated. The success of immunotherapy has improved the prognosis of some types of malignancies considerably,69,70 which has greatly encouraged researchers to combine the treatment with other conventional treatment strategies to further improve their efficacy. Among the treatment blends, the combination of RT and immunotherapy is considered promising.71
Immune checkpoints are inhibitory pathways hardwired into the immune system that are crucial for the maintenance of self-tolerance and also protect healthy tissues from damage when the immune system is responding to pathogenic infection.19 A growing number of clinical trials have confirmed that blocking inhibitory immune regulatory proteins ultimately results in immune activation against tumor cells.72 RT can effectively alter and eliminate both tumor cells and the surrounding stromal cells.73 RT has a unique advantage against tumor immune escape mechanisms by increasing antigen visibility.71 This advantage includes the enhancement of the clearance of damaged tumor cells by antigen-presenting cells, thereby promoting the activation of T-cells;71 the upregulation of the expression of MHC-I on tumor surfaces to enhance the visibility of tumors to cytotoxic T-cells,74 thus inducing the T-cell mediated abscopal effect and the possibility of RT-induced DNA damages generating neoantigen and enhancing the immune surveillance.75,76 An RT-induced modulation of tumor microenvironments can promote the recruitment and infiltration of immune cells and stimulate the recognition and killing of tumor cells by the immune system.77 Preclinical studies of mouse models by Deng et al. demonstrated that PD-L1 expression is upregulated in tumor microenvironments after RT, and combining of RT with PD-L1 checkpoint blockade could synergistically reduce the local accumulation of tumor-infiltrating myeloid-derived suppressor cells.78 According to Chen et al., RT increased PD-L1 expression in human EC cells through in vitro experiments.79 In addition, many studies have shown that RT can not only increase tumor antigen presentation but also enhance checkpoint inhibitor-induced antitumor immune responses.74,80,81 The above mechanisms are the rationale for combining RT with ICPi.
In this review, we have mainly summarized the immunosuppressive effect of RIL in patients with EC. It is crucial to maintain an intact adaptive immune system during cancer therapy to enhance the effectiveness of immunotherapy and improve cancer control.23 For a better combination of RT with ICPi, it is necessary to reduce the immunosuppressive effect of RIL. In Marciscano et al.’s preclinical model to evaluate the immunological differences of RT strategies with or without ENI and highlight the effect of field size on lymphocyte subpopulation from an immunological perspective54; the authors found that RT correlated with the up-regulation of an intratumoral T-cell chemoattractant chemokine signature (CXCR3, CCR5-related), which resulted in a robust infiltration of multiple subpopulations of lymphocytes. The addition of ENI reduced the expression of chemokines, inhibited immune infiltration, and adversely impacted survival when combined with ICPi. In yet another study, Pike et al. reported that prolonged courses (>five fractions) of RT increased the risk of severe lymphopenia, which was associated with more reduced survival in patients treated with ICPi.82 A study by Won Jin Ho demonstrated that pretreatment ALC is significantly associated with the response to PD1 inhibitors in patients with recurrent and/or metastatic head and neck squamous cell carcinoma.83 Patients with pretreatment ALC < 600 cells/μl were found to have significantly shorter PFS than patients with pretreatment ALC ⩾ 600 cells/μl.
More and more emerging evidence is showing that the choice of radiation dose, fractionation, and target volume plays a crucial role in the combination of RT with other treatment measures. It would, therefore, be beneficial to know whether some radiation regimens (optimum dose, fractionation, target site, timing, and target volume) have superior effects over others in terms of immune stimulation. We can infer from the studies discussed in this review that small radiation field sizes and shorter radiation courses appear to promote combinatorial efficacy in RT and immunotherapy.
Currently, immunotherapy is not the standard treatment for EC. It is, to that effect, unwise to change the radiation regimen blindly. A few clinical trials using RT combined with immunotherapy for EC are underway (Table 2).77,84,85 We expect to explore a better RT regimen through these clinical trials to maximize the anti-tumor response from the combination of RT with immunotherapy. The ultimate challenge is to integrate cancer immunotherapy into RT optimally. RT could quite possibly be an immunologic adjuvant if the RT plan is right.86
Table 2.
Ongoing clinical trials of radiotherapy combined with immunotherapy for EC.
| Clinical Trial. Gov number. | Phase/line | Condition | Target | Arms |
|---|---|---|---|---|
| NCT 03044613 | I/Neoadjvant | Operable Stage II/III Esophageal Gastroesophageal Junction Cancer |
PD-1 | Arm A: Nivolumab + carboplatin/paclitaxel + radiation Arm B: Nivolumab + relatlimab + carboplatin/paclitaxel + radiation |
| NCT 02830594 | II/Salvage | Adenocarcinoma of the gastroesophageal junction, gastric cancer, EC, metastatic malignant neoplasm in the stomach | PD-1 | Single-arm: pembrolizumab + palliative radiation |
| NCT 02642809 | I/1st, 2nd | Metastatic EC | PD-1 | Single-arm: pembrolizumab + radiation (brancytherapy 2 fractions × 8 Gy) |
| NCT 03087864 | II/1st | Stage II/III EC | PD-L1 | Single-arm: atezolizumab + carboplatin + paclitaxel + radiation (23 × 1.8 Gy) |
| NCT 02844075 | II/Neoadjvant | ESCC | PD-1 | Single-arm: pembrolizumab + taxol + carboplatin + radiation (21 × 2.1 Gy) + surgery |
| NCT 03064490 | II/Neoadjvant | Locally advanced EC and gastric cancer | PD-1 | Single-arm: weekly neoadjuvant pembrolizumab + concurrent CRT [carboplatin/paclitaxel + radiation (25 × 1.8 Gy)] + surgery |
| NCT 03278626 | I, II/1st | Locally advanced ESCC | PD-1 | Single-arm: Nivolimumab + Carboplatin/paclitaxel + Radiation (28 × 1.8 Gy) |
| NCT 03544736 | I, II/– | EC | PD-1 | Cohort A: Nivolumab + palliative radiation (2 Gy/day, to a total of 30–50 Gy) Cohort B: Nivolumab + definitive chemoradiotherapy [carboplatin/paclitaxel + radiation(23 × 1.8 Gy)] Cohort C: Nivolumab + neoadjuvant chemoradiotherapy [carboplatin/paclitaxel + radiation (23 × 1.8 Gy)] + surgery |
| NCT 03437200 | II/– | Inoperable EC | PD-1 | Arm A: Nivolumab + FOLFOX + radiation (25 × 2 Gy) Arm B: Nivolumab + Ipilimumab + FOLFOX + radiation (25 × 2 Gy) |
| NCT 03490292 | I, II/neoadjvant | Resectable EC Gastroesophageal cancer |
PD-L1 | Single-arm: Avelumab + Carboplatin/paclitaxel + radiation (25f) |
| NCT 02520453 | II/adjuvant | EC | PD-L1 | Neoadjuvant concurrent CRT + surgery + durvalumab versus neoadjuvant concurrent CRT + surgery + placebo |
| NCT 03377400 | II/1st, 2nd | ESCC | PD-L1 | Single-arm: durvalumab/tremelimumab + concurrent radiotherapy (5FU/CDDP) |
CDDP, cisplatin; CRT, chemoradiation therapy; EC, esophageal cancer; ESCC, esophageal squamous cell carcinoma; 5FU, 5-fluorouracil; NCT, national clinical trial.
Conclusion
RT can cause lymphopenia, and RIL is associated significantly with a poor outcome in EC patients. Tailoring RT regimens to spare the immune system may be an important future direction to improve prognosis in the EC population. Hence, it is critical to find effective treatment strategies to prevent or mitigate RIL in patients with EC. The RT target volume of EC is extensive, including the cervical, mediastinal, and upper abdominal nodes, which could be contained in multiple OARs of lymphopenia. To date, combining RT with immunotherapy has not only proven effective in preclinical studies but has also shown promise in clinical trials. To optimize RT regimens in the context of immunotherapy, several factors need to be considered: target volume, optimal dose and fractionation, and timing. We hope that the optimization of RT plans will further enhance the effectiveness of this combination. From the current evidence, the use of IFI, a radiation dose of 50.4 Gy, and the PBT technology could embody reasonable treatment strategies to reduce the incidence of RIL in EC. Further research is required to test this hypothesis.
Acknowledgments
The authors would such as to express their great thanks to the Natural Science Foundation of China and the Key Research and Development Program of Shandong Province.
Footnotes
Author contributions: MHL and JMY designed the study. XW drafted the manuscript. PLW, ZXZ, QFM coordinated, edited, and finalized the drafting of the manuscript. All authors read and approved the final manuscript.
Availability of data and material: The dataset supporting the conclusions of this article is included within the article.
Conflict of interest statement: The authors declare that there is no conflict of interest.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was funded by the Natural Science Foundation of China (NSFC 81672995) and the Key Research and Development Program of Shandong Province (2016GSF201133).
Contributor Information
Xin Wang, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
Peiliang Wang, Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; School of Medicine, Shandong University, Jinan, Shandong, China.
Zongxing Zhao, Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; School of Medicine, Shandong University, Jinan, Shandong, China.
Qingfeng Mao, Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; School of Medical and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, China.
Jinming Yu, Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
Minghuan Li, Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jiyan Road 440, Jinan, Shandong province 250117, China.
References
- 1. Fitzmaurice C, Dicker D, Pain A, et al. The Global Burden of Cancer 2013. JAMA Oncol 2015; 1: 505–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Cooper JS, Guo MD, Herskovic A, et al. Chemoradiotherapy of locally advanced esophageal cancer: long-term follow-up of a prospective randomized trial (RTOG 85-01). Radiation therapy oncology group. JAMA 1999; 281: 1623–1627. [DOI] [PubMed] [Google Scholar]
- 3. National Comprehensive Cancer Network. Clinical practice guidelines in oncology. Esophageal and esophagogastric junction cancers version 3, https://www.nccn.org/professionals/physician_gls/default.aspx#esophageal (2019, accessed 18 November 2019). [DOI] [PubMed]
- 4. Minsky BD, Pajak TF, Ginsberg RJ, 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: 1167–1174. [DOI] [PubMed] [Google Scholar]
- 5. Higuchi K, Koizumi W, Tanabe S, et al. Current management of esophageal squamous-cell carcinoma in Japan and other countries. Gastrointest Cancer Res 2009; 3: 153–161. [PMC free article] [PubMed] [Google Scholar]
- 6. Meng X, Wang J, Sun X, et al. Cetuximab in combination with chemoradiotherapy in Chinese patients with non-resectable, locally advanced esophageal squamous cell carcinoma: a prospective, multicenter phase II trail. Radiother Oncol 2013; 109: 275–280. [DOI] [PubMed] [Google Scholar]
- 7. Prenzel KL, Bollschweiler E, Schroder W, et al. Prognostic relevance of skip metastases in esophageal cancer. Ann Thorac Surg 2010; 90: 1662–1667. [DOI] [PubMed] [Google Scholar]
- 8. Van De Voorde L, Larue RT, Pijls M, et al. A qualitative synthesis of the evidence behind elective lymph node irradiation in oesophageal cancer. Radiother Oncol 2014; 113: 166–174. [DOI] [PubMed] [Google Scholar]
- 9. Trowell OA. The sensitivity of lymphocytes to ionising radiation. J Pathol Bacteriol 1952; 64: 687–704. [DOI] [PubMed] [Google Scholar]
- 10. Campian JL, Ye X, Brock M, et al. Treatment-related lymphopenia in patients with stage III non-small-cell lung cancer. Cancer Invest 2013; 31: 183–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. van Meir H, Nout RA, Welters MJ, et al. Impact of (chemo)radiotherapy on immune cell composition and function in cervical cancer patients. Oncoimmunology 2017; 6: e1267095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Yousefi H, Yuan J, Keshavarz-Fathi M, et al. Immunotherapy of cancers comes of age. Expert Rev Clin Immunol 2017; 13: 1001–1015. [DOI] [PubMed] [Google Scholar]
- 13. Rudra S, Hui C, Rao YJ, et al. Effect of radiation treatment volume reduction on lymphopenia in patients receiving chemoradiotherapy for glioblastoma. Int J Radiat Oncol Biol Phys 2018; 101: 217–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Cho O, Oh YT, Chun M, et al. Radiation-related lymphopenia as a new prognostic factor in limited-stage small cell lung cancer. Tumour Biol 2016; 37: 971–978. [DOI] [PubMed] [Google Scholar]
- 15. Davuluri R, Jiang W, Fang P, et al. Lymphocyte nadir and esophageal cancer survival outcomes after chemoradiation therapy. Int J Radiat Oncol Biol Phys 2017; 99: 128–135. [DOI] [PubMed] [Google Scholar]
- 16. 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: 259–265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Cho O, Chun M, Chang SJ, et al. Prognostic value of severe lymphopenia during pelvic concurrent chemoradiotherapy in cervical cancer. Anticancer Res 2016; 36: 3541–3547. [PubMed] [Google Scholar]
- 18. Fang P, Jiang W, Davuluri R, et al. High lymphocyte count during neoadjuvant chemoradiotherapy is associated with improved pathologic complete response in esophageal cancer. Radiother Oncol 2018; 128: 584–590. [DOI] [PubMed] [Google Scholar]
- 19. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 2012; 12: 252–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Cho Y, Miyamoto M, Kato K, et al. CD4+ and CD8+ T cells cooperate to improve prognosis of patients with esophageal squamous cell carcinoma. Cancer Res 2003; 63:1555–1559. [PubMed] [Google Scholar]
- 21. Schumacher K, Haensch W, Röefzaad C, et al. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res 2001; 61: 3932–3936. [PubMed] [Google Scholar]
- 22. Menetrier-Caux C, Ray-Coquard I, Blay JY, et al. Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer 2019; 7: 85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Shiraishi Y, Fang P, Xu C, et al. Severe lymphopenia during neoadjuvant chemoradiation for esophageal cancer: a propensity matched analysis of the relative risk of proton versus photon-based radiation therapy. Radiother Oncol 2018; 128: 154–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Nakamura N, Kusunoki Y, Akiyama M. Radiosensitivity of CD4 or CD8 positive human T-lymphocytes by an in vitro colony formation assay. Radiat Res 1990; 123: 224–227. [PubMed] [Google Scholar]
- 25. Meyer KK. Radiation-induced lymphocyte-immune deficiency. A factor in the increased visceral metastases and decreased hormonal responsiveness of breast cancer. Arch Surg 1970; 101: 114–121. [DOI] [PubMed] [Google Scholar]
- 26. Stjernsward J, Jondal M, Vanky F, et al. Lymphopenia and change in distribution of human B and T lymphocytes in peripheral blood induced by irradiation for mammary carcinoma. Lancet 1972; 1: 1352–1356. [DOI] [PubMed] [Google Scholar]
- 27. Raben M, Walach N, Galili U, et al. The effect of radiation therapy on lymphocyte subpopulations in cancer patients. Cancer 1976; 37: 1417–1421. [DOI] [PubMed] [Google Scholar]
- 28. Weeke E. The development of lymphopenia in uremic patients undergoing extracorporeal irradiation of the blood with portable beta units. Radiat Res 1973; 56: 554–559. [PubMed] [Google Scholar]
- 29. MacLennan IC, Kay HE. Analysis of treatment in childhood leukemia. IV. The critical association between dose fractionation and immunosuppression induced by cranial irradiation. Cancer 1978; 41: 108–111. [DOI] [PubMed] [Google Scholar]
- 30. Venkatesulu BP, Mallick S, Lin SH, et al. A systematic review of the influence of radiation-induced lymphopenia on survival outcomes in solid tumors. Crit Rev Oncol Hematol 2018; 123: 42–51. [DOI] [PubMed] [Google Scholar]
- 31. Ray-Coquard I, Cropet C, Van Glabbeke M, et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res 2009; 69: 5383–5391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. 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: 1225–1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Barbetta A, Nobel TB, Sihag S, et al. Neutrophil to lymphocyte ratio as predictor of treatment response in esophageal squamous cell cancer. Ann Thorac Surg 2018; 106: 864–871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Deng W, Xu C, Liu A, et al. The relationship of lymphocyte recovery and prognosis of esophageal cancer patients with severe radiation-induced lymphopenia after chemoradiation therapy. Radiother Oncol 2019; 133: 9–15. [DOI] [PubMed] [Google Scholar]
- 35. Wu YF, Chu SC, Chang BS, et al. Hematologic markers as prognostic factors in nonmetastatic esophageal cancer patients under concurrent chemoradiotherapy. Biomed Res Int 2019; 2019: 1263050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Mauch P, Constine L, Greenberger J, et al. Hematopoietic stem cell compartment: acute and late effects of radiation therapy and chemotherapy. Int J Radiat Oncol Biol Phys 1995; 31: 1319–1339. [DOI] [PubMed] [Google Scholar]
- 37. Chin AL, Aggarwal S, Pradhan P, et al. The role of bone marrow and spleen irradiation in the development of acute hematologic toxicity during chemoradiation for esophageal cancer. Adv Radiat Oncol 2018; 3: 297–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. 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: 1084–1091. [DOI] [PubMed] [Google Scholar]
- 39. Saito T, Toya R, Matsuyama T, et al. Dosimetric predictors of treatment-related lymphopenia induced by palliative radiotherapy: predictive ability of dose-volume parameters based on body surface contour. Radiol Oncol 2017; 51: 228–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Saito T, Toya R, Yoshida N, et al. Spleen dose-volume parameters as a predictor of treatment-related lymphopenia during definitive chemoradiotherapy for esophageal cancer. In Vivo 2018; 32: 1519–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Sini C, Fiorino C, Perna L, et al. Dose-volume effects for pelvic bone marrow in predicting hematological toxicity in prostate cancer radiotherapy with pelvic node irradiation. Radiother Oncol 2016; 118: 79–84. [DOI] [PubMed] [Google Scholar]
- 42. Newman NB, Sherry AD, Anderson J, et al. Dose-dependent association of vertebral body irradiation and lymphopenia during chemoradiation for esophageal cancer. Int J Radiat Oncol Biol Phys 2018; 102: e37–e38. [Google Scholar]
- 43. Kaiser A, Eley JG, Onyeuku NE, et al. Proton therapy delivery and its clinical application in select solid tumor malignancies. J Vis Exp. Epub ahead of print 6 February 2019. DOI: 10.3791/58372. [DOI] [PubMed] [Google Scholar]
- 44. Kang JI, Jr, Slater JD, Yang GY. Proton therapy for esophageal cancer. Thorac Cancer 2012; 3: 91–93. [DOI] [PubMed] [Google Scholar]
- 45. Fang P, Shiraishi Y, Verma V, et al. Lymphocyte-sparing effect of proton therapy in patients with esophageal cancer treated with definitive chemoradiation. Int J Part Ther 2018; 4: 23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Routman DM, Garant A, Lester SC, et al. A comparison of grade 4 lymphopenia with proton versus photon radiation therapy for esophageal cancer. Adv Radiat Oncol 2019; 4: 63–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Hirano Y, Onozawa M, Hojo H, et al. Dosimetric comparison between proton beam therapy and photon radiation therapy for locally advanced esophageal squamous cell carcinoma. Radiat Oncol 2018; 13: 23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Yovino S, Kleinberg L, Grossman SA, et al. 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: 140–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Bradley JD, Paulus R, Komaki R, 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: 187–199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ladbury CJ, Rusthoven CG, Camidge DR, et al. 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: 346–355. [DOI] [PubMed] [Google Scholar]
- 52. Jin JY, Hu C, Xiao Y, et al. Higher radiation dose to immune system is correlated with poorer survival in patients with stage III non–small cell lung cancer: a secondary study of a phase 3 cooperative group trial (NRG oncology RTOG 0617). Int J Radiat Oncol Biol Phys 2017; 99: S151–S152. [Google Scholar]
- 53. 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: 512–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Marciscano AE, Ghasemzadeh A, Nirschl TR, et al. Elective nodal irradiation attenuates the combinatorial efficacy of stereotactic radiation therapy and immunotherapy. Clin Cancer Res 2018; 24: 5058–5071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Sannohe Y, Hiratsuka R, Doki K. Lymph node metastasis in cancer of the thoracic esophagus. Am J Surg 1981; 141: 216–218. [DOI] [PubMed] [Google Scholar]
- 56. Fujita H, Kakegawa T, Yamana H, et al. Mortality and morbidity rates, postoperative course, quality of life, and prognosis after extended radical lymphadenectomy for esophageal cancer. Comparison of three-field lymphadenectomy with two-field lymphadenectomy. Ann Surg 1995; 222: 654–662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Kato H, Watanabe H, Tachimori Y, et al. Evaluation of neck lymph node dissection for thoracic esophageal carcinoma. Ann Thorac Surg 1991; 51: 931–935. [DOI] [PubMed] [Google Scholar]
- 58. Ajani JA, Winter K, Komaki R, et al. Phase II randomized trial of two nonoperative regimens of induction chemotherapy followed by chemoradiation in patients with localized carcinoma of the esophagus: RTOG 0113. J Clin Oncol 2008; 26: 4551–4556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Yamashita H, Takenaka R, Omori M, et al. Involved-field radiotherapy (IFRT) versus elective nodal irradiation (ENI) in combination with concurrent chemotherapy for 239 esophageal cancers: a single institutional retrospective study. Radiat Oncol 2015; 10: 171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Onozawa M, Nihei K, Ishikura S, et al. Elective nodal irradiation (ENI) in definitive chemoradiotherapy (CRT) for squamous cell carcinoma of the thoracic esophagus. Radiother Oncol 2009; 92: 266–269. [DOI] [PubMed] [Google Scholar]
- 61. Yamashita H, Okuma K, Wakui R, et al. Details of recurrence sites after elective nodal irradiation (ENI) using 3D-conformal radiotherapy (3D-CRT) combined with chemotherapy for thoracic esophageal squamous cell carcinoma–a retrospective analysis. Radiother Oncol 2011; 98: 255–260. [DOI] [PubMed] [Google Scholar]
- 62. Cheng YJ, Jing SW, Zhu LL, et al. Comparison of elective nodal irradiation and involved-field irradiation in esophageal squamous cell carcinoma: a meta-analysis. J Radiat Res 2018; 59: 604–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Morota M, Gomi K, Kozuka T, et al. Late toxicity after definitive concurrent chemoradiotherapy for thoracic esophageal carcinoma. Int J Radiat Oncol Biol Phys 2009; 75: 122–128. [DOI] [PubMed] [Google Scholar]
- 64. Zhao KL, Ma JB, Liu G, et al. Three-dimensional conformal radiation therapy for esophageal squamous cell carcinoma: is elective nodal irradiation necessary? Int J Radiat Oncol Biol Phys 2010; 76: 446–451. [DOI] [PubMed] [Google Scholar]
- 65. Li M, Zhang X, Zhao F, et al. Involved-field radiotherapy for esophageal squamous cell carcinoma: theory and practice. Radiat Oncol 2016; 11:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Li DJ, Li HW, He B, et al. Patterns of failure after involved field radiotherapy for locally advanced esophageal squamous cell carcinoma. J BUON 2016; 21: 1268–1273. [PubMed] [Google Scholar]
- 67. Zhang X, Li M, Meng X, et al. Involved-field irradiation in definitive chemoradiotherapy for locally advanced esophageal squamous cell carcinoma. Radiat Oncol 2014; 9: 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Culver ME, Gatesman ML, Mancl EE, et al. Ipilimumab: a novel treatment for metastatic melanoma. Ann Pharmacother 2011; 45: 510–519. [DOI] [PubMed] [Google Scholar]
- 69. Karlsson AK, Saleh SN. Checkpoint inhibitors for malignant melanoma: a systematic review and meta-analysis. Clin Cosmet Investig Dermatol 2017; 10: 325–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Herzberg B, Campo MJ, Gainor JF. Immune checkpoint inhibitors in non-small cell lung cancer. Oncologist 2017; 22: 81–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Wang Y, Deng W, Li N, et al. Combining immunotherapy and radiotherapy for cancer treatment: current challenges and future directions. Front Pharmacol 2018; 9: 185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Ramsay AG. Immune checkpoint blockade immunotherapy to activate anti-tumour T-cell immunity. Br J Haematol 2013; 162: 313–325. [DOI] [PubMed] [Google Scholar]
- 73. Menon H, Ramapriyan R, Cushman TR, et al. Role of radiation therapy in modulation of the tumor stroma and microenvironment. Front Immunol 2019; 10: 193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med 2006; 203: 1259–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Demaria S, Ng B, Devitt ML, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys 2004; 58: 862–870. [DOI] [PubMed] [Google Scholar]
- 76. Germano G, Lamba S, Rospo G, et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature 2017; 552: 116–120. [DOI] [PubMed] [Google Scholar]
- 77. Liao XY, Liu CY, He JF, et al. Combination of checkpoint inhibitors with radiotherapy in esophageal squamous cell carcinoma treatment: a novel strategy. Oncol Lett 2019; 18: 5011–5021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice. J Clin Invest 2014; 124: 687–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Chen MF, Chen PT, Chen WC, et al. The role of PD-L1 in the radiation response and prognosis for esophageal squamous cell carcinoma related to IL-6 and T-cell immunosuppression. Oncotarget 2016; 7: 7913–7924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Demaria S, Kawashima N, Yang AM, et al. Immune-mediated inhibition of metastases after treatment with local radiation and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res 2005; 11: 728–734. [PubMed] [Google Scholar]
- 81. Zhang B, Bowerman NA, Salama JK, et al. Induced sensitization of tumor stroma leads to eradication of established cancer by T cells. J Exp Med 2007; 204: 49–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Pike LRG, Bang A, Mahal BA, 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: 142–151. [DOI] [PubMed] [Google Scholar]
- 83. Ho WJ, Yarchoan M, Hopkins A, et al. Association between pretreatment lymphocyte count and response to PD1 inhibitors in head and neck squamous cell carcinomas. J Immunother Cancer 2018; 6: 84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Mimura K, Yamada L, Ujiie D, et al. Immunotherapy for esophageal squamous cell carcinoma: a review. Fukushima J Med Sci 2018; 64: 46–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Liu Y, Dong Y, Kong L, et al. Abscopal effect of radiotherapy combined with immune checkpoint inhibitors. J Hematol Oncol 2018; 11: 104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Schaue D, Ratikan JA, Iwamoto KS, et al. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys 2012; 83: 1306–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]

