| Home | E-Submission | Sitemap | Contact Us |  
top_img
Radiation Oncology Journal > Volume 41(1); 2023 > Article
Novikov, Baldueva, Zozulya, Emelyanova, Girdyuk, Arsenyev, Alexandrovna, Tyuryaeva, Antipov, Girshovich, and Kanayev: Peripheral blood lymphocyte changes after stereotactic ablative body radiotherapy to lung or liver metastases in patients with oligometastatic cancers

Abstract

Purpose

To perform the analysis of the peripheral blood lymphocyte changes after stereotactic ablative radiotherapy (SABR) in patients with oligometastatic cancers.

Materials and Methods

The dynamics of the immune status in peripheral blood was prospectively evaluated in 46 patients with lung (17 cases) or liver (29 cases) metastases treated by SABR. Flow cytometry of peripheral blood lymphocyte subpopulations was performed before SABR, 3–4 weeks and 6–8 weeks after the end of SABR: 3 fractions of 15–20 Gy or 4 fractions of 13.5 Gy. The number of treated lesions varied from 1 (32 patients) to 2–3 (14 patients).

Results

SABR induced a significant increase of T-lymphocytes (CD3+CD19) (p = 0.001), T-helper (CD3+CD4+) (p = 0.004), activated cytotoxic T-lymphocytes (CD3+CD8+HLA-DR+) (p = 0.001), activated T-helpers (CD3+CD4+HLA-DR+) (p < 0.001). A significant decrease of T-regulated immune suppressive lymphocytes (CD4+CD25brightCD127low) (p = 0.002) and NKT-cells (CD3+CD16+CD56+) (p = 0.007) was recorded after the SABR. The comparative analysis demonstrated that lower doses of SABR (EQD2Gy(α/β=10) = 93.7–105.7 Gy) induced significant increase of T-lymphocytes, activated cytotoxic T-lymphocytes, and activated CD4+CD25+ T-helpers, while SABR with higher doses (EQD2Gy(α/β=10) = 150 Gy) was not associated with these effects. A more efficient activations of T-lymphocytes (p = 0.010), activated T-helpers (p < 0.001), and cytotoxic T-lymphocytes (p = 0.003) were associated with SABR to a single lesion. A significant increase of T-lymphocytes (p = 0.002), T-helpers (p = 0.003), and activated cytotoxic T-lymphocytes (p = 0.001) was observed after SABR for hepatic metastases in contrast to SABR for lung lesions.

Conclusion

Changes in peripheral blood lymphocytes after SABR could be influenced by the location or the number of irradiated metastasis, and the dose of SABR.

Introduction

Radiotherapy is an important and obligatory component of treatment programs in 50%–70% patients with cancer [1]. It has been traditionally considered as a method that improved the locoregional control. Radiobiologically, the radiation exposure is perceived as a cytotoxic treatment causing DNA breaks with lethal events that manifested by cell death through necrosis, apoptosis, and autophagy [2]. During the last decades, there appeared numerous evidence that radiotherapy and especially stereotactic ablative radiotherapy (SABR) are also characterized by systemic effects mainly represented by antitumor immune response usually called an abscopal effect.
A certain number of hypotheses tried to explain systemic effects of radiotherapy. It was proposed that the radiation damage of various tumors is associated with the release of increased presentation of antigens and release of effector molecules (HMGB1) triggering molecular patterns associated with damage of tumor cells (DAMPs), activation of proteasomal pathways and major histocompatibility complex molecules (HLA) of the 1st class, NKG2D ligand expression, complement fixation, interferon 1st type production, and induction of cell immunogenic death [3-6]. Besides, it should be mentioned that irradiation affects vessel, tumor stoma and tumor stem cells. Tumor irradiation, as is pointed out, activates the antigen-presenting cells and cytotoxic T-lymphocytes that can finally induce tumor cell death both in the irradiation area and in unirradiated sites.
Unfortunately, until now the immunologic changes occurring during and after the SABR have not been sufficiently studied in clinical practice. In the presented prospective observation study, we have performed a complex analysis of the dynamics of immune status before and after SABR of various (in number and localization) metastatic lesions.

Materials and Methods

The dynamics of immune status in peripheral blood was prospectively evaluated in 46 patients (30 females and 16 males; mean age, 55.9 years; 95% CI, 52.4–59.4) with lung or liver metastases treated by SABR from September 2018 to July 2021.
This study was prospective observation single center study, and it was approved by the local ethics committee of N.N. Petrov National Cancer Center (Protocol No. 13, dated November 29, 2018). Voluntary informed consent for stereotactic radiotherapy and immunological studies was obtained in writing form from all patients.
Inclusion criteria were as follows: morphologically confirmed disease; oligometastatic disease (less than four lesions); the first SABR session; anatomical localization of metastases in liver or lungs, allowing for a radical SABR course; the size of liver or lung metastasis is less than 5 cm; no treatment by immune checkpoints inhibitors or/and chemotherapy at least 2 months before and after SABR.
Exclusion criteria were radiotherapy 6 or less months before SABR; immunotherapy before or during the treatment; age <18 years; Karnofsky performance status <70%; chemotherapy 2 months before or during SABR.
The immune status of patients was evaluated before irradiation, 3–4 and 6–8 weeks after the end of SABR. Distribution of the patients according to the localization of the primary lesion and irradiated metastatic sites is presented in Table 1.
SABR was performed according to the standard volumetric modulated arc therapy technique with respiratory gating and obligatory cone-beam CT guidance of every session. Lung metastases were irradiated as 3 fractions of 20 Gy (peripheral metastases) or 4 fractions of 13.5 Gy (lesions with proximity to ribs or central “fly zone”). Liver metastases were irradiated in 3 fractions of 15–20 Gy. The number of treated lesions varied from 1 (32 patients) to 2–3 (14 patients). The dynamics of immune status was analyzed in patients with liver (29 cases) and lung (17 cases) metastases separately after SABR within the equivalent dose—EQD2Gy(α/β=10) of 93.75–105.75 Gy (group A, 23 patients) or EQD2Gy(α/β=10) of 150 Gy (group B, 23 patients). All included patients underwent SABR treatment for all oligometastatic lesions.
The assessment of immune status was made in three checkpoints: before the radiation (checkpoint 1), in 3–4 weeks (checkpoint 2), and in 6–8 weeks (checkpoint 3) after the end of SABR. The immune status was characterized by the count of several main peripheral blood lymphocyte subpopulations.
The calculations were performed with flow cytometry method. To process the data, we used BD FACSDiva software (version v8.0.1; BD Biosciences, San Jose, CA, USA). The subpopulation composition of peripheral blood immunocompetent cells was studied on a BD FACSCanto II flow cytometer. We evaluated the relative value of lymphocytes; T-lymphocytes (CD3+CD19); В-lymphocytes (CD3–CD19+); T-helpers (CD3+CD4+); activated T-helpers with different phenotypes (CD3+CD4+HLA-DR+ and CD4+CD25+); cytotoxic Т-lymphocytes (CD3+СD8+); activated cytotoxic Т-lymphocytes (CD3+СD8+HLA-DR+); NK-cells (CD3CD16+CD56+); NKТ-cells (CD3+CD16+CD56+); T-regulatory lymphocytes (CD4+CD25brightCD127low); double-positive T-lymphocytes (CD3+CD4+СD8+); double-negative T-lymphocytes (CD3+CD4СD8); and the immunoregulatory index (CD4+/CD8+).
The values of this immune status indicators (T-lymphocytes; В-lymphocytes; T-helpers; activated T-helpers with phenotype CD3+CD4+HLA-DR+; cytotoxic Т-lymphocytes; activated cytotoxic Т-lymphocytes; NK-cells; NKТ-cells; double-positive T-lymphocytes; double-negative T-lymphocytes and the immunoregulatory index) were calculated from the total count of lymphocytes. CD4+CD25+ activated T-helpers and T-regulatory lymphocytes were calculated as a percentage of the T-helper (CD3+CD4+) population.
For calculation of the absolute value of the immunocompetent cells, a two-platform analysis system was used; the calculation was made using the results of a hematological analyzer.
We used antibodies for a multicolor panel (BD FACSCanto2 HIV reagents kits, 6-color TBNK reagent) mouse anti-human HLA-DR monoclonal antibodies labeled with V450, CD25 antibodies labeled with FITC (BD Biosciences), CD4 monoclonal antibodies labeled with BD PerCP-Cy5.5 fluorochrome and CD127 antibodies labeled with Pe-Cy7.
The statistical analysis of the data was performed in R version 3.6.2 (https://www.r-project.org/). Friedman test was used to determine statistically significant differences in immune status dynamics [7]. Nemenyi’s criterion was used as a post hoc test for multiple pair-wise comparisons [8]. A p-value less than 0.05 was considered statistically significant. The relative value (percentage) of the immunocompetent cells was used for the statistical analysis.

Results

The analysis of immune status parameters in patients with extensive irradiated solid tumors revealed a statistically significant increase of T-lymphocytes (CD3+CD19) in 3–4 weeks after the irradiation compared to the analysis performed before the stereotactic radiotherapy (χ2 Friedman = 13.781; omnibus p = 0.001; pairwise p = 0.001). Besides, a statistically significant increase of T-helper subpopulation (CD3+CD4+) (χ2 Friedman = 11.01; omnibus p = 0.004; pairwise p = 0.003) was noted in 3–4 weeks after the irradiation compared to the analysis performed before the radiotherapy and to non-significant decrease by the period of 6–8 weeks after the irradiation.
A statistically significant increase of activated cytotoxic T-lymphocytes carrying the HLA-DR+ antigen was noted in 3–4 weeks after SABR (pairwise p = 0.001) with a continuing rise of this subpopulation in 6–8 weeks after the irradiation (pairwise p = 0.004) (χ2 Friedman = 15.4; omnibus p = 0.001) (Fig. 1A). A detailed calculation is presented in Supplementary Table S1).
An increase of activated T-helpers was noted 3–4 and 6–8 weeks after the SABR compared to the values obtained before the irradiation (χ2 Friedman = 38.8; omnibus p < 0.001; pairwise p = 0.001 and 0.001, respectively) (Fig. 1B). A detailed calculation is presented in Supplementary Table S1.
A decrease of T-regulated immune suppressive lymphocytes (CD4+CD25brightCD127low) was recorded in 3–4 and 6–8 weeks after the SABR (χ2 Friedman = 12.96; omnibus p = 0.002, pairwise p = 0.004 and 0.010, respectively) compared to the values obtained before the irradiation (Fig. 1C). A detailed calculation is presented in Supplementary Table S1.
Besides, a decrease of B-lymphocytes (CD3CD19+) was recorded in 3–4 and 6–8 weeks after the SABR compared to the values recorded before the irradiation (χ2 Friedman = 35.4; omnibus p = 0.001; pairwise p = 0.001 and 0.002, respectively).
A decrease of NKT-cells (CD3+CD16+CD56+) was noted in 6–8 weeks after the irradiation compared to the analysis performed before the SABR (χ2 Friedman = 9.8; omnibus p = 0.007; pairwise p = 0.006).
Statistically significant differences in the dynamics of other immune cells in the peripheral blood (activated CD4+CD25+ T-helpers, NK-cells, double positive T-lymphocytes, double negative T-lymphocytes) were not revealed.
The comparative analysis demonstrates a significant correlation between changes in the peripheral blood immune cells and the EQD2Gy(α/β=10) delivered to the metastatic lesions. While examining T-lymphocytes (CD3+CD19) after the SABR, a statistically significant increase of T-lymphocytes was discovered in 3–4 weeks after the irradiation in both dose groups—group A: EQD2Gy(α/β=10) = 93.75–105.75 Gy (χ2 Friedman = 7.3; omnibus p = 0.030; pairwise p =0.020); group B: EQD2Gy(α/β=10) = 150 Gy (χ2 Friedman = 7.1; omnibus p = 0.030; pairwise p =0.030).
The dynamic pattern in activated cytotoxic T-lymphocytes (CD3+CD8+HLA-DR+) is quite interesting. When administering EQD2Gy(α/β=10) = 93.75–105.75 Gy, a statistically significant increase of cells in the given population was noted both in 3–4 and 6–8 weeks after the irradiation (χ2 Friedman = 16.1; omnibus p < 0.001; pairwise p = 0.001 and 0.003, respectively). At the same time, statistically significant differences in the number of activated cytotoxic T-lymphocytes after SABR with EQD2Gy(α/β=10) = 150 Gy were not recorded (Fig. 2). A detailed calculation is presented in Supplementary Table S2.
A statistically significant increase of T-helpers was recorded in 3–4 weeks after irradiating the lesions with EQD2Gy(α/β=10) = 93.75–105.75 Gy (χ2 Friedman = 6.96; omnibus p = 0.030; pairwise p = 0.030). There were no differences in cells of the given subpopulation in dose group В (EQD2Gy(α/β=10) = 150 Gy).
A statistically significant increase of activated T-helpers phenotype (CD3+CD4+HLA-DR+) was noted in 3–4 and 6–8 weeks after the SABR when compared to the values obtained before SABR. These differences were significant in both groups: in group A (EQD2Gy(α/β=10) = 93.75–105.75 Gy) (χ2 Friedman = 26.2; omnibus p < 0.001; pairwise p = 0.001 and 0.001, respectively) and in group В (EQD2Gy(α/β=10) = 150 Gy) (χ2 Friedman = 14.2; omnibus p < 0.001; pairwise p = 0.001 and 0.003, respectively). Concurrently, in group B we mentioned a tendency for decrease in the number of activated T-helpers 6–8 weeks after the SABR when the values were compared to the data obtained 3–4 weeks after irradiation.
Moreover, the analysis of another subpopulation of the activated T-helpers (CD4+CD25+) demonstrated a statistically significant increase of activated T-helpers in 6–8 weeks after the SABR within the EQD2Gy(α/β=10) = 93.75–105.75 Gy (group A) when compared to the values obtained before the SABR (χ2 Friedman = 6.3; omnibus p = 0.040; pairwise p = 0.030, respectively). In group B (EQD2Gy(α/β=10) = 150 Gy) we did not mention statistically significant differences in the number of activated T-helpers before and at various time points after SABR.
In the subgroup of T-regulatory (suppressors) lymphocytes we also found dose dependent effects. In particular, a significant reduction of T-regulatory lymphocytes (χ2 Friedman = 7.7; omnibus p = 0.022; pairwise p = 0.040) was mentioned 3–4 weeks after SABR to metastatic lesions with EQD2Gy(α/β=10) = 150 Gy. On the contrary, we did not reveal significant differences in the number of T-regulatory lymphocytes after SABR within EQD2Gy(α/β=10) = 93.75–105.75 Gy.
In 46 consecutive patients, we compared the dynamics of immune status according to the number of the irradiated lesions. Group I consisted of 32 patients with one irradiated metastatic lesion and group II (14 patients) with 2–3 irradiated metastases. After SABR of a single lesion we mentioned numerous changes in immune profile. First of all, a statistically significant increase of T-lymphocytes (CD3+CD19) was recorded in 3–4 weeks after the SABR (χ2 Friedman = 9.2; omnibus p = 0.010; pairwise p = 0.008). In addition, an increase in the number of T-helper quantity (χ2 Friedman = 9.1; omnibus p = 0.010; pairwise p = 0.008) was noted in the same timeframe (Fig. 3). On the contrary, we did not find any change in the number of the above-mentioned cells in the group with 2–3 irradiated lesions (group II). A detailed calculation is presented in Supplementary Table S3.
The increase of activated T-helpers (CD3+CD4+HLA-DR+) was noted in group I in 3–4 weeks and 6–8 after the irradiation compared to the values obtained before the SABR (χ2 Friedman = 33.2; omnibus p < 0.001; pairwise p = 0.001 and 0.001, respectively), and in group II, these changes were non-significant. However, an upward tendency was evident (χ2 Friedman = 6.9; omnibus p = 0.033; pairwise p = 0.060 and 0.060, respectively).
A statistically significant increase of the cytotoxic T-lymphocytes was revealed in 3–4 and 6–8 weeks after the SABR of a single lesion (χ2 Friedman = 11.6; omnibus p = 0.003; pairwise p = 0.005 and 0.020, respectively), and in group II, these changes were non-significant.
The analysis of T-regulatory lymphocytes in group with one irradiated lesion demonstrated a decrease of this subpopulation in 3–4 and 6–8 weeks after SABR compared to the values detected before the start of radiotherapy (χ2 Friedman = 13.2; omnibus p = 0.001; pairwise p = 0.005 and 0.006, respectively). T-regulatory lymphocytes demonstrated a statistically insignificant downward tendency in the group having 2–3 irradiated lesions.
We also evaluated changes of the immune status in connection with the localization (lung vs. liver) of irradiated metastases. An increase of activated T-helpers was observed in both groups 3–4 and 6–8 weeks after SABR when compared to the values obtained before the irradiation: in group with SABR for hepatic lesions (χ2 Friedman = 29.2; p = 0.001; pairwise p = 0.001 and 0.001, respectively) and for lung lesions (χ2 Friedman = 8.9; omnibus p = 0.012; pairwise p = 0.020 and 0.040, respectively).
Significant changes in the number of T-lymphocytes (χ2 Friedman = 12.8; omnibus p = 0.002; pairwise p = 0.002 and 0.020, respectively) and T-helpers (χ2 Friedman = 10.6; omnibus p = 0.003; pairwise p = 0.006 and 0.051, respectively) were observed after SABR for hepatic metastases: both populations increased in 3–4 weeks after the irradiation followed by true decrease of T-lymphocytes in 6–8 weeks after the SABR. Statistically significant differences of the above parameters after SABR for lung metastases were not revealed (Fig. 4). A detailed calculation is presented in Supplementary Table S4.
A statistically significant increase of activated cytotoxic T-lymphocytes carrying antigen HLA-DR+ was registered in 3–4 weeks (pairwise p = 0.004) and in 6–8 weeks (pairwise p = 0.010) (χ2 Friedman = 12.2; omnibus p = 0.001) after the SABR for liver metastases. Similar differences were not revealed after irradiation of lung lesions.
Moreover, a statistically significant increase of the immunoregulatory index was noted 3–4 weeks after SABR for liver metastases (pairwise p = 0.040) followed by a decrease in 6–8 weeks after the irradiation (χ2 Friedman = 10.8; omnibus p = 0.004; pairwise p = 0.010).
On the contrary, a statistically significant decrease of T-regulatory lymphocytes (χ2 Friedman = 15.6; omnibus p < 0.001) was registered only after SABR for lung metastases: 3–4 weeks (pairwise p = 0.001) and 6–8 weeks (pairwise p = 0.006) after irradiation.

Discussion and Conclusion

Different populations of immune cells characterizing SABR induced changes of the immune status were dynamically evaluated during this study. These parameters reflect differences in both T-cell and humoral component of the immune system. The obtained results identify the activation of T-cell immune response mainly by the increase of T-lymphocytes (CD3+CD19); T-helpers (CD3+CD4+); activated T-helpers (CD3+CD4+HLA-DR+) and activated cytotoxic T-lymphocytes (CD3+СD8+HLA-DR+) against the decrease in activity of the antibody producer cells (CD3CD19+).
Chua et al. [9] investigated the immune responses to radiotherapy in 10 consecutive patients using in-depth mass cytometric profiling of circulating lymphocytes. It is interesting that the authors demonstrated significant heterogeneity of the immune status among patients before the initiation of radiotherapy and in seven non-irradiated people of the control group. Nevertheless, they demonstrated immune stimulatory effect of radiotherapy that was characterized by significant increase in the frequency of CD8 positive T cells from baseline.
Other authors [10,11] also point out the significance of tumor infiltration by activated CD8+ T-cell. In 2018, Zheng et al. [12] presented data confirming that a higher level of cytotoxic CD8+ T-cells in peripheral blood after the irradiation predicts a better survival without progression in patients with stage I non-small-cell lung cancer treated with SABR. They concluded that the enhancement of the tumor antigen specific cell immunity by combining radiotherapy and immunotherapy might become a decisive strategy in improving survival of these patients. Other authors [13,14] underline the importance of activated T-helpers and the role of T-regulatory cells in realization of antitumor immune response.
Considering the above-mentioned issues, a special focus in interpretation of our research must be concentrated on the post-SABR activation of T-cell component: T-lymphocytes (CD3+CD19), T-helpers (CD3+CD4+), activated T-helpers (CD3+CD4+HLA-DR+), and activated cytotoxic T-lymphocytes (CD3+СD8+HLA-DR+) with parallel reduction in the number of T-regulatory lymphocytes. These changes indicated that the state of T-cell inflammatory phenotype or “hot” tumor is likely to be reached after SABR. As is proved by Turajlic et al. [15], this condition is associated with a good response to immunotherapy by checkpoint inhibitors.
Most of the discussions about the possible additive or supra-additive effect of immune check point inhibitors with irradiation underline an important role of proper sequencing of these modalities. In particular, Buchwald et al. [16] demonstrated that a concurrent administration of both modalities was characterized by the effective immune response. On the other hand, in the PEMBRO-RT randomized trial therapy by check point inhibitors started 1 week after radiotherapy and was associated with the improvement in local control and disease-free survival [17]. Similar results were obtained in multicenter prospective randomized PACIFIC demonstrating that the start of therapy by PD-L1 inhibitors must be initiated during the first 2 weeks after the end of radiotherapy [18]. Results of our study indicated that the highest activation of T-cell immune response was recorded 3–4 weeks after the SABR and probably this timing can be optimal for combining radiotherapy with other immunotherapeutic interventions as, for instance, treatment with immune checkpoint inhibitors (PD-1/PD-L1; СTLA-4).
The effect of radiotherapy technique and the value of absorbed dose on the antitumor immune response intensity is still a matter for debates. Some authors reported that irradiation with large (>10 Gy) dose per fraction and high absorbed radiation dose causes massive release of tumor antigens and increased frequency of abscopal effects [19,20]. Many other authors found out that the optimal immunomodulation effects of radiotherapy can be obtained with the help of less aggressive regimes, most often as 3 fractions of 8–10 Gy [16,19,21]. It was proposed that these SABR schemes are characterized by the optimal balance between accumulation of neoantigens and activation of immunosuppressive exonuclease Trex1 [21]. Our results suggest that SABR for hepatic and lung metastases with EQD2Gy(α/β=10) = 93.75–105.75 Gy was associated with a more active immune response through the increase of T-lymphocytes (CD3+CD19); activated T-helpers (CD3+CD4+HLA-DR+ and CD4+CD25+); activated cytotoxic T-lymphocytes (CD3+СD8+HLA-DR+). On the contrary, SABR with EQD2Gy(α/β=10) =150 Gy shows only a statistically significant decrease of T-regulatory lymphocytes.
Many authors [4,22] indicate the importance of SABR to all metastatic sites in order to decrease tumor burden, which is associated with an improved response, irradiates all manifested sites that are immunologically heterogeneous because this comprehensive radiotherapy program can maximize the probability of inducing compelling local immune effects [22]. Surprisingly, our data indicate that in patients with 2–3 irradiated metastatic lesions activation of immune cells in peripheral blood was significantly lower. We also demonstrated that the number of irradiated lesions influences the dynamic of immune response with early activation of T-cell subpopulations after SABR for one lesion and later activation of T-cells after SABR for 2–3 metastatic lesions.
These conflicting results can be partly explained by the fact that in preclinical and clinical studies, response to immunotherapy has been shown to be strongly associated with tumor burden, with higher response in lower burden [23,24]. However, the restricted number of analyzed patients and an observational design of the study makes the conclusion about the association between SABR-induced immunologic changes and the number of irradiated metastases to be very preliminary and speculative.
Currently, there is no clear understanding of what kinds of immune system changes occur in metastatic patients after SABR administered to lesions of various localizations. Tang et al. [25] reported that in patients with non-small-cell lung cancer the antitumor immunologic response is more active after the SABR for hepatic metastases than after irradiation of lung lesions. Our data obtained in mixed population of oncologic patients indicated the same trend: a more pronounced activation of T-cell immune component was mentioned in patients with SABR for liver lesions compared to the group with irradiated lung metastases. Probably this phenomenon can be explained by significant differences in immunogenic potential of lung and liver tissues with high concentration of macrophages and other lymphatic elements in liver. Additionally, it is well-known that macrophages play an important role in the functioning of immune system: first, as the cells that presented tumor antigens to CD8+ T-cells; second, as the activated macrophages that damage neighboring cells by various signaling factors [26,27].
Our study has a number of meaningful limitations. First, it is a relatively small number of evaluated patients that might have underpowered some changes in the immune landscape (as in irradiating lung lesions or in SABR to multiple lesions). Moreover, the obtained data might have been critically impacted by the group heterogeny with regard to the localization and histological variant of the primary tumors. In order to overcome these limitations we tried to perform multivariate analysis, but this has not been successful due to a very small sample size of the subgroups.
However, it seems possible to make several preliminary conclusions: (1) SABR is associated with the activation of T-cell component of the immune system (p ≤ 0.01) which is manifested by the increase of T-lymphocytes (CD3+CD19), T-helpers (CD3+CD4+), activated T-helpers (CD3+CD4+HLA-DR+), and activated cytotoxic T-lymphocytes (CD3+СD8+HLA-DR+) affected by the decrease of regulatory T-lymphocytes (CD4+CD25brightCD127low) and antibody producer cells (CD3CD19+). The activation is most evident in 3–4 weeks after the SABR. (2) Lower doses of SABR (EQD2Gy(α/β=10) = 93.7–105.7 Gy) are associated with the most significant activation of T-cell immune response (p ≤ 0.04) than SABR with higher total dose (EQD2Gy(α/β=10) = 150 Gy). (3) SABR for a single metastatic lesion induces a more pronounced activation of T-cell immune response than irradiation of two or three lesions (p ≤ 0.03). (4) SABR for a hepatic lesion result in a more pronounced activation of T-cell immune response than SABR for lung metastases (p ≤ 0.04).
Immunogenic effects of radiation therapy are complex, controversial and not perfectly understood. It becomes clear that many factors (localization and number of irradiated lesions, the dose and fractionation scheme, irradiated volume, etc.) can significantly influence the immunomodulating activity of radiation therapy, and further research in this field is of paramount clinical importance.

Notes

Statement of Ethics

This study was approved by the local ethics committee of N.N. Petrov National Cancer Center (Protocol No. 13 dated November 29, 2018). Voluntary informed consent for stereotactic radiotherapy and immunological studies was obtained in writing form from all patients.

Conflict of Interest

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

Funding

None.

Author Contributions

Conceptualization, Novikov SN, Aleksandrovna BI, Vasilevich KS. Formal analysis, Novikov SN, Aleksandrovna BI, Yurievich ZA. Data curation, Viktorovich GD, Viktorovich GD, Ivanovna TE, Evgenevich AF, Ivanovich AA, Markovich GM. Investigation and methodology, Viktorovna EN. Writing–original draft, Novikov SN, Yurievich ZA. Writing–review and editing, Novikov SN, Aleksandrovna BI, Yurievich ZA, Viktorovna EN, Viktorovich GD, Alexandrovna FE, Ivanovna TE, Evgenevich AF, Ivanovich AA, Markovich GM, Vasilevich KS.

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Supplementary Materials

Supplementary materials can be found via https://doi.org/10.3857/roj.2022.00521.
Table S1.
Statistical data of immune status indicators in the studied points against the background of SABR
roj-2022-00521-suppl1.pdf
Table S2.
Statistical data of immune status indicators in the studied points against the background of SABR depending on delivered EQD2Gy(ɑ/β=10)
roj-2022-00521-suppl2.pdf
Table S3.
Statistical data of immune status indicators in the studied points against the background of SABR depending to the number of the irradiated lesions
roj-2022-00521-suppl3.pdf
Table S4.
Statistical data of immune status indicators in the studied points against the background of SABR depending to localization of irradiated metastases
roj-2022-00521-suppl4.pdf

Fig. 1.
Dynamics in the number of (A) activated cytotoxic lymphocytes, (B) activated T-helpers, and (C) T-regulatory lymphocytes. SABR, stereotactic ablative radiotherapy.
roj-2022-00521f1.jpg
Fig. 2.
Dynamics in the number of (A) activated cytotoxic lymphocytes, (B) activated T-helpers, and (C) T-regulatory lymphocytes depends on delivered EQD₂Gy (ɑ/β = 10). SABR, stereotactic ablative radiotherapy; EQD, equivalent dose.
roj-2022-00521f2.jpg
Fig. 3.
Dynamics in the number of (A) T-helpers, (B) activated T-helpers, and (C) T-regulatory lymphocytes according to the number of the irradiated lesions. SABR, stereotactic ablative radiotherapy.
roj-2022-00521f3.jpg
Fig. 4.
Dynamics in the number of (A) T-helpers, (B) activated cytotoxic lymphocytes, and (C) T-regulatory lymphocytes according to localization of irradiated metastases. SABR, stereotactic ablative radiotherapy.
roj-2022-00521f4.jpg
Table 1.
Patient distribution according to the primary tumor and localization of the irradiated metastases
Primary n Localization of the irradiated metastases
Liver Lung
Colorectal adenocarcinoma 20 17 3
Breast cancer
 Luminal B subtype 5 4 1
 HER2-enriched subtype 1 0 1
 Triple-negative subtype 1 1 0
Non-small cell lung cancer
 Adenocarcinoma 1 0 1
 Squamous cell carcinoma 2 1 1
 Adenosquamous carcinoma 1 1 0
 Poorly differentiated 1 0 1
Soft tissue sarcoma
 Leiomyosarcoma 4 2 2
 Extraskeletal myxoid chondrosarcoma 1 0 1
Parotid cancer
 Adenoid cystic carcinoma 1 0 1
 Clear cell carcinoma 1 0 1
 Low grade adenocarcinoma 1 0 1
Epithelioid malignant melanoma 2 2 0
Esophageal squamous cell carcinoma 1 1 0
Prostate acinar adenocarcinoma 1 0 1
Hepatocellular carcinoma 1 0 1
Clear cell renal cell carcinoma 1 0 1
Total 46 29 17

HER2, human epidermal growth factor receptor 2.

References

1. Stubblefield MD. Cancer rehabilitation: principles and practice. New York, NY: Demos Medical Publishing; 2009.

2. Amaravadi RK, Thompson CB. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin Cancer Res 2007;13:7271–9.
crossref pmid pdf
3. Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC. Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death. Oncoimmunology 2014;3:e28518.
crossref pmid pmc
4. Hanna GG, Illidge T. Radiotherapy and immunotherapy combinations in non-small cell lung cancer: a promising future? Clin Oncol (R Coll Radiol) 2016;28:726–31.
crossref pmid
5. Matsumura S, Demaria S. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat Res 2010;173:418–25.
crossref pmid pmc
6. Kwilas AR, Donahue RN, Bernstein MB, Hodge JW. In the field: exploiting the untapped potential of immunogenic modulation by radiation in combination with immunotherapy for the treatment of cancer. Front Oncol 2012;2:104.
crossref pmid pmc
7. Friedman M. A comparison of alternative tests of significance for the problem of m rankings. Ann Math Stat 1940;11:86–92.
crossref
8. Nemenyi PB. Distribution-free multiple comparisons [dissertation] Ann Arbor, MI: Princeton University; 1963.

9. Chua KL, Fehlings M, Yeo EL, et al. High-dimensional characterization of the systemic immune landscape informs on synergism between radiation therapy and immune checkpoint blockade. Int J Radiat Oncol Biol Phys 2020;108:70–80.
crossref pmid
10. Gupta A, Probst HC, Vuong V, et al. Radiotherapy promotes tumor-specific effector CD8+ T cells via dendritic cell activation. J Immunol 2012;189:558–66.
crossref pmid pdf
11. Lee Y, Auh SL, Wang Y, et al. Therapeutic effects of ablative radiation on local tumor require CD8+ T cells: changing strategies for cancer treatment. Blood 2009;114:589–95.
crossref pmid pmc pdf
12. Zheng Y, Shi A, Wang W, et al. Posttreatment immune parameters predict cancer control and pneumonitis in stage I non-small-cell lung cancer patients treated with stereotactic ablative radiotherapy. Clin Lung Cancer 2018;19:e399.
crossref pmid
13. Martinez-Zubiaurre I, Chalmers AJ, Hellevik T. Radiation-induced transformation of immunoregulatory networks in the tumor stroma. Front Immunol 2018;9:1679.
crossref pmid pmc
14. Schaue D, Ratikan JA, Iwamoto KS, McBride WH. Maximizing tumor immunity with fractionated radiation. Int J Radiat Oncol Biol Phys 2012;83:1306–10.
crossref pmid
15. Turajlic S, Litchfield K, Xu H, et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: a pan-cancer analysis. Lancet Oncol 2017;18:1009–21.
crossref pmid
16. Buchwald ZS, Wynne J, Nasti TH, et al. Radiation, immune checkpoint blockade and the abscopal effect: a critical review on timing, dose and fractionation. Front Oncol 2018;8:612.
crossref pmid pmc
17. Theelen WS, Peulen HM, Lalezari F, et al. Effect of pembrolizumab after stereotactic body radiotherapy vs pembrolizumab alone on tumor response in patients with advanced non-small cell lung cancer: results of the PEMBRO-RT phase 2 randomized clinical trial. JAMA Oncol 2019;5:1276–82.
crossref pmid pmc
18. Breen WG, Leventakos K, Dong H, Merrell KW. Radiation and immunotherapy: emerging mechanisms of synergy. J Thorac Dis 2020;12:7011–23.
crossref pmid pmc
19. Marconi R, Strolin S, Bossi G, Strigari L. A meta-analysis of the abscopal effect in preclinical models: Is the biologically effective dose a relevant physical trigger? PLoS One 2017;12:e0171559.
crossref pmid pmc
20. Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S, Formenti SC. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer 2018;18:313–22.
crossref pmid pmc pdf
21. 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
22. Bauml JM, Mick R, Ciunci C, et al. Pembrolizumab after completion of locally ablative therapy for oligometastatic non-small cell lung cancer: a phase 2 trial. JAMA Oncol 2019;5:1283–90.
crossref pmid pmc
23. Huang AC, Postow MA, Orlowski RJ, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545:60–5.
crossref pmid pmc
24. Joseph RW, Elassaiss-Schaap J, Kefford R, et al. Baseline tumor size is an independent prognostic factor for overall survival in patients with melanoma treated with pembrolizumab. Clin Cancer Res 2018;24:4960–7.
crossref pmid pmc pdf
25. Tang C, Welsh JW, de Groot P, et al. Ipilimumab with stereotactic ablative radiation therapy: phase I results and immunologic correlates from peripheral T cells. Clin Cancer Res 2017;23:1388–96.
crossref pmid pdf
26. Manda K, Glasow A, Paape D, Hildebrandt G. Effects of ionizing radiation on the immune system with special emphasis on the interaction of dendritic and T cells. Front Oncol 2012;2:102.
crossref pmid pmc
27. Obeid M, Tesniere A, Ghiringhelli F, et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat Med 2007;13:54–61.
crossref pmid pdf
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