1,655
Views
5
CrossRef citations to date
0
Altmetric
Original Research

Stereotactic Ablative Radiation Therapy in 3 Fractions Induces a Favorable Systemic Immune Cell Profiling in Prostate Cancer Patients

, , , , , , , , , & show all
Article: 2174721 | Received 19 Dec 2022, Accepted 26 Jan 2023, Published online: 13 Feb 2023

ABSTRACT

The impact of radiotherapy (RT) on immune cell status in prostate cancer (PCa) is only partially determined. The aim of this study was to assess the effect of different RT strategies on peripheral B, T, and Natural killer (NK) lymphocytes at precise longitudinal time-points in PCa. 18 patients treated with stereotactic body radiation therapy (SBRT) (40 Gy/3FRX), definitive moderate-hypofractionation (62 Gy/20FRX), or post-operative conventional-fractionation RT (66–69 Gy/30FRX) were prospectively evaluated for the immune cell profile in terms of immune cell composition, differentiation stage, cytokine production and inhibitory receptor (IR) expression. The immune-monitoring of the 18 patients revealed that RT affects the balance of systemic immune cells, with the main differences observed between SBRT and conventionally fractionated RT. SBRT favorably impacts immune response in term of increased B cells, central-memory and effector-memory CD8+ T cells, along with decreased Treg cells after treatment. On the contrary, conventional fractionated RT had a long-term negative effect on the systemic immune profile, including a decrease of total lymphocyte counts accompanied by an increase of neutrophils-to-lymphocytes ratio. Total B and T cells decreased and Treg-to-CD8+ ratio increased. Functionality of T lymphocytes were not affected by any of the 3-fractionation schedules. Interestingly, SBRT significantly up-regulates the expression of V-domain immunoglobulin suppressor of T-cell activation (VISTA) in CD8+ T cells in the absence of other IRs. Our results indicate the relevance of systematic immunomonitoring during RT to identify novel immune-related target to design trials of combined radio-immunotherapy as a promising strategy in the clinical management of PCa.

Introduction

Prostate cancer (PCa) is the second most common cancer and the fifth cause of cancer related deaths overall.Citation1 Radiotherapy (RT) plays a pivotal role in the treatment of localized disease, either as adjuvant-salvage therapy after surgery or as curative treatment option alone or combined with androgen deprivation (AD).

Different fractionation schedules and volumes of treatment can be delivered depending on disease stage. Stereotactic body radiation therapy (SBRT), defined as a dose per fraction higher than 5 Gy,Citation2 is an accepted treatment modality for localized PCa.Citation3 Controlled trials have shown favorable clinical results in terms of local control and gastro-intestinal and genitourinary toxicity rates,Citation4–6 compared to conventionally fractionated RT.Citation7

Moreover, several studies have found that peripheral immune cells might be significantly modified by conventionally fractionated RTCitation8 though the clinical role of this immune cell modification is yet to be determined. Moreover, the effect on peripheral immune cells of a higher dose per fraction in SBRT treated patients is still poorly investigated.

To enhance SBRT efficacy, studies are warranted to investigate the effect of this high dose per fraction on patients’ immune cells, by collecting and analyzing longitudinal blood samples. An in-depth immunomonitoring may in fact highlight “the fine tuning” of immune response during RT and may allow to define complementary strategies of treatment.

Immunotherapy and in particular immune checkpoint inhibitors (ICI) have revolutionized the systemic treatment of cancer, though the survival benefit in PCa patients remains elusive. To date, ICI in PCa have been only tested in advanced castration resistant metastatic state and failed to provide clinical benefit when used as monotherapy.Citation9 The mechanism of resistance, mainly related to the tumor microenvironment (TME) characteristicsCitation10,Citation11 is still under investigation, even though the combination of Nivolumab and Ipilimumab has improved clinical outcomes at the cost of higher toxicity.Citation12

The aim of this prospective study is to evaluate the immunological changes on peripheral T, B and Natural killer (NK) lymphocytes at precise time-points in PCa patients treated with 3 different treatment strategies, in order to identify new therapeutic targets to be combined with radiotherapy.

Materials and methods

Patients and Treatments

This prospective study was conducted at a single Institution between February 2019 and May 2021 (Clinicaltrials.gov NCT04774133). The study was approved by the Institutional Review Board (RS1163/18). Eligibility criteria were: histologically confirmed PCa undergoing RT, age >18 years. Exclusion criteria were: previous malignancies, chemotherapy, autoimmune and hematological disease in leukemic phase. All accrued patients were treated according to our Institutional guidelines/ongoing research protocol: SBRT (40 Gy/3 fractions (FRX); Group-1); definitive moderately hypofractionationated RT (62 Gy/20FRX; Group-2); post-operative conventionally fractionated RT (66–69 Gy/30FRX; Group-3). Target volumes were: prostate only (Group-1), prostate + seminal vesicles (SV) (Group-2), prostate bed + pelvic lymph nodes (Group-3), including bilateral internal, external and common iliac lymph nodes as well as pre-sacral lymph nodes. Androgen deprivation (AD) was prescribed to selected patients. RT was delivered as follow: SBRT, every other day; definitive moderately hypofractionationated RT, four days per week; conventionally fractionated RT, five days per week.

Peripheral blood collection

30 ml of blood were collected at different timepoints () from 18 treated patients and 12 sex/age-matched healthy donors (HD), after written informed consent, in EDTA vacutainer tubes (BD Bioscience, 367864). 10 ml of whole blood EDTA was analyzed for complete blood count (CBC) (DXH 900, Beckman Coulter) and flow cytometry staining. Peripheral blood mononuclear cells (PBMC) were isolated from 20 ml of blood using Ficoll-paque separation (Euroclone, DVCL5020) and cryopreserved in 10% Dimethyl sulfoxide (DMSO) (Sigma-Aldrich, D2650) until use.

Figure 1. Flow chart of the study and CBC analysis. (a) Treatment schedule and blood sample collection. (b-f) Longitudinal analysis of immune cells in 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7), determined by full blood counts (Wilcoxon U-test). Middle panels, comparison among Groups (Mann-Whitney U-test). Right panels, normalization calculated as intra-individual patient ratio between 12 months (POST) and baseline level (POST/PRE ratio). The red line indicates a ratio of 1, representing no change between PRE and POST treatment. The P value is calculated by Wilcoxon U-test between POST versus baseline and follow-up versus treatment-end. *p ≤ .05. Abbreviation: FRX = fraction; WBC = white blood cells; MP = Middle point; mo = months.

Figure 1. Flow chart of the study and CBC analysis. (a) Treatment schedule and blood sample collection. (b-f) Longitudinal analysis of immune cells in 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7), determined by full blood counts (Wilcoxon U-test). Middle panels, comparison among Groups (Mann-Whitney U-test). Right panels, normalization calculated as intra-individual patient ratio between 12 months (POST) and baseline level (POST/PRE ratio). The red line indicates a ratio of 1, representing no change between PRE and POST treatment. The P value is calculated by Wilcoxon U-test between POST versus baseline and follow-up versus treatment-end. *p ≤ .05. Abbreviation: FRX = fraction; WBC = white blood cells; MP = Middle point; mo = months.

Flow cytometry

200 µl of fresh blood was stained with a combination of antibodies (mAbs) (Supplementary Table S1) and acquired on Lyse-Wash-Assistant (LWA) of Becton Dickinson (BD Pharm Lyse, 555899). Surface-staining of isolated PBMC was performed for 30 min at 4°C using combinations of mAbs (Supplementary Table S1). 3 × 105 cells were activated with plate-bound anti-CD3 mAb (2 μg/ml) (CBT3 IgG2a), in the presence of Golgistop (BD Bioscience 554724) and Golgiplug (BD Bioscience 555029), for 5–6 h. Intracellular staining was performed by the use of Intrasure kit (BD Bioscience, 641778) according with the manufactory instruction, combining surface and intracellular mAbs (Supplementary Table S1). Dead cells were excluded by Fixable Viability Stain 700 (BD Bioscience 564997). Cells were acquired in the BD FACSCelesta and BD FACSCantoII flow cytometers and analyzed by BD FACSDiva and Flowjo software.

Statistical analysis

For the comparison of two or more groups non-parametric Mann-Whitney U-test and Kruskal-Wallis test were used. For intra-individual comparison among different times the Wilcoxon U-test and Friedman test were used. Box-graphs represent the 25th and 75th percentiles, median and outliers are shown. Statistical analysis was performed by comparing different timepoints versus baseline and follow-up (6 or 12 months) versus treatment-end. A P value ≤.05 was considered significant. Significance is denoted as *p ≤ .05, **p ≤ .01, ***p ≤ .001. p value supporting a trend toward statistical significance is shown as values. Statistical evaluation was performed with SPSS 21.0 (SPSS Inc., Chicago, IL, USA) for Windows. Barplot and boxplot were generated using R environment.

Results

Effect of different RT strategies on complete blood counts

Eighteen patients were included in this analysis (Group-1 3FRX, 5 pts; Group-2 20FRX, 6 pts; Group-3 30FRX, 7 pts). Patients, treatment characteristics and disease status after a median follow-up of 38.4 months (range 33.7–44.1 months) are reported in . Six patients were additionally treated with AD (Group-2, 4 pts; Group-3, 2 pts) for 6 to 24 months according to disease stage.

Table 1. Clinical characteristics of patients.

To identify which immune cell subsets might be modulated by the 3 RT strategies, including different fractionation and treatment volumes (Group-1, 40 Gy/3FRX, prostate; Group-2, 62 Gy/20FRX, prostate + SV; Group-3, 66–69 Gy/30FRX, prostate bed + pelvic lymph nodes), we performed an in-depth immune profiling at different time-points, before, during, at the end and post-RT in peripheral blood ().

shows the results of CBC analysis from baseline to 12 months after RT. At baseline, immune cell counts did not show significant differences among Groups except for a trend in highest monocytes count in Group-1 (, middle panels). White blood cell count (WBC) increased in Group-1 during RT, and returned to baseline at 6 and 12 months (, left panel). This was also evident after the normalization of counts calculated as intra-individual ratio between 12 months and baseline (, right panel). Differently, in Group 2 and 3 WBC significantly decreased at treatment-end, but did not return at baseline after 1 year (). Absolute lymphocyte counts underwent a slight reduction during RT in Group-1, recovering to baseline 6 months after RT (). In both Group-2 and Group-3 a significant drop was observed at treatment-end. After 1 year of follow-up (POST 12 months) we evidenced a recovery in Group-2 and only a partial recovery not reaching baseline value in Group-3 ().

Monocyte’s analysis did not show significant changes during RT (). Neutrophil counts increased in Group-1 from 3 h and returned to baseline after 6 months (), as evidenced in neutrophil-to-lymphocyte ratio (). A significant increase was observed as well at 3 h and 6 months in Group-3 (), with a higher neutrophil-to-lymphocyte ratio at treatment-end, which was not fully restored after 1 year ().

Comparison of CBC after 1 year among the 3 groups clearly showed that lymphocyte count was higher in Group-1 than in Groups-2 and 3 (, middle panel). The comparison of CBC between the 18 patients (at baseline) and 12 sex/age-matched HD did not evidence statistically significant differences (Supplementary Figure S1).

Effect of RT strategies on immune cell frequency and functionality

Immunophenotyping of peripheral immune cells was performed by multicolor flow cytometry (gating strategy Supplementary Figure S2) and results are shown in . The frequency of T and B lymphocytes (CD3, CD4, CD19) were detected in whole fresh blood and isolated PBMCs, with comparable results (Supplementary Figure S3).

Figure 2. Impact of RT-strategies on immune cell frequency. (a, g) Dot-plots representing immune cell population, evaluated by multicolor flow cytometry, in two patients, at PRE and POST (6mo). (b-f, h-m) Longitudinal analysis of 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7), showing the frequency of total T cells (CD3+CD45+ on total lymphocytes) (b), CD8+ T cells (on CD3+CD45+ T cells) (c), CD4+ T cells (on CD3+CD45+ T cells) (d), CD8-to-CD4 ratio (e), ɣδ T cells (CD3+CD4CD8 on CD3+CD45+ T cells) (f), Treg cells (CD4+CD25highCD127 on CD3+CD45+CD4+ T cells) (h), Treg-to-CD8+ ratio (i), NK cells (CD3CD56+ on CD45+ lymphocytes) (l) and B cells (CD3CD19+ on CD45+ lymphocytes) (m) (Wilcoxon U-test). Middle panels, comparison among Groups (Mann-Whitney U-test). Right panels, normalization calculated as intra-individual patient ratio between 12 months (POST) and baseline level (POST/PRE ratio). The red line indicates a ratio of 1, representing no change between PRE and POST treatment. The P value is calculated by Wilcoxon U-test between POST versus baseline and follow-up versus treatment-end. *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point; POST = 6 months after RT.

Figure 2. Impact of RT-strategies on immune cell frequency. (a, g) Dot-plots representing immune cell population, evaluated by multicolor flow cytometry, in two patients, at PRE and POST (6mo). (b-f, h-m) Longitudinal analysis of 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7), showing the frequency of total T cells (CD3+CD45+ on total lymphocytes) (b), CD8+ T cells (on CD3+CD45+ T cells) (c), CD4+ T cells (on CD3+CD45+ T cells) (d), CD8-to-CD4 ratio (e), ɣδ T cells (CD3+CD4−CD8− on CD3+CD45+ T cells) (f), Treg cells (CD4+CD25highCD127− on CD3+CD45+CD4+ T cells) (h), Treg-to-CD8+ ratio (i), NK cells (CD3−CD56+ on CD45+ lymphocytes) (l) and B cells (CD3−CD19+ on CD45+ lymphocytes) (m) (Wilcoxon U-test). Middle panels, comparison among Groups (Mann-Whitney U-test). Right panels, normalization calculated as intra-individual patient ratio between 12 months (POST) and baseline level (POST/PRE ratio). The red line indicates a ratio of 1, representing no change between PRE and POST treatment. The P value is calculated by Wilcoxon U-test between POST versus baseline and follow-up versus treatment-end. *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point; POST = 6 months after RT.

Immune cells frequency did not show significant differences among Groups at baseline (, middle panels), similarly to CBC, with the exception of γδ-T cells higher in Group-3.

Figure 2. (Continued)

Figure 2. (Continued)

A significant increase of total T cells (CD3+CD45+) and conventional CD4+ T cells was observed in all Groups () at 3 h. At treatment-end, both subsets returned to baseline but Group-3 showed a significant decrease after 6 months compared to baseline and treatment-end values (). Conversely, within CD8+ T cells a slight decrease was observed in Group-1 at 3 h and treatment-end, showing a significant recovery after 6 months compared to baseline. Groups-2 and 3 showed a significant increase of CD8+ cells after 6 months, accompanied by an increment of CD8-to-CD4 cell ratio in all groups at 6 months ().

Looking at Treg subset, defined as CD3+CD4+CD25 highCD127, a progressive increase during RT was noticed with highest value at treatment-end, although not reaching statistical significance in any groups. Notably, only in Group-1 a significant decrease of Treg was observed at 6 months compared to baseline (). Furthermore, while in Group-1 the Treg-to-CD8 ratio remained stable during treatment and then significantly decreased at 6 months, in Group-3 an increase was observed during and at treatment-end, recovering after 6 months ().

The percentage of γδ-T cells, evaluated in terms of CD3+CD4CD8, showed a reduction at 3 h in all groups, then increased at 6 months in Group-2 and Group-3 with respect to baseline and treatment-end (). The evaluation of total NK cells (CD3CD56+) showed a significant increase in Group-1 and 3 at 6 months ().

As regard to B-cell frequency (CD3CD19+), we observed a progressive increase in Group-1 and a progressive decrease in Groups-2 and 3 during RT. Notably, while in Group-1 the proportion of B cells at 6 months resolve to baseline, in Groups-2 and 3 there is only a partial recovery that not reached baseline value ().

Comparison of immune cell frequency among different groups after 6 months of follow-up showed significant differences in CD3+ T-cell subset, higher in Groups-1 and 2 then in Group-3, and in γδ-T cells, higher in Group-3 (, middle panels).

No significant differences were observed in the proportion of T and NK cells between patients before treatments and sex/age-matched HD. Otherwise, B-cells were higher in HD (Supplementary Figure S5), in line with literature.Citation8,Citation13,Citation14

To determine whether different RT treatment strategies may influence functionality of T and B cells, we performed an intracellular staining for cytokines production (IFN-γ, TNF-α and IL-10) after anti-CD3 activation ( and Supplementary Figure S4 a-c). In Group-1 and 3 functionality of CD8+ T cells was not affected by RT treatments, with only a slight reduction of IFN-γ and TNF-α 3 h after RT in Group-1 (). Differently, CD8+ T cells from Group-2 showed a decrease in IFN-γ and TNF-α production along the treatment, with a significant drop at 6 months, compared with treatment-end and baseline. Although at baseline the frequency of T cells was comparable in all groups (, middle panels), we noticed that CD8+ T cells of Group-3 showed a baseline higher level of IL-10, stable at treatment-end and significantly higher with respect to Group-1 ().

Figure 3. Intracellular multicolor staining for cytokine- production (IFN-γ, TNF-α and IL-10), after 5h of anti-CD3 activation, in CD8+ (a-b), CD4+ (c-d) and B (e-f) cells, on 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7). (a, c, e) Dot-plots representing cytokine production in gated CD8+ (a), CD4+ (c) and B (e) cells, from two representative patients, at baseline (PRE) and treatment-end (END-RT). (b-d) Proportion of CD8+ (b) and CD4+ (d) producing IFN-γ (top panels) TNF-α (middle panels) and IL10 (bottom panels), during treatment (left panels) (Wilcoxon U-test). (f) Proportion of B cells producing IL10 during the treatment (Wilcoxon U-test). Right panels, comparison among Groups, (Mann-Whitney U-test). *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point; POST = 6 months after RT; ND = not determined.

Figure 3. Intracellular multicolor staining for cytokine- production (IFN-γ, TNF-α and IL-10), after 5h of anti-CD3 activation, in CD8+ (a-b), CD4+ (c-d) and B (e-f) cells, on 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7). (a, c, e) Dot-plots representing cytokine production in gated CD8+ (a), CD4+ (c) and B (e) cells, from two representative patients, at baseline (PRE) and treatment-end (END-RT). (b-d) Proportion of CD8+ (b) and CD4+ (d) producing IFN-γ (top panels) TNF-α (middle panels) and IL10 (bottom panels), during treatment (left panels) (Wilcoxon U-test). (f) Proportion of B cells producing IL10 during the treatment (Wilcoxon U-test). Right panels, comparison among Groups, (Mann-Whitney U-test). *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point; POST = 6 months after RT; ND = not determined.

Within total CD4+ T cells, we observed a weak functional decrease of TNF-α production in Group-1 at 6 months from RT (), likely to ascribe at the reduction of Treg cells (). In agreement, Group-3 showed higher production of all three cytokines in CD4+ T-cell population at 6 months compared to Group-1, suggesting that 30FRX among the CD4 pool advantages Treg frequency () and functionality ().

Looking at B-cells we found an increase of IL-10 production, at 3 h and treatment-end only in Group-3 (). No significant differences were observed in the cytokine production between PCa patients before treatments and HD, with the exception of IL-10 that was lower in CD4+ T cells of PCa patients (Supplementary Figure S5).

Effect of RT strategies on maturative differentiation

Radiotherapy has been reported to preferentially eliminate CD45RA+ T-cells,Citation15 thus we assessed the longitudinal differentiation status of CD8+ and CD4+ T lymphocytes, based on CD45RA and CCR7 expression ( and Supplementary Figure S4 d,e). Baseline values showed no significant differences among groups ( right panels).

Figure 4. SBRT determines an increase of central-memory and effector-memory T cells. (a-c) Dot-plots showing CCR7 versus CD45RA staining, evaluated by multicolor flow cytometry, in gated CD8+ (a) and CD4+ (c) T cells from two representative patients, during and after RT. (b-d) Pooled results from 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7), showing the proportion of central-memory (CM, CCR7+CD45RA), naïve (CCR7+CD45RA+), effector-memory (EM, CCR7CD45RA) and terminally-effector (EMRA, CCR7CD45RA+) T cells, in gated CD8+ (b) and CD4+ (d) T cells (left panels) (Wilcoxon U-test). Right panels, comparison among Groups (Mann-Whitney U-test). *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point; POST = 6 months after RT.

Figure 4. SBRT determines an increase of central-memory and effector-memory T cells. (a-c) Dot-plots showing CCR7 versus CD45RA staining, evaluated by multicolor flow cytometry, in gated CD8+ (a) and CD4+ (c) T cells from two representative patients, during and after RT. (b-d) Pooled results from 18 patients (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7), showing the proportion of central-memory (CM, CCR7+CD45RA−), naïve (CCR7+CD45RA+), effector-memory (EM, CCR7−CD45RA−) and terminally-effector (EMRA, CCR7−CD45RA+) T cells, in gated CD8+ (b) and CD4+ (d) T cells (left panels) (Wilcoxon U-test). Right panels, comparison among Groups (Mann-Whitney U-test). *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point; POST = 6 months after RT.

As regard to total CD8+ T cells, we observed a progressive increase of central-memory (CM, CD45RACCR7+) CD8+ T cells until the end of RT in all groups, returning to baseline after 6 months for Groups-2 and 3, with a trend of higher percentage in Group 1. Notably, only in Group-1 we observed an increase of the effector-memory subset (EM, CD45RACCR7), accompanied by a decrease of naïve (CD45RA+CCR7+) and terminally-effector cells (EMRA, CD45RA+CCR7) at 6 months after RT ().

As regard to total CD4+ T cells, similarly to CD8+ lymphocytes, an increase of CM subsets occurred at treatment-end in all groups, returning to normal baseline after 6 months, along with a progressive decrease of naïve cells in all groups. Looking at EM subsets, we observed an increase, mostly evident in Group-1 and Group-3 at 6 months of follow-up, compared with baseline and treatment-end ().

From comparing patients versus HD emerged that patients have higher level of both CD4+ CM and CD8+ CM T-cells, and lower levels of CD4+ EM T-cells (Supplementary Figure S5).

Effect of RT strategies on inhibitory receptor (IR) expression

The analysis of total single inhibitory receptor expression in CD8+ T cells revealed that lymphocyte-activation gene-3 (LAG3) was the lowest receptor expressed at baseline, and was almost not affected by treatment, with only a trend of up-regulation in Group-3, similarly to mucin domain containing 3 (TIM3) (). Interestingly, the expression of V-domain immunoglobulin suppressor of T cell activation (VISTA) showed a significant up-regulation at 3 h and treatment-end in Group-1 and at midpoint in Group-3 (, lower panel). Differently, the expression of programmed cell death 1 (PD1) was not affected by RT, with the only exception of transient up-regulation at 3 h in Group-3.

Figure 5. Impact of RT-strategies on inhibitory receptor (IR) expression. (a-c) Analysis of single IR expression (LAG3, PD1, TIM3 and Vista) performed by multicolor staining, in gated CD8+ (a) and CD4+ (c) T cells, in 18 patients during treatment (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7) (Wilcoxon U-test). Right panels, comparison among Groups (Mann-Whitney U-test). (b-d) Quantification of 16 IR simultaneous co-expression in gated CD8+ (b) and CD4+ (d) T cells. Each bar represents the mean (± SEM) percentage (Wilcoxon U test). *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point.

Figure 5. Impact of RT-strategies on inhibitory receptor (IR) expression. (a-c) Analysis of single IR expression (LAG3, PD1, TIM3 and Vista) performed by multicolor staining, in gated CD8+ (a) and CD4+ (c) T cells, in 18 patients during treatment (Group1-3FRX, n = 5; Group2-20FRX, n = 6; Group3-30FRX, n = 7) (Wilcoxon U-test). Right panels, comparison among Groups (Mann-Whitney U-test). (b-d) Quantification of 16 IR simultaneous co-expression in gated CD8+ (b) and CD4+ (d) T cells. Each bar represents the mean (± SEM) percentage (Wilcoxon U test). *p ≤ .05. Abbreviation: FRX = fraction; MP = Middle point.

When evaluating the concomitant expression of IRs we found that PD1, TIM3 and VISTA were merely expressed singularly, with a significant increase of VISTA+PD1LAGTIM CD8+ T cells in Group-1 (). We did not observe simultaneous expression of the 4 IRs in any group.

Regarding total CD4+ T cells, we evidenced that at baseline the IRs expression was almost absent in Group-1, whereas TIM3 and LAG3 were the most expressed in Groups-2 and 3 respectively (). Considering the effect of the different fractionation doses, Group-3 showed the most robust changes, with a significant up-regulation of LAG3 and VISTA, and a trend of increase of PD1 and TIM3. Group-1 displayed only a trend of rise for VISTA, while Group-2 showed a significant increase of LAG3 (). The analysis of simultaneous expression reflected the results from singular study, including a significant up-regulation of LAG3+PD1TIM3VISTA in Group-3 ().

No significant differences were observed in the IRs expression between PCa patients before treatments and HD (Supplementary Figure S5).

Overall, these results indicate that the different strategies did not affect the simultaneous expression of IRs in both CD8+ and CD4+ T cells, but selectively influenced the expression of single IRs, and in particular VISTA in CD8+ T cells in patients treated with SBRT (Group-1).

Discussion

To define optimal combination strategies of radiotherapy and immunotherapy we need to gather information on the effect of different RT fractionation schedules on the systemic immune cell functions. To date, only few data are available in PCa, a tumor often treated with RT, and known to be resistant to immunotherapy approaches such as ICI.Citation9,Citation16 Herein, we performed an in-depth immunomonitoring of peripheral immune cell composition, differentiation stage, cytokine production and IRs expression, from baseline to one year after three different RT treatment strategies.

We have previously shown that prostate SBRT in 3 fractions is feasible and tolerated at one year.Citation6 Here we report, for the first time, that this schedule (Group-1) exerts a favorable effect on immune cell functionality with respect to moderately-hypofractionated RT (Group2-20FRX) and conventionally-fractionated RT (Group3-30FRX). Although our SBRT setting was in 3 fractions, we argue that similar effect on immune cells may be obtained with SBRT in 5 fractions, as reported for other tumors.Citation17,Citation18

In the SBRT cohort we found a transient lymphocyte reduction and a neutrophil increase, returning to baseline within 6 months after RT. Conversely, patients treated with 30 fractions showed a more persistent lymphocyte toxicity with a partial recovery, and a higher neutrophils-to-lymphocytes ratio after 1 year of follow-up (). These results are in agreement with those observed after 20–30 fractions.Citation8,Citation13,Citation15 Furthermore, these data suggest that larger irradiated immune-related volumes (bone marrow and vessels) may impair anti-tumor immunity.Citation19 Indeed, in the present analysis we cannot tease out the effects of dose/fractionation from those of treated volumes. Therefore, the results are to be interpreted as the consequence of a given treatment strategy rather than a single characteristic.

T cell multiparametric flow cytometry analysis highlights that total CD3+ and CD4+ T-cell frequency and functionality are not affected, differently from Groups-2 and 3 where a decrease of both cells was observed 6 months post-treatment (). Notably, Treg frequency and Treg-to-CD8+ ratio decreased only in Group-1, in agreement with data showing that SBRT treatment in lung cancer determines a Treg reduction,Citation18,Citation20 thus supporting the hypothesis that the larger the fraction size the more is the effect on Tregs. On the other hand, the findings that, in patients treated with 30 fractions T-reg/CD8+ ratio increases toward an immune-suppressive environment is consistent with the observation from PCa patients treated with multiple fractions over large pelvic treatment volumes.Citation8,Citation19 This is in agreement with data showing that Tregs are more radio-resistant than effector T cells,Citation21 due to a reduced apoptosis and an increased proliferation ability, thus underscoring the relevance of monitoring Treg population during radiotherapy.Citation22

Although we did not detect any difference in CD8+ effector cell frequency modulation with respect to RT strategies, with a CD8+ increase in all Groups, we found relevant differences evaluating the CD8+ T-cell differentiation and maturation stage. Indeed, only after SBRT we observed a significant increase of both central-memory and effector-memory CD8+ T cells (), a subset with a crucial role in long-lasting immunity and protection against tumor recurrence. We could hence speculate that this schedule may favor the induction of molecular pathways able to increase an efficient antigen-presentation and in turn amplify the anti-tumor specific immune response. This is in line with data from McGee and colleagues that observed an increase of activated memory CD4+ and CD8+ after SBRTCitation23 and from Evans et al. who described an increase of tumor-reactive CD8+ T cells as protective against the risk of PCa progression when treated with SBRT.Citation24 The increase of effector-memory cells 14 days after SBRT was associated with superior overall survival in oligometastatic PCa.Citation25

Another hypothesis that could be generated is that the increase of memory T-cell subsets is due to a preferential elimination of naïve T cells by RT and the resistance of CD45RA T cells to RT-induced apoptosis.Citation15 These data support the rationale for combination of RT with antigen-based vaccination, also considering the several immunogenic antigens specific for PCa such as the prostatic acid phosphatase and the prostate-specific antigen.Citation26

Considering the crucial role of B cells in anti-tumor immune response, B-cell frequency in SBRT-treated patients increases at the end of treatment while this effect was not detectable when RT was performed in 20 or 30FRX. Noteworthy, we found that only in Group-3, B cells produce IL-10 while Group-1 and 2 did not. Further studies are needed to evaluate the composition of B cell subsets and their maturation state, considering the recent role of plasma cells in prolonged recurrence-free survival after surgery for PCa.Citation27

Interestingly, the frequency of NK cells, a key population of innate immune response, increased 6 months after RT (), independently from the schedule, indicating no toxic effect on this population. Our results are in line with several studies in PCa,Citation8,Citation13 lung cancer,Citation20,Citation28 and breast cancer.Citation29 A possible explanation for NK increase could be ascribed to the high intracellular glutathione levels of these cells, which allow to maintain homeostasis after radiation stress, as demonstrated in NK-resistance to antineoplastic agents.Citation30

To date, there is still lack of consensus on the dose, fractionation and timing of RT in combination with immunotherapy. So far ICI therapy, alone or combined with RT, has been tested mostly in metastatic castration-resistant PCa,Citation9,Citation16,Citation31,Citation32 and the translation to earlier stages of disease may raise further challenges. Gao et al. reported that patients treated with Ipilimumab had an increased expression of VISTA,Citation33 indicating that is crucial to in-depth analyze the impact of RT fractionation on the modulation of IRs expression in T cells and in turn in anti-tumor immune response. In the present study, the analysis of IRs showed a significant increase of VISTA, in absence of other IRs (VISTA+PD1LAGTIM- CD8+ subset, ) at the end of SBRT. Therefore, these results confirm the potential role of VISTA, which is expressed as immune suppressive receptor on the myeloid and lymphoid lineages with an impact on both innate and adaptive immunitiesCitation34 and might represent an important new potential immunotherapy target in PCa. Moreover, our results suggest the potential role of VISTA as a new target after SBRT completion, also in consideration of the ability of this dose-fractionation to preserve central-memory and effector CD8+ T-cell functions (). Noteworthy, SBRT has been reported to promote immunogenic cell death, tumor antigens release and in turn immunotherapy efficacy.Citation35 However, differently from PD-1,Citation36 the precise role of VISTA as inhibitorCitation16,Citation37 or activator receptor is still unclear in PCa. Indeed, Loeser et al.Citation38 showed that VISTA expression on tumor-infiltrating lymphocytes was correlated with improved overall survival within esophageal adenocarcinoma, suggesting that VISTA could represents a positive prognostic marker other that a therapeutic target in this cohort of patients.

Differently from what observed with SBRT, patients of Group-3 presented an up-regulation of LAG3+PD1TIM3VISTA (), likely to ascribe to the concomitant increase of immune suppressive Treg. Noteworthy, this phenotype is associated with other immune suppressive factors one year after treatment (), suggesting that conventional-fractionated RT in PCa could negatively impact the immune response.

Our results argue on a favorable role of SBRT on the immune response, and support SBRT as the treatment choice for both localizedCitation7,Citation39 and oligometastatic PCa.Citation40 In the framework of this treatment with logistic convenience, shorter treatment time and equivalent efficacy compared to conventional RT,Citation7 the identification of immunological advantages may be of great impact for both patients and physicians.

It is important to note that this was an exploratory study without a clinical endpoint. The limits of our study were the small number of patients, and the heterogeneity between Groups, especially in terms of AD and treatment volumes. According to Hoffman and colleaguesCitation19 the early suppression of anti-cancer immunity is directly related to large volumes of treatment, and in particular with immune related structures such as blood vessels and bone marrow. Recently two different head and neck studies highlight that lymphatic preserving treatments favor systemic antitumor immunity, whereas elective nodal irradiation may decrease immune cell functionality.Citation41,Citation42 One may argue that this was the case of conventionally fractionated RT (Group-3) where both prostatic bed and pelvic lymph nodes were irradiated. Sini et al.Citation43 has shown how whole pelvis RT in PCa can induce hematological toxicity, and the use of specific constraints on bone marrow may help limiting this effect. In order to try to tease out the effect of treated volume from dose/fractionation, we have decided to contour these additional immune related structures for a further analysis on a larger cohort and to investigate dosimetric parameters which may correlate with a higher immune toxicity in all 3 Groups.

Finally, regarding AD, since in a previous analysisCitation43 no detrimental effect of AD was found in terms of both acute and late hematological toxicities, we do not believe this to be a major confounding factor.

We think that other than the comparison between Groups and the different RT strategies, one of the strengths of our results was the preservation of immune cells functionality in SBRT patients, where the effect measured was entirely pure without any potential influence of AD and/or larger volume with immune related structures irradiated. Further studies with more balanced Groups are needed to strongly confirm our results, and a new study directly comparing CFRT and SBRT in the salvage setting is recruiting patients.

Furthermore, it would be of major clinical interest to determine if all the changes in periphery could reflect microenvironment alteration, as reported by Kane et al.Citation44

Conclusion

Our study provides a detailed analysis on the role of different RT strategies on systemic immune cell functions. The in-depth monitoring at precise timing evaluating frequency, differentiation stage and functionality of several immune cells, the analysis of four IRs, singularly or concomitantly expressed, were, to our knowledge, analyzed for the first time in PCa patients. Importantly, we found that SBRT, other than advantageous for shorter treatment schedule and the health care cost benefit, preserve central-memory and effector-memory T-cell functions and decrease the immunosuppressive Treg population. Further studies are needed to correlate the systemic immune changes with patient’s clinical outcome and to define potential biomarkers of recurrence. Finally, the increase of VISTA observed after SBRT may provide a rationale for combining VISTA blockade after SBRT in PCa patients.

Trial registration

Clinicaltrials.gov NCT04774133, registered February 23 2021, retrospectively registered; https://clinicaltrials.gov/ct2/show/NCT04774133

Ethics approval and consent to participate

The study was conducted according to the guidelines of the Declaration of Helsinki and approved by the Ethics Committee of the ‘Regina Elena’ National Cancer Institute (protocol code RS1163/18).

Author contribution

B.P., A.F., M.B. P.N. and G.S. contributed conception and design of the study. P.N. and G.S. supervised the data. M.B., A.F. and G.S. enrolled and treated patients and collected the clinical information. B.P., I.C and P.N. designed the experiments. B.P., M.P. and S.M. performed the experiments and analyzed the data. B.P., M.B., A.F., I.C. and P.N interpreted data. I.S. and G.F performed statistical analysis. M.L.F. supplied blood samples from healthy donors. B.P., M.B. P.N. and G.S. wrote the manuscript. All authors read and agreed on the final version of the submitted manuscript.

Supplemental material

Supplemental Material

Download PDF (2.3 MB)

Data availability statement

Research data are stored in an institutional repository and will be shared upon reasonable request to the corresponding author.

Disclosure statement

The authors report there are no competing interests to declare

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/2162402X.2023.2174721

Additional information

Funding

This work was financially supported through funding from the “Ricerca Corrente 2022” granted by the Italian Ministry of Health and partially supported by the Italian Association for Cancer Research PN AIRC IG 19822.

References

  • Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–14. doi:10.3322/caac.21660.
  • Morgan SC, Hoffman K, Loblaw DA, Buyyounouski MK, Patton C, Barocas D, Bentzen S, Chang M, Efstathiou J, Greany P, et al. Hypofractionated radiation therapy for localized prostate cancer: executive summary of an ASTRO, ASCO, and AUA evidence-based guideline. Pract Radiat Oncol. 2018;8(6):354–360. doi:10.1016/j.prro.2018.08.002.
  • NCCN. NCCN clinical practice guidelines in oncology prostate cancer. 2022.
  • Kishan AU, Dang A, Katz AJ, Mantz CA, Collins SP, Aghdam N, Chu F-I, Kaplan ID, Appelbaum L, Fuller DB, et al. Long-term outcomes of stereotactic body radiotherapy for low-risk and intermediate-risk prostate cancer. JAMA Netw Open. 2019;2(2):e188006. doi:10.1001/jamanetworkopen.2018.8006.
  • Jackson WC, Silva J, Hartman HE, Dess RT, Kishan AU, Beeler WH, Gharzai LA, Jaworski EM, Mehra R, Hearn JWD, et al. Stereotactic body radiation therapy for localized prostate cancer: a systematic review and meta-analysis of over 6,000 patients treated on prospective studies. Int J Radiat Oncol Biol Phys. 2019;104(4):778–789. doi:10.1016/j.ijrobp.2019.03.051.
  • Magli A, Farneti A, Faiella A, Ferriero M, Landoni V, Giannarelli D, Moretti E, de Paula U, Gomellini S, Sanguineti G. Toxicity at 1 year after stereotactic body radiation therapy in 3 fractions for localized prostate cancer. Int J Radiat Oncol Biol Phys. 2021;111:93–100. doi:10.1016/j.ijrobp.2021.03.027.
  • Widmark A, Gunnlaugsson A, Beckman L, Thellenberg-Karlsson C, Hoyer M, Lagerlund M, Kindblom J, Ginman C, Johansson B, Bjornlinger K, et al. Ultra-hypofractionated versus conventionally fractionated radiotherapy for prostate cancer: 5-year outcomes of the HYPO-RT-PC randomised, non-inferiority, phase 3 trial. Lancet. 2019;394:385–395. doi:10.1016/S0140-6736(19)31131-6.
  • Eckert F, Schaedle P, Zips D, Schmid-Horch B, Rammensee HG, Gani C, Gouttefangeas C. Impact of curative radiotherapy on the immune status of patients with localized prostate cancer. Oncoimmunology. 2018;7:e1496881. doi:10.1080/2162402X.2018.1496881.
  • Venkatachalam S, McFarland TR, Agarwal N, Swami U. Immune checkpoint inhibitors in prostate cancer. Cancers. 2021:13. doi:10.3390/cancers13092187.
  • Maleki Vareki S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J Immunother Cancer. 2018;6(1):157. doi:10.1186/s40425-018-0479-7.
  • Sfanos KS, Bruno TC, Maris CH, Xu L, Thoburn CJ, DeMarzo AM, Meeker AK, Isaacs WB, Drake CG. Phenotypic analysis of prostate-infiltrating lymphocytes reveals TH17 and Treg skewing. Clin Cancer Res. 2008;14(11):3254–3261. doi:10.1158/1078-0432.CCR-07-5164.
  • Sharma P, Pachynski RK, Narayan V, Flechon A, Gravis G, Galsky MD, Mahammedi H, Patnaik A, Subudhi SK, Ciprotti M, et al. Nivolumab Plus ipilimumab for metastatic castration-resistant prostate cancer: preliminary analysis of patients in the checkMate 650 trial. Cancer Cell. 2020;38(4):489–499 e483. doi:10.1016/j.ccell.2020.08.007.
  • Sage EK, Schmid TE, Geinitz H, Gehrmann M, Sedelmayr M, Duma MN, Combs SE, Multhoff G. Auswirkungen von definitiver und Salvage-Strahlentherapie auf die Verteilung von Lymphozytensubpopulationen bei Prostatakarzinompatienten. Strahlentherapie und Onkologie. 2017;193(8):648–655. doi:10.1007/s00066-017-1144-7.
  • Lepone LM, Donahue RN, Grenga I, Metenou S, Richards J, Heery CR, Madan RA, Gulley JL, Schlom J. Analyses of 123 peripheral human immune cell subsets: defining differences with age and between healthy donors and cancer patients not detected in analysis of standard immune cell types. Journal of Circulating Biomarkers. 2016;5:5. doi:10.5772/62322.
  • Tabi Z, Spary LK, Coleman S, Clayton A, Mason MD, Staffurth J. Resistance of CD45RA− T cells to apoptosis and functional impairment, and activation of tumor-antigen specific T cells during radiation therapy of prostate cancer. J Immunol. 2010;185(2):1330–1339. doi:10.4049/jimmunol.1000488.
  • Cha HR, Lee JH, Ponnazhagan S. Revisiting immunotherapy: a focus on prostate cancer. Cancer Res. 1615-1623;2020(80). doi:10.1158/0008-5472.CAN-19-2948.
  • Zhang T, Yu H, Ni C, Zhang T, Liu L, Lv Q, Zhang Z, Wang Z, Wu D, Wu P, et al. Hypofractionated stereotactic radiation therapy activates the peripheral immune response in operable stage I non-small-cell lung cancer. Sci Rep. 2017;7:4866. doi:10.1038/s41598-017-04978-x.
  • Rutkowski J, Slebioda T, Kmiec Z, Zaucha R. Changes in systemic immune response after stereotactic ablative radiotherapy. Preliminary results of a prospective study in patients with early lung cancer. Pol Arch Intern Med. 2017;127(4):245–253. doi:10.20452/pamw.3997.
  • Hoffmann E, Paulsen F, Schaedle P, Zips D, Gani C, Rammensee H-G, Gouttefangeas C, Eckert F. Radiotherapy planning parameters correlate with changes in the peripheral immune status of patients undergoing curative radiotherapy for localized prostate cancer. Cancer Immunol Immunother. 2022;71(3):541–552. doi:10.1007/s00262-021-03002-6.
  • Navarro-Martin A, Galiana IL, Berenguer Frances MA, Cacicedo J, Canas Cortes R, Comas Anton S, Padrones Sanchez S, Bolivar Cuevas S, Parry R, Guedea Edo F. Preliminary study of the effect of stereotactic body radiotherapy (SBRT) on the immune system in lung cancer patients unfit for surgery: immunophenotyping analysis. Int J Mol Sci. 2018;19(12):3963. doi:10.3390/ijms19123963.
  • Persa E, Balogh A, Safrany G, Lumniczky K. The effect of ionizing radiation on regulatory T cells in health and disease. Cancer Lett. 2015;368(2):252–261. doi:10.1016/j.canlet.2015.03.003.
  • Oweida A, Hararah MK, Phan A, Binder D, Bhatia S, Lennon S, Bukkapatnam S, Van Court B, Uyanga N, Darragh L, et al. Resistance to radiotherapy and PD-L1 blockade is mediated by TIM-3 upregulation and regulatory T-cell infiltration. Clin Cancer Res. 2018;24(21):5368–5380. doi:10.1158/1078-0432.CCR-18-1038.
  • McGee HM, Daly ME, Azghadi S, Stewart SL, Oesterich L, Schlom J, Donahue R, Schoenfeld JD, Chen Q, Rao S, et al. Stereotactic ablative radiation therapy induces systemic differences in peripheral blood immunophenotype dependent on irradiated site. Int J Radiat Oncol Biol Phys. 2018;101(5):1259–1270. doi:10.1016/j.ijrobp.2018.04.038.
  • Evans JD, Morris LK, Zhang H, Cao S, Liu X, Mara KC, Stish BJ, Davis BJ, Mansfield AS, Dronca RS, et al. Prospective Immunophenotyping of CD8+ T cells and associated clinical outcomes of patients with oligometastatic prostate cancer treated with metastasis-directed SBRT. Int J Radiat Oncol Biol Phys. 2019;103(1):229–240. doi:10.1016/j.ijrobp.2018.09.001.
  • Zhang H, Orme JJ, Abraha F, Stish BJ, Lowe VJ, Lucien F, Tryggestad EJ, Bold MS, Pagliaro LC, Choo CR, et al. Phase II evaluation of stereotactic ablative radiotherapy (SABR) and immunity in 11C-Choline-PET/CT–Identified Oligometastatic Castration-Resistant Prostate Cancer. Clin Cancer Res. 2021;27(23):6376–6383. doi:10.1158/1078-0432.CCR-21-2510.
  • Fay EK, Graff JN. Immunotherapy in Prostate Cancer. Cancers. 2020;12(7):1752. doi:10.3390/cancers12071752.
  • Weiner AB, Vidotto T, Liu Y, Mendes AA, Salles DC, Faisal FA, Murali S, McFarlane M, Imada EL, Zhao X, et al. Plasma cells are enriched in localized prostate cancer in Black men and are associated with improved outcomes. Nat Commun. 2021;12(1):935. doi:10.1038/s41467-021-21245-w.
  • Shen G, Wang W, Da Y, Li D, Bai L, Tai J, Liu G. Immunomodulatory features of radiotherapy in lung carcinoma. Transl Cancer Res. 2020;9(1):42–48. doi:10.21037/tcr.2019.11.07.
  • Linares-Galiana I, Berenguer-Frances MA, Canas-Cortes R, Pujol-Canadell M, Comas-Anton S, Martinez E, Laplana M, Perez-Montero H, Pla-Farnos MJ, Navarro-Martin A, et al. Changes in peripheral immune cells after intraoperative radiation therapy in low-risk breast cancer. J Radiat Res. 2021;62(1):110–118. doi:10.1093/jrr/rraa083.
  • Multhoff G, Meier T, Botzler C, Wiesnet M, Allenbacher A, Wilmanns W, Issels RD. Differential effects of ifosfamide on the capacity of cytotoxic T lymphocytes and natural killer cells to lyse their target cells correlate with intracellular glutathione levels. Blood. 1995;85(8):2124–2131. doi:10.1182/blood.V85.8.2124.bloodjournal8582124.
  • Cheng K, Wang Y, Chen Y, Zhu J, Qi X, Wang Y, Zou Y, Lu Q, Li Z. Multisite radiotherapy combined with tislelizumab for metastatic castration-resistant prostate cancer with second-line and above therapy failure: study protocol for an open-label, single-arm, phase Ib/II study. Front Oncol. 2022;12:888707. doi:10.3389/fonc.2022.888707.
  • Kwan EM, Spain L, Anton A, Gan CL, Garrett L, Chang D, Liow E, Bennett C, Zheng T, Yu J, et al. Avelumab combined with stereotactic ablative body radiotherapy in metastatic castration-resistant prostate cancer: the phase 2 ICE-PAC clinical trial. Eur Urol. 2022;81(3):253–262. doi:10.1016/j.eururo.2021.08.011.
  • Gao J, Ward JF, Pettaway CA, Shi LZ, Subudhi SK, Vence LM, Zhao H, Chen J, Chen H, Efstathiou E, et al. Vista is an inhibitory immune checkpoint that is increased after ipilimumab therapy in patients with prostate cancer. Nat Med. 2017;23(5):551–555. doi:10.1038/nm.4308.
  • ElTanbouly MA, Croteau W, Noelle RJ, Lines JL. Vista: a novel immunotherapy target for normalizing innate and adaptive immunity. Semin Immunol. 2019;42:101308. doi:10.1016/j.smim.2019.101308.
  • Adorno Febles VR, Blacksburg S, Haas JA, Wise DR. Translating the immunobiology of SBRT to novel therapeutic combinations for advanced prostate cancer. Front Oncol. 2020;10:830. doi:10.3389/fonc.2020.00830.
  • Simon S, Labarriere N. PD-1 expression on tumor-specific T cells: friend or foe for immunotherapy? Oncoimmunology. 2018;7(1):e1364828. doi:10.1080/2162402X.2017.1364828.
  • Huang X, Zhang X, Li E, Zhang G, Wang X, Tang T, Bai X, Liang T. Vista: an immune regulatory protein checking tumor and immune cells in cancer immunotherapy. J Hematol Oncol. 2020;13(1):83. doi:10.1186/s13045-020-00917-y.
  • Loeser H, Kraemer M, Gebauer F, Bruns C, Schroder W, Zander T, Persa O-D, Alakus H, Hoelscher A, Buettner R, et al. The expression of the immune checkpoint regulator Vista correlates with improved overall survival in pT1/2 tumor stages in esophageal adenocarcinoma. Oncoimmunology. 2019;8(5):e1581546. doi:10.1080/2162402X.2019.1581546.
  • Tree AC, Ostler P, van der Voet H, Chu W, Loblaw A, Ford D, Tolan S, Jain S, Martin A, Staffurth J, et al. Intensity-modulated radiotherapy versus stereotactic body radiotherapy for prostate cancer (PACE-B): 2-year toxicity results from an open-label, randomised, phase 3, non-inferiority trial. Lancet Oncol. 2022;23(10):1308–1320. doi:10.1016/S1470-2045(22)00517-4.
  • Chinniah S, Stish B, Costello BA, Pagliaro L, Childs D, Quevedo F, Lucien F, Bryce A, Park SS, Orme JJ. Radiation therapy in oligometastatic prostate cancer. Int J Radiat Oncol Biol Phys. 2022;114(4):684–692. doi:10.1016/j.ijrobp.2022.07.014.
  • Darragh LB, Gadwa J, Pham TT, Court BV, Neupert B, Olimpo NA, Nguyen K, Nguyen D, Knitz MW, Hoen M, et al. Elective nodal irradiation mitigates local and systemic immunity generated by combination radiation and immunotherapy in head and neck tumors. Nat Commun. 2022;13(1). doi:10.1038/s41467-022-34676-w.
  • Saddawi-Konefka R, O’Farrell A, Faraji F, Clubb L, Allevato MM, Jensen SM, Yung BS, Wang Z, Wu VH, Anang N-A. Lymphatic-preserving treatment sequencing with immune checkpoint inhibition unleashes cDC1-dependent antitumor immunity in HNSCC. Nat Commun. 2022;13(1). doi:10.1038/s41467-022-31941-w.
  • Sini C, Fiorino C, Perna L, Noris Chiorda B, Deantoni CL, Bianchi M, Sacco V, Briganti A, Montorsi F, Calandrino R, 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(1):79–84. doi:10.1016/j.radonc.2015.11.020.
  • Kane N, Romero T, Diaz-Perez S, Rettig MB, Steinberg ML, Kishan AU, Schaue D, Reiter RE, Knudsen BS, Nickols NG. Significant changes in macrophage and CD8 T cell densities in primary prostate tumors 2 weeks after SBRT. Prostate Cancer Prostatic Dis. 2022. doi:10.1038/s41391-022-00498-6.