Abstract
Purpose
The goal of this study was to determine if radiation therapy (RT) of human cancer enhances or diminishes tumor-specific T-cell reactivity. This is important if immunotherapy is to be harnessed to improve the outcome of cancer radiotherapy.Experimental design
Lymphocytes were isolated from colorectal cancer (CRC) patients before, during, and after presurgical chemoradiotherapy. Similar samples were taken from prostate cancer patients receiving standard RT. The level of CD8(+) T cells capable of binding tetramers for the tumor-associated antigen survivin, which is overexpressed in both cancer types, was enumerated in HLA-A*0201 patient samples. CD4(+), CD25(high), Foxp3(+) cells were also enumerated to evaluate therapy-induced changes in T(regulatory) cells. For CRC patients, most of whom were enrolled in a clinical trial, pathologic response data were available, as well as biopsy and resection specimens, which were stained for cytoplasmic and intranuclear survivin.Results
Survivin-specific CD8(+) T lymphocytes were detected in the peripheral blood of CRC and prostate cancer patients and increased after therapy in some, but not all, patients. Increases were more common in CRC patients whose tumor was downstaged after chemoradiotherapy. Biopsy specimens from this cohort generally had higher nuclear to cytoplasmic survivin expression. T(regulatory) cells generally increased in the circulation following therapy but only in CRC patients.Conclusion
This study indicates that RT may increase the likelihood of some cancer patients responding to immunotherapy and lays a basis for future investigations aimed at combining radiation and immunotherapy.Free full text
T-Cell Responses to Survivin in Cancer Patients Undergoing Radiation Therapy
Abstract
Purpose
The goal of this, study was to determine if radiation therapy (RT) of human cancer enhances or diminishes tumor-specific T-cell reactivity. This is important if immunotherapy is to be harnessed to improve the outcome of cancer radiotherapy.
Experimental Design
Lymphocytes were isolated from colorectal cancer (CRC) patients before, during, and after, presurgical chemoradiotherapy. Similar samples were taken from pro-state cancer patients receiving standard RT. The level of CDB+ T cells capable of binding tetramers for the tumor-associated antigen survivin, which is overexpressed in both cancer types; was enumerated in HLA-A*0201 patient samples. CD4+, CD25high, Foxp3+ cells were also enumerated to evaluate therapy-induced changes in Tregulatory cells. For CRC-patients, most of whom were enrolled in a clinical trial, pathologic response data were available, as well as biopsy and resection specimens, which were stained for cytoplasmic and intranuclear survivin.
Results
Survivin-specific CD8+ T lymphocytes were detected in the peripheral blood of CRC and prostate cancer patients and increased after therapy in some, but not all, patients. Increases were more common in CRC patients whose tumor was downstaged after chemo-radiotherapy. Biopsy specimens from this cohort generally had higher nuclear to cytoplasmic survivin expression. Tregulatory cells generally increased in the circulation following therapy but only in CRC patients.
Conclusion
This study indicates that RT may increase the likelihood of some cancer patients responding to immunotherapy and lays a basis for future investigations aimed at combining radiation and immunotherapy.
Management of cancers of the rectum and the prostate relies heavily on radiation therapy (RT), but later-stage disease is often hard to control. Harnessing the immune system to assist in the elimination of cancer cells within, and outside, the radiation field could be beneficial in such situations but this will require knowledge of the effects of RT on tumor-specific immunity in humans, about which little is known. Preclinical data are not very helpful, suggesting consequences ranging anywhere from favoring tolerance to enhancing immunity (1–4).
Here, we used a sensitive tetramer assay for the tumor-associated antigen survivin to ask what happens to tumor-specific T-cell responses in colorectal and prostate cancer patients during and after RT. Survivin is highly expressed in many human cancers but is largely undetectable in most normal tissues (5, 6). It augments cell proliferation and survival (7), either by inhibiting caspase-9 and hence apoptosis (8) or by directing chromosome movement during mitosis (9). Its location in the cytoplasm or nucleus may be crucial in determining its function as well its prognostic potential (10–12). Importantly, it is associated with resistance to therapy, including RT (7, 13).
The evidence that survivin is immunogenic is strong. In preclinical models, survivin-reactive CD8+ CTLs can be generated in vitro that efficiently lyse target cells and confer tumor protection on adoptive transfer in vivo (14–16). In humans, survivin-reactive T cells can be detected in primary breast cancer and melanoma lesions and in lymph nodes and blood of cancer patients (17, 18), who also develop anti-survivin antibodies (19). Both animals and patients respond to vaccination with this antigen (20–22).
Unfortunately, clinical experience indicates that adaptive antitumor immune responses generally fail to translate into measurable tumor regression. This has been ascribed to a variety of immune escape mechanisms, one of which is the presence of Tregulatory cells (23). Different types of Tregulatory cells have been described (24 – 26) but the CD4+, CD25high, Foxp3+ subsets are generally considered important in suppressing antitumor immunity (27). Human tumors are frequently infiltrated with such cells (28 – 30), which suppress effector T cells through multiple mechanisms (31, 32). Although older studies in mice showed that tumor-induced suppressor T cells were more radiosensitive than other T-cell subsets (33), there is little data on the radiosensitivity of Tregulatory subsets in humans.
This study asks whether survivin-specific cytotoxic CDB+ T cells can be detected in patients with prostate or colorectal cancer (CRC), whether cancer treatment with RT or chemo-radiotherapy (CRT) alters the tumor-specific immune status in these patients, whether the level of circulating Tregulatory cells is affected, and whether any immune variables correlate with pathologic tumor regression.
Materials and Methods
Chemicals
The following were used: Ficoll-Paque (GE Healthcare Bio-Sciences); human AB serum (OmegaSci.); DMSO and DNase (Sigma); RPMI 1640 with l-glutamine and antibiotics (Fisher); tetramers and anti-CD8 antibody (T8-FlTC; clone SFC121Thy2D3; Beckman Coulter, Inc.); 7-aminoactinomycin D, FITC-anti-human HLA-A2 (clone BB7.2), phycoerythrin (PE)-Cy5-anti-human CD25 (clone M-A251), and R-PE-anti-human CD4 (clone RPA-T4; PharMingen); F1TC-anti-human Foxp3 (clone hFOXY), PE-Cy5-anti-human Foxp3 (clone PCH101), FITC-anti-human CD4 (clone OKT4), and PE-anti-human CD25 (clone BC96; eBioscience); rabbit anti-human survivin (clone NB500-201; Novus Biologicals); and biotinylated anti-rabbit (BA-1000; Vector).
Patients and sample collection
Blood samples came from patients with colorectal cancer (CRC; n = 28) or prostate cancer (n = 20) in the University Hospital Gasthuisberg (Leuven, Belgium), with local Institutional Review Board approval and consent. Prostate patients received conventional RT. All but three with CRC were part of a phase II randomized, double-blind, placebo-controlled clinical trial with the cyclooxygenase-2 inhibitor celecoxib described previously (34). Preoperative CRT was 45 Gy in 25 fractions with continuous 5-fluorouracil infusion. Patients were randomized to celecoxib (2 × 400 mg/d) or placebo before surgery, which was on week 6. Blood samples were taken into Vacutainer CPT tubes (Becton Dickinson) before, during (week 3), and after CRT (week 5). Peripheral blood mononuclear cells (PBMC) were isolated following gradient centrifugation and frozen in human AB serum containing 10% (v/v) DMSO. Frozen blood samples were shipped on dry ice and stored in liquid nitrogen on arrival in the United States. Serial samples of individual patients were assayed for tetramer and Tregulatory cell staining (see below) on the same day. PBMCs from eight healthy volunteers were isolated on Ficoll-Paque at the University of California at Los Angeles and stored as above, Pretreatment biopsies (21) and residual tumor surgery specimens (10) of CRC patients were fixed and processed for immunohistology. This trial was delayed because of cyclooxygenase-2 inhibitor safety issues, resulting in some incompleteness of data.
HLA-A2 testing
PBMCs from patients and healthy subjects were thawed by dilution in prewarmed RPMI 1640 with 10% (v/v) human AB serum. Cells were treated with DNase, washed, and resuspended at 5 × l06/mL in human AB serum. Cells (1 × 105; 20 µl) were stained with 1 µl of FITC anti-HLA-A2 antibody for 30 min at 4°C, washed, and resuspended in 300 µ1 PBS for flow cytometry (FACSCalibur, BD Biosciences).
Terramer-binding assay
Cells (1 ×106) in human AB serum (200 µl) from HLA-A*0201-positive subjects were stained with 8 µl of the MHC tetramer for the HLA-A2-restricted survivin epitope SurIM2 (LMLCEFLKL; ref. 18) along with 8 µl of anti-CD8 antibody. Sample volume permitting (28 samples) a MHC class I human negative tetramer with no known specificity that does not bind CD8+ T cells of any HLA allele (Beckman Coulter) was used to determine back-ground PE fluorescence, After incubation for 30 min at room temperature and washing, samples were resuspended in PBS. 7-Aminoactinomycin D was added to detect nonviable cells 5 to 10 min before flow cytometry. PBMCs from a single HLA-A*0201-positive volunteer were run as an internal control for each assay. Events (1 × 105 to 2 × 105) were accumulated. Quality control required ≥10,000 viable events and ≥2,000 CD8+ T cells.
The gating strategy was
plot FL3, set viability gate (gate 1); (fixed cells as control);
plot FL1 versus FSC of population in gate 1; set gate 2 for CD8highlymphocytes, excluding natural killer cells (CD8low); and
plot FL-2 versus FL-1 of cells in gates 1 and 2 (viable CD8high lymphocytes). Samples of one healthy volunteer stained with negative tetramer were used to set an arbitrary FL-2 lower limit of 0.03% double positives (35).
Two batches of survivin tetramer were used and correction had to be made for differences in binding. Positivity was based on a HLA-A*0201+ healthy volunteer having 0.053 ± 0.023% reactive CD8+ T cells for one batch and five HLA-A*0201+ healthy volunteers having 0.100 ± 0.075% for the other (Table 1). The low limit for a positive value was taken to be the mean ± 2 SD of these values (i.e., 0.099% for batch 1 and 0.25% for the second batch).
Table 1
Tetramer (% of CD8) | CD8 (% of PBMC) | ||||||
---|---|---|---|---|---|---|---|
Before | During | After | Tetramer set | Before | During | After | |
CRC 7 | 0.04 | 0.09 | 1 | 14.9 | 14.1 | ||
CRC 19 | 0 | 0.18 | 1 | 3.2 | 5.4 | ||
CRC 21 | 0.1 | 0.16 | 1 | 15.5 | 1.6 | ||
CRC 22 | 0.76 | 2 | 1.2 | ||||
CRC 23 | 1.57 | 1.93 | 2 | 12.8 | 10.9 | ||
CRC 24 | 1.16 | 0.27 | 0.97 | 2 | 5.78 | 15.5 | 12.1 |
CRC 27 | 1.67 | 0.75 | 2 | 1.9 | 4.8 | ||
CRC 28 | 0.42 | 2 | 9.4 | ||||
CRC 29 | 0.68 | 2.25 | 2 | 18.1 | 24.5 | ||
CRC 30 | 1.48 | 1.46 | 2 | 7.24 | 10.1 | ||
CRC 33 | 0.12 | 0.17 | 2 | 28.2 | 20.8 | ||
CRC 34 | 0.23 | 0.15 | 0.12 | 2 | 8.4 | 7.6 | 5.3 |
Rectum 5 | 0.02 | 0.18 | 1 | 11.9 | 14.9 | ||
Rectum 8 | 0.04 | 0.03 | 0.07 | 1 | 14.7 | 11.5 | 10.5 |
Rectum 10 | 0.01 | 0.07 | 1 | 18.2 | 8.95 | ||
prostate 05 | 0.1 | 0.11 | 1 | 15.2 | 21.6 | ||
prostate 1 | 0.23 | 0.29 | 0.37 | 2 | 16.4 | 13.7 | 13.8 |
prostate 2 | 0.12 | 0.05 | 0.25 | 2 | 17 | 18.7 | 17.7 |
prostate 3 | 0.28 | 0.23 | 0.53 | 2 | 4 | 4.1 | 3.3 |
prostate 4 | 1.33 | 0.16 | 2 | 17.7 | 18.5 | ||
prostate 5 | 0.22 | 0.12 | 2 | 8.9 | 8.1 | ||
prostate 6 | 0.15 | 0.16 | 0.15 | 2 | 13.9 | 15.7 | 7.3 |
prostate 9 | 0.46 | 0.16 | 0.15 | 2 | 5.4 | 7.7 | 9 |
prostate 11 | 0.32 | 0.31 | 0.21 | 2 | 28 | 40 | 36 |
prostate 12 | 0.15 | 0.5 | 0.32 | 2 | 6 | 2.5 | 5.5 |
prostate 13 | 0.1 | 0.14 | 2 | 5.4 | 5.5 |
NOTE: Data are from 15 HLA-A2 – positive patients with CRC and 11 patients with prostate cancer. CRC indicates patients that were part of a cyclooxygertase-2 inhibitor clinical trial. Data are CD8+ Tcells staining positive with the tetramer for survivin (%) and levels of CD8+ PBMCs (%). Two batches of survivin tetra merwere used and are indicated. Five healthy volunteers served as control. Gray fields highlight those samples that stained above the background level (≥mean ± 2× SD of % survivin-reactive CD8+ T cells in the healthy volunteers for that tetramer batch).
Tregulatory cell staining
CD4+, CD25high cells with intracellular Foxp3+ were examined. For most samples, 1 × 106 cells were stained in 100 µl human AB serum with 20 µl FITC-anti-human CD4 and 20 µl PE-anti-human CD25 and incubated for 30 min on ice. Cells were washed and incubated in 1 mL of fixation/permeabilization buffer for 45 min on ice, washed twice, and resuspended in 2% (v/v) normal rat serum in 100 µl of permeabilization buffer. PE-Cy5-anti-Foxp3 (20 µl; clone PCH101) was added followed by 30 min on ice. Cells were washed, resuspended in 200 µl of 10% fetal bovine serum, and analyzed by flow cytometry. In an earlier protocol, the antibody cocktail containing PE-Cy5-anti-CD25, R-PE-anti-CD4, and the first-generation FITC-anti-Foxp3 antibody (clone hFOXY) were applied simultaneously after fixation and permeabilization. PBMCs from one volunteer served as an internal control for each assay. If possible, 1 × 105 events were accumulated. Quality control required all acquired data to be ≥70% viable and ≥2,000 CD4+ T cells.
The gating strategy was
FL-1 versus FSC of all events, set gate 1 for CD4* cells, excluding debris and monocytes (CD4low);
FL2 versus FLI of population in gate 1, set gate 2 for CD4+CD25high double-positive lymphocytes; and.
plot FL-3 of cells in gates 1 and 2 to determine fraction of CD4+CD25highFoxp3+ triple-positive cells.
Immunohistochemistry for survivin
Tissues were deparaffinized at 75°C (30 min) in xylene and decreasing percentages of alcohol and washed five times in water. Sections were steamed in citrate buffer (100 mmol/L, pH 6.0; 25 min) and washed five times with PBS. Endogenous peroxidase was blocked with 3% H2O2 in methanol (15 min), washed, and incubated with 5% normal goat serum in 0.05% Tween 20 in PBS. Polyclonal rabbit anti-human survivin (1:200) was added and slides were incubated (30 min, room temperature; overnight at 4°C). Biotinylated anti-rabbit Ig (1:200) in 5% normal goat serum in 0.05% Tween 20 was added (40 min) followed by 3,3′-diaminobenzidine (3 min). After counterstaining with hematoxylin (10 s), slides were dehydrated and mounted. The identity of slides was blinded through a number code and scored in Belgium. Cytoplasmatic survivin was scored for percentage tumor tissue staining positive and for intensity on a scale from 0 to 3. For the nuclear staining, we only scored the percentage because the intensity did not vary.
Statistics
All but one set of data were analyzed for statistical significance with the sign test and, after a square root transformation, with a Student's t test (36). Whether the level of circulating Tregulatory cells was significantly different from control levels was determined with the Wilcoxon signed rank test (36). Statistical significance was at the 5% level. In general, pooled patient data sets were compared as cohorts to the healthy control levels. Longitudinal responses for each patient compared outcome values (during or after) to individual baseline levels (before) and were then summarized for the whole cohort.
Box plots are box whisker diagrams summarizing the distribution of data as (a) the box spanning the 75% to 25% percentile and (b) the median (line), (c) the minimum and maximum (whiskers above and below the box), and (d) individual outlying data points (open circles).
Results
Survivin-specific CD8+ T cells
Of 49 patients, 28 (57%) were HLA-A*0201 positive and eligible for tetramer analysis. However, two samples did not meet the quality control standards and were excluded from analyses.
Levels of survivin tetramer-reactive CD8+ cells were significantly higher in patients than five healthy controls (P < 0.001; Supplementary Figure), indicating the presence of antigen-specific T cells, and exceeded those for the negative control tetramer for all cohorts (P < 0.001).
Samples for tetramer analysis were available for 10 patients before, during, and after treatment, for 14 patients from two time points, and for 2 patients at a single time point. Overall, there were 19 lymphocyte samples before RT, 21 during RT, and 20 after RT (Table 1).
Samples from four of nine (44%) CRC patients before CRT treatment were positive for survivin tetramer binding (>2 SD from the mean of healthy controls), 5 of 10 (50%) during treatment, and 8 of 12 (67%) after treatment (Table 1). This trend toward increased responses on completion of CRT in CRC patients (P = 0.499) was not apparent in prostate cancer patients (P = 0.352) who exceeded the criterion of positivity in 5 of 10 (50%) cases before RT, 4 of 11 (36%) during RT, and 4 of 8 (50%) after RT.
Treatment-dependent responses to survivin
Because samples were taken before, during, and after treatment, we were able to evaluate individual patient responses over time (Fig. 1A). The percent survivin-specific CD8+ T cells increased in 9 of 13 (69%) CRC patients as a result of treatment, including before → during RT (2 of 5 cases), during → after RT (4 of 7), and before → after RT (5 of 7). Statistical significance was not reached (P = 0.267), but in only 2 of 13 cases (15%) was there a clear decrease in survivin-specific CD8+ T cells after CRT. Seven of 11 (64%; P = 0.599) prostate cancer patients also responded to RT with an increase in survivin-specific CD8+ T cells, including before → during RT (3 of 10 cases), during → after RT (5 of 8 cases), and before → after RT (5 of 7 cases). Hence, CRT of CRC and RT of prostate cancer generally increased the percent of circulating survivin-specific T cells in the CD8+ pool (P = 0.076).
Interestingly, the ratio of CD4+ to CD8+ T cells increased in most patients from before to after completion of therapy (PCRCpatients = 0.071 and Pprostate = 0.47; Fig. 1B). This was most marked for CRC patients, who had a low CD4 to CD8 ratio before therapy compared with all other cohorts (P = 0.008, compared with healthy volunteers). Overall, cancer patients tended to have less CD4+ in their circulation than healthy control subjects (33.6 ± 6.5%) at the beginning of therapy (Supplementary Table).
Time course of response of circulating Tregulatory cells
The number of Tregulatory cells varied in eight healthy volunteers (aged 30–64) from 1% to 4.6% of total CD4+ (mean, 2.8 ± 1.1% CD4+CD25highFoxp3+) cells, which compares well with the literature, indirectly validating our protocol. Most patients started treatment with less Tregulatory cells in their circulation than the volunteers (Fig. 2A; Supplementary Table). This was true for 7 of 8 (88%; P = 0.182) CRC patients and for 13 of 17 (76%; P = 0.083) prostate cancer patients. All seven CRC patients with matched samples from before and after treatment ended the study with more Tregulatory cells than they had initially; hence, there was an overall trend for these values to increase (P = 0.039; Fig. 2B). Notably, the majority of prostate cancer patients (8 of 13) did not adhere to this trend (Fig. 2B).
Intratumoral survivin expression
Tumor biopsies and resections from patients who were part of the CRC trial were stained with an antibody that recognizes both cytoplasmic and nuclear surviving. Examples are shown in Fig. 3. All biopsies, with one exception, tested positive for cytoplasmic survivin, in most cases covering an extensive area of the tumor, whereas survivin was mostly undetectable in normal colon tissue, with the exception of crypts. Nuclear staining was seen more sporadically and in a smaller area of the tumor (Table 2).
Table 2
CRC patient | T downstaging | N downstaging | HLA-A2 | Tetramer increasing | Tregulatory increasing | Survivin expression in biopsy | Survivin expression in resect | ||||
---|---|---|---|---|---|---|---|---|---|---|---|
% cyto. | intens. cyto. | % nuclear | % cyto. | intens. cyto. | % nuclear | ||||||
11 | 0 | 0 | 74 | 3 | 14 | 52.5 | 2 | 3.75 | |||
12 | 0 | 1 | 0 | 1 | 80 | 1 | 10 | ||||
16 | 0 | 1 | 0 | 90 | 2 | 0 | 100 | 3 | 5 | ||
17 | 0 | 0 | 73.3 | 1 | 3.3 | ||||||
19 | 0 | N/A | 1 | 1 | 77.5 | 2 | 3 | ||||
24 | 0 | 1 | 0 | 1 | 92 | 2 | 24 | ||||
26 | 0 | 1 | 0 | 1 | 0 | 0 | 20 | 34.2 | 1 | 1.4 | |
27 | 0 | 0 | 1 | 0 | N/A | 58 | 1 | 0 | 77 | 3 | 7 |
33 | 0 | 1 | 1 | 1 | 1 | ||||||
1 | 1 | 1 | 0 | 66.7 | 2 | 6.7 | |||||
2 | 1 | 1 | 0 | 70 | 2 | 7.5 | |||||
3 | 1 | 1 | 74 | 2 | 12 | ||||||
4 | 1 | 0 | 90 | 3 | 2 | ||||||
7 | 1 | 1 | 1 | 1 | 50 | 1 | N/A | 55.4 | 2 | 0.38 | |
8 | 1 | N/A | N/A | 88 | 3 | 1 | |||||
9 | 1 | 1 | 68.3 | 3 | 28.3 | ||||||
10 | 1 | 1 | 88 | 2 | 40 | ||||||
18 | 1 | N/A | 70 | 3 | 1.25 | ||||||
20 | 1 | 1 | 0 | 32.8 | 2 | 37.14 | 100 | 3 | 0 | ||
21 | 1 | 0 | 1 | 1 | 65 | 1 | 2.5 | ||||
22 | 1 | 1 | 1 | N/A | N/A | ||||||
23 | 1 | 1 | 1 | 1 | 30 | 1 | 0 | ||||
28 | 1 | 1 | 1 | N/A | N/A | 10 | 1 | 40 | |||
29 | 1 | 1 | 1 | 1 | 1 | 40 | 1 | 0 | |||
30 | 1 | 1 | 1 | 0 | 1 | 75 | 2 | 0 | |||
31 | 1 | 1 | 0 | 1 | |||||||
32 | 1 | 1 | 0 | 1 | |||||||
34 | 1 | 1 | 1 | 0 | 0 | ||||||
35 | 1 | 1 | 0 | 1 | |||||||
36 | 1 | 1 | 0 | 1 |
NOTE: “1” is a positive indicator for the respective variable, whereas “0” specifies a negative response. Tumor (n = 21) and lymph node (n = 22) stage decreased or did not (n = 9 and 5, respectively). Twelve patients were HLA-A2 positive. The number of survivin-reactive CDs+ T cells increased in the peripheral blood in four of six responding patients and in two of four nonresponders. The frequency of systemic Tregulatory cells increased in six of seven responding patients and in four of four nonresponders. All biopsies tested stained positive for survivin, which was mostly in the cytoplasm and lesser so in the nucleus. In most samples, nuclear but not cytoplasmic survivin staining decreased and, in all resection specimens from the three responders, was essentially zero.
Abbreviations: % cyto., percent tumor staining positive for survivin in cytoplasm; intens. cyto., intensity of the cytoplasmic staining; % nuclear, percent tumor staining positive for survivin in the nucleus; N/A, not available.
Clinical responses
For patients in the clinical CRC trial, Table 2 summarizes the data on clinical tumor and lymph node stage and immunologic variables of survivin-specific CD8+ T cells, Tregulatory cells, and intratumoral survivin expression. The patient numbers and subgroups were too small and the responses were too variable to derive statistical significance, but there were indications that a larger study could valuably explore.
Biopsy specimens from patients whose tumors were downstaged tended to have higher nuclear survivin levels at biopsy than those who were not downstaged (Fig. 4A, right). Both the ratio of nuclear to cytoplasmic survivin and staining intensity in biopsy specimens were higher for the responders (Fig. 4B and C). The number of resection specimens was inevitably low, but both cytoplasmic and nuclear survivin were decreased over the comparable biopsy specimens, particularly for nuclear staining (Fig. 4A and B). Only six patients who responded with T downstaging and four who did not respond could be evaluated for therapy-related changes in survivin tetramer-positive T cells, with the former showing more promising changes (Fig. 4D).
Discussion
Although reverse immunology has clearly established that survivin is immunogenic, it remains uncertain whether anti-survivin responses occur in untreated cancer patients. We were clearly able to detect T cells binding survivin tetramer in almost half of CRC and prostate patients before treatment when compared with healthy controls. This agrees with Coughlin et al. (37), who measured similar frequencies in pediatric cancer patients using the same tetramer, and Grube et al. (38), who detected survivin-reactive T cells in 40% of patients with multiple myeloma using IFN-γ mRNA as a readout, It contrasts Casati et al. (15), who, using a different tetramer, reported that <0.1% of CD8+ T cells from a CRC patient bound survivin, although this level could be boosted by in vitro stimulation.
From a phase l clinical vaccination trial, we know that anti-survivin responses are not easily induced in CRC patients with only 1 of 15 responding (39). Assessing whether responses increase or decrease following RT or CRT alone therefore inevitably stretches the sensitivity and reproducibility of the assays being used. Nonetheless, we observed that tumor-specific T cells clearly increase in most CRC patients after completion of CRT and in most prostate cancer patients after RT. Perhaps more important is that only a few patients showed a decrease in survivin-reactive CD8+ T cells, which suggests that their ability to respond is not compromised by treatment. This was also suggested in a randomized phase II clinical trial in prostate cancer patients where RT did not obstruct T-cell responses to prostate-specific antigen when given at the end of a cancer vaccination regimen (40).
It is tempting to ascribe the increase in tumor-specific T cells to a radiation-induced increase in antigenic peptide liberation (41) and presentation by dendritic cells, which we have shown can be boosted by radiation (42). The rate of tumor regression (i.e., tumor kinetics) may modulate this and hence the generation of immunity. The fact that this was not detected (40) by enzyme-linked immunospot in patients with prostate cancer receiving RT could be due to the relative sensitivity of the assays. We have not been able to distinguish the two treatment arms of the CRC cohort with some patients receiving the cyclooxygenase-2 inhibitor celecoxib because this trial is still ongoing. There are suggestions in the literature that celecoxib might further enhance the development of antigen-specific Teffector cells (43, 44), but this will not alter the conclusions from the study. The fact that CRC patients who responded with T downstaging tended to have higher levels of nuclear survivin suggests that this is a potential predictive marker of response, with the understanding that nuclear survivin relates more to proliferation and hence may signal a more rapid response (11, 45).
There are several ways to interpret radiation-induced antitumor-reactive CD8+ T-cell levels. Loss of immune suppression due to decreased tumor burden is one. In recent years, Tregulatory cells have gained prominence as a powerful suppressive mechanism. The frequency of Tregulatory cells that we detected in the majority (80%) of our patients was actually lower than in healthy subjects, who were well within the published range for normal individuals (30, 46). This contrasts to several reports of high levels of circulating Tregulatory cells in cancer patients (30, 46, 47).
In our study, levels of Tregulatory cells in CRC patients increased on completion of CRT, whereas this did not happen in prostate cancer patients. It may be that in CRC Tregulatory cells, and perhaps other CD4+ T-cell subsets, relocate to tumor sites, for which there is evidence (28), and are remobilized by therapy-induced changes in the tumor micro-environment. RT-induced adhesion molecule and chemokine expression could alter migratory behavior of Tregulatory cells (30, 48). It is also feasible that systemic effects induced by the CRT more so than by RT could have contributed to such a selective increase in Tregulatory cells in CRC by affecting the balance in lymphocyte subpopulations (49). Such increases in circulating Tregulatory cells may therefore be more apparent than real. Even local irradiation can affect the lymphocyte balance because lymphocyte subsets have differential sensitivity to radiation, with the simplified picture being that naive T cells are more sensitive than effector cells (50), whereas antigen-induced Tregulatory cells gain in radiosensitivity (33). The fact that we observed an increase in CD4+CD25high Foxp3+ Tregs might simply reflect changes in other CD4+ subpopulations.
Perhaps, the most important point from this study is that CRT and RT do not induce immune tolerance to survivin, making immunotherapy approaches feasible in combination with RT. Furthermore, tetramer technology coupled with other flow-based methods provides us with powerful tools to study the antitumor immune status in patients undergoing such treatments.
Acknowledgments
Grant support: Jonsson Comprehensive Cancer Center at University of California at Los Angeles and Department of Defense postdoctoral training award W81XWH-87-1-0014 (D. Schaue) and National Cancer Institute grant R01 CA-101752 (W.H. McBride). K. Haustermans is supported by a fundamental clinical mandate of the Foundation for Scientific Research Flanders.
Footnotes
Disclosure of Potential Conflicts of Interest
No potential conflicts of interest were disclosed.
Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/)
References
Full text links
Read article at publisher's site: https://doi.org/10.1158/1078-0432.ccr-07-4462
Read article for free, from open access legal sources, via Unpaywall: https://aacrjournals.org/clincancerres/article-pdf/14/15/4883/1976283/4883.pdf
Free to read at clincancerres.aacrjournals.org
http://clincancerres.aacrjournals.org/cgi/content/abstract/14/15/4883
Free after 12 months at clincancerres.aacrjournals.org
http://clincancerres.aacrjournals.org/cgi/content/full/14/15/4883
Free after 12 months at clincancerres.aacrjournals.org
http://clincancerres.aacrjournals.org/cgi/reprint/14/15/4883.pdf
Citations & impact
Impact metrics
Citations of article over time
Smart citations by scite.ai
Explore citation contexts and check if this article has been
supported or disputed.
https://scite.ai/reports/10.1158/1078-0432.ccr-07-4462
Article citations
Immune effects of α and β radionuclides in metastatic prostate cancer.
Nat Rev Urol, 21(11):651-661, 27 Aug 2024
Cited by: 1 article | PMID: 39192074
Review
Radiotherapy enhances efficacy of PD-1 inhibitors in advanced hepatocellular carcinoma: A propensity-matched real-world study.
Chin Med J (Engl), 137(11):1332-1342, 09 May 2024
Cited by: 1 article | PMID: 38725345 | PMCID: PMC11191029
A new target of radiotherapy combined with immunotherapy: regulatory T cells.
Front Immunol, 14:1330099, 08 Jan 2024
Cited by: 0 articles | PMID: 38259489 | PMCID: PMC10800811
Review Free full text in Europe PMC
TORCH-R trial protocol: hypofractionated radiotherapy combined with chemotherapy and toripalimab for locally recurrent rectal cancer: a prospective, single-arm, two-cohort, phase II trial.
Front Oncol, 13:1304767, 20 Nov 2023
Cited by: 0 articles | PMID: 38053659 | PMCID: PMC10694348
A review on lymphocyte radiosensitivity and its impact on radiotherapy.
Front Oncol, 13:1201500, 03 Aug 2023
Cited by: 10 articles | PMID: 37601664 | PMCID: PMC10435323
Review Free full text in Europe PMC
Go to all (94) article citations
Data
Similar Articles
To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.
The apoptosis inhibitor protein survivin induces tumor-specific CD8+ and CD4+ T cells in colorectal cancer patients.
Cancer Res, 63(15):4507-4515, 01 Aug 2003
Cited by: 60 articles | PMID: 12907624
CD8+ T cells reactive to survivin antigen in patients with multiple myeloma.
Clin Cancer Res, 13(3):1053-1060, 01 Feb 2007
Cited by: 20 articles | PMID: 17289902
Circulating tumor antigen-specific regulatory T cells in patients with metastatic melanoma.
Proc Natl Acad Sci U S A, 104(52):20884-20889, 18 Dec 2007
Cited by: 112 articles | PMID: 18093940 | PMCID: PMC2409236
Survivin-derived peptide epitopes and their role for induction of antitumor immunity in hematological malignancies.
Leuk Lymphoma, 47(6):978-985, 01 Jun 2006
Cited by: 13 articles | PMID: 16840186
Review
Funding
Funders who supported this work.
NCI NIH HHS (3)
Grant ID: R01 CA-101752
Grant ID: R01 CA101752
Grant ID: R01 CA101752-04