Abstract
Immune checkpoint blockade enhances antitumor responses, but can also lead to severe immune-related adverse events (IRAE). To avoid unnecessary exposure to these potentially hazardous agents, it is important to identify biomarkers that correlate with clinical activity and can be used to select patients that will benefit from immune checkpoint blockade. To understand the consequences of CTLA-4 blockade and identify biomarkers for clinical efficacy and/or survival, an exploratory T cell monitoring study was performed in a phase I/II dose escalation/expansion trial (n = 28) of combined Prostate GVAX/ipilimumab immunotherapy. Phenotypic T cell monitoring in peripheral blood before and after Prostate GVAX/ipilimumab treatment revealed striking differences between patients who benefited from therapy and patients that did not. Treatment-induced rises in absolute lymphocyte counts, CD4+ T cell differentiation, and CD4+ and CD8+ T cell activation were all associated with clinical benefit. Moreover, significantly prolonged overall survival (OS) was observed for patients with high pre-treatment frequencies of CD4+CTLA-4+, CD4+PD-1+, or differentiated (i.e., non-naive) CD8+ T cells or low pre-treatment frequencies of differentiated CD4+ or regulatory T cells. Unsupervised clustering of these immune biomarkers revealed cancer-related expression of CTLA-4+ in CD4+ T cells to be a dominant predictor for survival after Prostate GVAX/ipilimumab therapy and to thus provide a putative and much-needed biomarker for patient selection prior to therapeutic CTLA4 blockade.
Electronic supplementary material
The online version of this article (doi:10.1007/s00262-012-1330-5) contains supplementary material, which is available to authorized users.
Keywords: Ipilimumab, Prostate GVAX, Biomarker, Patient selection, Survival prediction
Introduction
Prostate cancer is the third leading cause of cancer-related death in men worldwide [1, 2]. Curative treatment options are only available for localized disease. In patients that develop castration-resistant prostate cancer (CRPC), the median survival is 16–21 months [3–5]. Since chemotherapy only offers limited survival benefit at this stage, alternative treatment options are widely investigated [6]. Recent advances have led to novel immunotherapeutic strategies with clinical efficacy in metastatic CRPC (mCRPC) patients, notably the prostate-specific antigen-expressing poxvirus vaccine PROSTVAC and sipuleucel-T (Provenge), which led to survival benefits of 8.5 and 4.5 months, respectively [2–4].
In recent years, progress has also been made in cancer immunotherapy by the clinical exploration of the new regulatory targets cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed death-1 (PD-1) [4, 7, 8]. CTLA-4 and PD-1 are co-inhibitory molecules that are expressed on T cells upon activation and that play crucial roles in maintaining immune homeostasis by down-regulating T cell signaling, thereby preventing unbridled T cell proliferation while maintaining tolerance to self-antigens. Interference with these molecules has shown great promise in enhancing antitumor responses in mice and man [9, 10]. CTLA-4 blockade through systemic administration of the CTLA-4-blocking antibody ipilimumab has shown clinical activity in a variety of human cancers [11–13]. Moreover, CTLA-4 blockade enhanced antitumor immunity when combined with other agents, including GM-CSF and GM-CSF-secreting cancer vaccines, for example, GVAX immunotherapy [14–16]. Yet, caution is warranted as immune checkpoint blockade has led to the breaking of tolerance, thereby inducing immune-related adverse events (IRAE) like colitis, dermatitis, hepatitis, alveolitis, and hypophysitis [17, 18]. Although the reported IRAE were generally manageable by interruption or cessation of CTLA-4 therapy and/or high-dose steroids, they can be severe and even potentially life-threatening. To date, attempts to reduce autoimmunity have been unsuccessful [19]. To avoid unnecessary exposure to such a potentially hazardous agent, it is of great importance to identify biomarkers that correlate with clinical activity and can be used to recognize and select patients that will not benefit from CTLA-4 blockade therapy.
We recently reported clinical findings from a phase I/II dose escalation/expansion trial of patients with mCRPC, receiving a combination of a GM-CSF-engineered allogeneic immunotherapy (Prostate GVAX) and escalating doses of the anti-CTLA-4 antibody ipilimumab [20]. Clinical results included partial responses (PR) and survival benefit exceeding survival rates observed in control arms of recent phase III trials [3–5]. To understand the underlying mechanism(s) of action of the Prostate GVAX/ipilimumab combination and identify biomarkers for clinical efficacy and/or survival, extensive T cell monitoring was performed. To this end, T cell-related immune markers were selected prospectively and monitored during treatment, after which cut-off points for response to treatment and/or survival were determined in retrospect. This exploratory study led to the identification of a number of preexisting and treatment-induced T cell-related immune biomarkers that provide a predictive profile for clinical benefit after Prostate GVAX and/or ipilimumab immunotherapy. Most notably, CTLA4 expression in CD4+ T cells was pinpointed as a dominant factor in this regard.
Materials and methods
Prostate GVAX and ipilimumab
The Prostate GVAX vaccine is a cellular vaccine consisting of two prostate cancer cell lines, LNCaP (CG8711) and PC-3 (CG1940), which have been transduced with an adeno-associated viral vector to secrete GM-CSF. These cell lines were propagated, frozen, and irradiated to arrest further cell division [21, 22]. The product was stored and shipped in gaseous nitrogen phase and administered within 60 min after thawing. All manufacturing was conducted according to good manufacturing practice. Ipilimumab (MDX-010), a fully human IgG1κ monoclonal antibody directed against CTLA-4, was provided by Medarex/Bristol-Myers Squibb (Plainsboro, NJ, USA).
Study population and sampling of peripheral blood
Twenty-eight chemonaive patients with asymptomatic mCRPC received 13 biweekly vaccinations of the Prostate GVAX vaccine (as described [21, 22]) and 6 four-weekly infusions of ipilimumab from the time of prime vaccination. In the first 12 patients, ipilimumab was administered at escalating doses of 0.3, 1, 3, and 5 mg/kg (3 patients each). In the expansion phase, 16 additional patients were included at 3 mg/kg ipilimumab [20].
Responses to treatment were defined as described [23]. In brief, PSA partial response (PR) was defined as >50 % PSA decline from baseline, which was confirmed by a second PSA test 3 or more weeks later. PSA progressive disease (PD) was defined as >25 % PSA increase and an absolute increase of 2 ng/ml or more from baseline, whereas stable disease (SD) was defined as no PR and no PD on treatment.
For immunological monitoring, blood samples were taken from the patients before start of therapy and every 4 weeks until 4 weeks after the last treatment (i.e., follow-up (fu)). Peripheral blood was used for whole-blood analysis by flow cytometry, or for peripheral blood mononuclear cell (PBMC) isolation by density centrifugation (Nycomed AS, Oslo, Norway). PBMC was either immediately stained and assessed by flow cytometry, or cryopreserved for later analysis. Absolute lymphocyte counts (ALC) were determined by routine differentiated white blood cell counts (WBC). For CD4+CTLA-4 frequency analysis, we also collected blood from eight sex- and age-matched healthy volunteers.
Antibodies and 4-color flow cytometry
Peripheral blood-circulating lymphocyte frequencies and activation status were flow cytometrically assessed before, during, and after treatment by routine whole-blood or PBMC staining. Whole-blood analysis was performed with lysing solution (BD Biosciences, Mountain view, CA) as described [24]. Cell surface antibody staining of PBMC was performed in FACS buffer for 30 min at 4 °C. Intracellular FoxP3 and CTLA-4 staining was conducted with the antihuman FoxP3 staining kit (eBioscience, San Diego, CA) according to manufacturers’ protocol. The following antibodies were used: fluorescein isothiocyanate (FITC)-, phycoerythrin (PE)-, peridinin chlorophyll protein-Cy5.5 (PerCP)-, or allophycocyanin (APC)-labeled antibodies directed against human CD3, CD4, CD8, CD14, CD16, CD19, CD25, CD27, CD45, CD45RO, CD45RA, CTLA4, HLA-DR, PD-1 (all BD Bioscience), CD56 (IQ products, Groningen, the Netherlands) FoxP3 and ICOS-biotin (eBioscience, San Diego, CA), APC-conjugated streptavidin (BD Biosciences), and matching isotype control antibodies. Stained cells were analyzed on a FACScalibur (BD Biosciences) using Cell Quest software. Events collected were 100,000–150,000 per sample.
T cell subset and differentiation state definitions
Naive CD4+ T cells were defined as CD4+CD45RO− cells and effector/effector memory CD4+ T cells as CD4+CD45RO+ cells (also referred to as non-naive CD4+ T cells). For CD8+ T cells, naive T cells were defined as CD27+CD45RA+, effector cells as CD27−CD45RA+, effector memory cells as CD27−CD45RA−, and central memory cells as CD27+CD45RA− [25]. The non-naive CD8+ T cell population is the sum of effector, effector memory, and central memory T cells. Tregs were defined as CD3+CD4+CD25hi and FoxP3+. As FoxP3 has also been described to be transiently up-regulated on dividing (activated) effector T cells [26–28], we also analyzed FoxP3 expression within these activated (effector-like) T cells, which we defined as CD4+CD25intermediate (CD4+CD25int) cells. Conventional CD4+ T cells were defined as CD4+CD25−/int. As CTLA-4 expression was determined within the total T cell population, the percentage of CTLA-4-expressing conventional T cells was determined by subtracting the percentage of CD4+ Tregs (which all express CTLA-4 [29]) from the total percentage of CD4+CTLA-4+ T cells.
Statistical analyses
Differences between immune parameters before treatment (w0v1; w = week; v = visit; i.e., baseline levels) and during and/or after treatment (w4v3, w8v5, w12v7, w16v9, w20v11, w24v13, and follow-up [fu, i.e., 1 month after last Prostate GVAX and 2 months after last ipilimumab administration]) were analyzed with the repeated measures ANOVA with a post hoc Dunnett’s multiple comparisons test. To determine whether the identified immune parameters were indicative of response to treatment or useful for survival prediction, optimal cut-off points were determined by Cox regression analysis, according to which patients were subsequently divided into two groups. OS for the two groups was plotted using the Kaplan–Meier method, and statistical significance of the survival distribution was analyzed by log-rank testing. In case changes occurred after the first event, patients were excluded from the analysis (this proved only necessary for 1 patient and only in the survival analysis according to absolute lymphocyte counts). To analyze whether prognosis impacted the value of the identified response/survival parameters, the median HPS was also determined for both groups [30]. Differences in HPS between groups of patients and in parameters between prostate cancer patients and age- and sex-matched healthy volunteers were analyzed with the two-sample Mann–Whitney U test. Above-listed statistical analyses were performed with either GraphPad or SPSS software. Differences were considered significant when p < 0.05. To identify clusters of correlated markers, hierarchical cluster analysis using TIGR software was performed and complete linkage analysis was done by Pearson correlation analysis. For this purpose, the values of the treatment-induced and pre-treatment parameters were taken for each patient and divided by the cut-off value, after which the resulting ratios were log-transformed (base 2). Three patients were excluded from this analysis since <70 % of the analyzed biomarkers were available for these patients due to withdrawal from the study or sampling failure.
Results
Clinical results
In this study 28 CRPC patients received 13 biweekly injections of the Prostate GVAX vaccine and 6 four-weekly infusions of ipilimumab. Based on serum PSA levels, five patients experienced PR with PSA declines of more than 50 % and 12 demonstrated disease stabilization (SD) during treatment; PR/SD was significantly correlated with prolonged overall survival (p = 0.0034; [20]). Moreover, the median overall survival (OS) was 29 months, whereas median survival of patients with similar disease characteristics is generally around 16–21 months [3–5]. IRAE were observed in nine patients. Seven patients, five of which with a PR, developed grade 2–3 endocrinopathies consistent with hypophysitis; two of these patients also developed grade 4 alveolitis or grade 1–2 colitis [20].
Absolute lymphocyte count (ALC) and T cell differentiation in relation to survival
As shown in Fig. 1a, absolute lymphocyte counts (ALC) significantly increased upon prostate GVAX/ipilimumab treatment, and increases of more than 25 % were associated with prolonged OS (median survival 41 vs. 20.5 months, HR = 0.181, 95 % CI = 0.046–0.720, p = 0.015; Fig. 1b). To assess the frequency and activation state of peripheral blood-circulating lymphocytes before, during, and after treatment, routine whole-blood or PBMC samples were analyzed by flow cytometry. Despite the observed ALC rises, no differences were observed in overall frequencies of circulating CD3+, CD4+, and CD8+ T lymphocytes and of CD3−CD56+ NK cells following treatment (Fig. 1c). Prostate GVAX/ipilimumab therapy induced T cell differentiation, as reflected by increased percentages of CD4+CD45RO+ memory T cells and of effector and effector/central memory CD8+ T cells (defined by CD45RA and CD27 expression) (see Fig S1A and C, and Table 1). Treatment-induced increases of >30 % of non-naive (memory) CD4+ T cells, but not of non-naive (effector and central/effector memory) CD8+ T cells, were associated with significantly prolonged OS (median survival, 41 vs. 20 months, HR = 0.402, 95 % CI = 0.147–0.940, p = 0.036; Fig. S1B and D, and Table 1).
Fig. 1.
Increased absolute lymphocyte counts (ALC) following Prostate GVAX/ipilimumab therapy are associated with prolonged survival. Absolute numbers of lymphocytes were determined before (w0), during (w4, w8, w16), and after (fu) Prostate GVAX/ipilimumab treatment. a The number of lymphocytes (×1,000) per μl blood is shown for 28 patients. b Kaplan–Meier curve for increases in ALC. The number of patients and corresponding median survival for each group are given. c Levels of circulating CD3+, CD4+, and CD8+ T lymphocytes and CD3−CD56+ NK cells were determined before (w0), during (w4, w8, w12, w16, w20, and w24), and after (follow-up (fu)) Prostate GVAX/ipilimumab treatment by whole-blood flow cytometric analysis. Mean percentage ± SEM of CD3+, CD4+, CD8+, and CD3-CD56+ cells is shown for 28 patients. Differences between pre- and on- or post-treatment were analyzed with the repeated measures ANOVA with a post hoc Dunnett’s multiple comparisons test. Differences were considered significant when p < 0.05, as indicated with an asterisk (*p < 0.05, **p < 0.01)
Table 1.
Characteristics and survival distribution of treatment-induced T cell activation and differentiation parameters
Immune parameter | Mean treatment-induced increase (range)a | Cut-offb | Median Survival between groupsc (# of patients in each group) | p value§ | Hazard ratio (95 % CI HR) |
---|---|---|---|---|---|
Absolute lymphocytes/μl blood | 51 % (0–155) | 25 % | 41.0 vs. 20.5 (21 vs. 6) | 0.015 | 0.181 (0.046–0.720) |
Non-naïve CD8+ cells | 23.6 % (−7 to +88) | 30 % | 41.0 vs. 29.0 (7 vs. 21) | 0.472 | 0.717 (0.230–1.94) |
Non-naïve CD4+ cells | 32 % (2–104) | 30 % | 41.0 vs. 20.0 (12 vs. 16) | 0.036 | 0.402 (0.147–0.940) |
CD8+ HLA-DR+ | 80 % (−50 to +470) | 100 % | 32.5 vs. 31.5 (6 vs. 22) | 0.960 | 0.972 (0.35–2.7) |
CD4+ HLA-DR+ | 136 % (10–480) | 140 % | 35.0 vs. 27.5 (12 vs. 16) | 0.467 | 0.721 (0.30–1.75) |
CD8+ ICOS+ | 110 % (10–310) | 100 % (and sustained)d | 21.0 vs. 57.0 (7 vs. 11) | 0.043 | 2.99 (1.05–20.1) |
CD4+ ICOS+ | 140 % (50–280) | 100 % (and sustained)d | 34.0 vs. 37.0 (5 vs. 13) | 0.894 | 1.08 (0.348–3.47) |
CD4+ CD25intFoxP3+ | 73.6 % (0–356) | 50 % | 41.0 vs. 21.0 (13 vs. 11) | 0.030 | 0.37 (0.13–0.90) |
CD8+ PD-1+ | 64.5 % (−32 to +464) | 40 % | 20.0 vs. 36.0 (8 vs. 16) | 0.109 | 2.07 (0.81–7.63) |
CD4+ PD-1+ | 90.9 % (−40 to +242) | 40 % | 24.0 vs. 52.0 (17 vs. 7) | 0.464 | 1.45 (0.54–3.90) |
CD8+ CTLA-4+ | 135 % (−72 to +1,395) | 100 % | 24.0 vs. 36.0 (9 vs. 15) | 0.530 | 1.347 (0.565–3.77) |
CD4+ CTLA-4+ (total T cells) | 76.7 % (−6 to +280) | 40 % | 31.5 vs. 28.0 (8 vs. 16) | 0.222 | 0.569 (0.195–1.46) |
CD4+ CD25hiFoxP3 + Tregs | 20.9 % (−30 to +140) | 50 % | 37.0 vs. 21.0 (5 vs. 17) | 0.045 | 2.63 (1.03–19.5) |
aMean and range of treatment-induced increases are given in percentage positive CD4+ or CD8+ T cells
bCut-off points for survival prediction were determined using the Cox regression model and are given as percentage positive cells
c Median survival was calculated using the Kaplan–Meier method and is given in months for groups above and below designated cut-offs
dIncreased and sustained ICOS expression in T cells was defined as >twofold increase in ICOShi T cells over baseline that was sustained over 12 weeks after therapy
§Statistical significance of the survival distribution was analyzed by log-rank testing and considered significant when p < 0.05 (in bold)
T cell activation in relation to treatment response and survival
To assess treatment-induced T cell activation, circulating CD4+ and CD8+ T cells were analyzed for the expression of HLA-DR, ICOS, FoxP3, CTLA-4, and PD-1 over the course of treatment. As shown in Figure S2 and Table 1, Prostate GVAX/ipilimumab treatment resulted in generalized CD4+ and CD8+ T cell activation. Expression of the activation markers increased with escalating doses of ipilimumab (data not shown). As we recently reported, early up-regulation of HLA-DR surface expression during treatment (at week 4, i.e., after one ipilimumab infusion) was more pronounced in patients with PR/SD versus patients with PD [20]. Yet, this early up-regulation did not correlate with survival (Table 1 and [20]). In fact, of all the aforementioned activation markers, only up-regulation of FoxP3 in activated, non-regulatory CD4+ T cells (defined by intermediate CD25 expression levels) was related to improved OS (median survival, 41 vs. 21 months, HR = 0.37, 95 % CI = 0.13–0.90, p = 0.03; see also Table 1).
Treatment-induced increase in Treg frequency is negatively associated with OS
Treg frequencies gradually increased over the course of treatment in patients with SD or PD, but not in patients with PR (Fig. 2a). Increases of more than 50 % were associated with a shorter OS following treatment (median survival, 21 vs. 37 months, HR = 2.63, 95 % CI = 1.03–19.5, p = 0.045; see also Table 1 and Fig. 2b). Previous data from (pre)clinical studies on CTLA-4 blockade have shown that increases in the Teff/Treg ratio in both blood and tumors were associated with clinical responses [31, 32]. As shown in Table 1 and Fig 2c, we also observed increases in the CD4CD45RO Teff/Treg ratio, and increases of more than 50 % were associated with prolonged OS following treatment (median survival, 57 vs. 21 months, HR = 0.294, 95 % CI = 0.109–0.794, p = 0.0157).
Fig. 2.
Increased Treg frequencies following Prostate GVAX/ipilimumab therapy are associated with reduced survival. The number of circulating CD4+CD25hiFoxP3+ regulatory T cells was determined before (w0), during (w4, w8, w12, w16, w20, and w24), and after (fu) Prostate GVAX/ipilimumab treatment by flow cytometric analysis on isolated PBMC. a Individual percentages (black lines) and means with range (open box plot) of circulating CD4+CD25hiFoxP3+ cells are given for patients that experienced partial PSA response (PR; left panel), disease stabilization (SD; middle panel), or disease progression (PD; right panel) during/after Prostate GVAX/ipilimumab treatment. b Kaplan–Meier curve for increases in CD4+CD25hiFoxP3+ Tregs. The number of patients and the corresponding median survival for each group are given. c Kaplan–Meier curve for increases in Teff/Treg ratio. Teff/Treg ratio was determined by dividing frequency of CD4+CD45RO+ T cells by the CD4+CD25highFoxP3+ Treg frequency. The number of patients and the corresponding median survival for each group are given. Differences between pre- and on- or post-treatment were analyzed with the repeated measures ANOVA with a post hoc Dunnett’s multiple comparisons test. Differences were considered significant when p < 0.05, as indicated with an asterisk (*p < 0.05, **p < 0.01)
Preexisting T cell characteristics in relation to survival
In order to identify biomarkers for patient selection, we correlated pre-treatment frequencies of non-naive CD4+ and CD8+ cells, and of ICOS+, HLA-DR+, FoxP3+, PD-1+, or CTLA-4+ T cells and Tregs with OS after Prostate GVAX/ipilimumab treatment. An overview of significant and near-significant associations is listed in Table 2.
Table 2.
Characteristics and survival distribution of pre-treatment T cell activation and differentiation parameters
Immune parameter | Mean pre-treatment frequencies (range)a | Cut-offb | Median survival between groupsc (# of patients in each group) | p value§ | Hazard ratio (95 % CI HR) |
---|---|---|---|---|---|
Non-naïve CD8+ cells | 67.8 % (26–94) | 76.0 % | Not reached vs. 20.5 (10 vs. 18) | 0.028 | 0.334 (0.135–0.892) |
Non-naïve CD4+ cells | 63.6 % (31.2–89.1) | 66.0 % | 19.0 vs. 41.0 (11 vs. 17) | 0.02 | 2.666 (1.21–9.09) |
CD8+PD-1+ | 8.1 % (2.5–17.8) | 5.0 % | Not reached vs. 24.0 (19 vs. 5) | 0.096 | 3.19 (0.86–6.4) |
CD4+PD-1+ | 5.8 % (1.9–14.8) | 5.0 % | 41.0 vs. 18.0 (12 vs. 12) | 0.014 | 0.336 (0.103–0.772) |
CD8+CTLA-4+ | 3.0 % (0.4–12.5) | 1.5 % | 38.5 vs. 21.0 (16 vs. 9) | 0.358 | 0.646 (0.222–1.723) |
CD4+CTLA-4+ (conv. T cells) | 3.4 % (0–12.0) | 2.4 % | 52.0 vs. 20.5 (13 vs. 12) | 0.011 | 0.343 (0.083–0.725) |
CD4+CD25hiFoxP3+ Tregs | 5.5 % (3.1–7.7) | 6.3 % | 20.0 vs. 36.0 (6 vs. 18) | 0.087 | 2.26 (0.86–9.86) |
aMean and range of pre-treatment frequencies are given in percentage positive CD4+ or CD8+ T cells
bCut-off points for survival prediction were determined using the Cox regression model and are given as percentage positive cells
cMedian Survival was calculated using the Kaplan–Meier method and is given in months for groups above and below designated cut-offs
§Statistical significance of the survival distribution was analyzed by log-rank testing and considered significant when p < 0.05 (in bold)
High pre-treatment frequencies of non-naive CD8+ cells or CD4+PD-1+ cells were associated with a significantly prolonged OS (median survival, not reached vs. 20.5 months, HR = 0.334, 95 % CI = 0.135–0.892, p = 0.028 for non-naïve CD8+ cells; and median survival, 41 vs. 18 months, HR = 0.336, 95 % CI = 0.103–0.772, p = 0.014 for CD4+PD-1+ cells; see also Fig. 3a, b). In contrast, high pre-treatment frequencies of non-naive CD4+ cells or Tregs were associated with shorter OS (median survival, 19 vs. 41 months, HR = 2.66, 95 % CI = 1.21–9.09, p = 0.020 for non-naive CD4+ cells; Fig. 3c and d).
Fig. 3.
High pre-treatment frequencies of CD8+ Teff/Tcm/Tem, CD4+PD-1+, or CD4+CTLA4+ T cells, and low pre-treatment frequencies of CD4+ Teff/Tem and Tregs are predictive parameters for survival benefit after Prostate GVAX/ipilimumab therapy. Pre-treatment frequencies of circulating CD4+ and CD8+ effector/memory, CD4+PD-1+, CD4+CD25hiFoxP3+, or CD4+CTLA-4+ T cells were determined by flow cytometry. Kaplan–Meier curves for pre-treatment frequencies of a CD8+ effector/memory T cells, b CD4+PD-1+, c CD4+CD25hiFoxP3+, and d CD4+ effector/memory cells. The number of patients and the corresponding median survival for each group are given. e Percentage of CTLA-4-expressing cells within total and conventional CD4+ T cells (i.e., minus Tregs) in patients (black bars) and age- and sex-matched healthy volunteers (open bars) before treatment. f Kaplan–Meier curve for pre-treatment frequencies of CD4+CTLA-4+ conventional T cells. The number of patients and the corresponding median survival for each group are given
Significantly higher levels of intracellular CTLA-4+ were detected in CD4+ T cells of prostate cancer patients compared with age- and sex-matched healthy volunteers (Fig. 3e). Interestingly, CTLA-4 could be detected in both total CD4+ and conventional CD4+ T cells (i.e., total CD4+ T cells minus Tregs) of prostate cancer patients, whereas CTLA-4 was virtually undetectable in conventional CD4+ T cells of healthy donors (Fig. 3e). Patients who displayed these cancer-related high levels of conventional CD4+CTLA-4+ cells before treatment had a significantly longer OS than patients who did not (median survival, 52 vs. 20.5 months, HR = 0.343, 95 % CI = 0.083–0.725, p = 0.011; Fig. 3f). Of note, this positive correlation with survival was also observed for total CD4+CTLA-4+ T cells (see Fig. S3B).
CD4+CTLA-4+ T cell frequency as a dominant predictor of OS
To assess whether clinical prognosis impacted the predictive value for treatment outcome of any of the identified response/survival parameters, the median Halabi predicted survival (HPS) was determined for the patient groups above and below the designated cut-offs [30]. No significant differences were observed, indicating that better prognosis before treatment was not the determining factor for any of these parameters (data not shown).
Unsupervised clustering of all pre-treatment and treatment-induced T cell biomarkers with predictive value for survival on treatment revealed two major clusters of patients, designated group 1 and group 2 (Fig. 4a). There was a clear trend for higher OS of patients in group 1 (Fig. 4b). Interestingly, all except two patients from group 1 exhibited the cancer-related expression of CTLA-4+ in CD4+ T cells, and these patients (designated clustered group 3, highlighted in Fig. 4a) experienced significantly prolonged OS after therapy (median survival, 46.5 vs. 21 months, HR = 0.271, 95 % CI = 0.0791–0.931, p = 0.036; Fig. 4c). Thus, unsupervised clustering analysis demonstrated the cancer-related CTLA-4 expression in CD4+ T cells to be a dominant positive predictor for survival after Prostate GVAX/ipilimumab therapy.
Fig. 4.
Cancer-related expression of CTLA-4+ in CD4+ T cells is the dominant predictor for survival after Prostate GVAX/ipilimumab therapy. a Unsupervised cluster analysis of the expression of the treatment-induced and pre-treatment T cell markers. To identify clusters of correlated markers, hierarchical cluster analysis using TIGR software was performed, and average linkage analysis was done by Pearson correlation analysis. Values of the treatment-induced and pre-treatment parameters are given relative to the cut-off value (determined by Cox regression model as described in “Materials and methods”): below cut-off in green and above cut-off in red. Kaplan–Meier curve for b group 1 versus group 2 and c group 3 (with high cancer-related CD4+CTLA-4+ T cell rates: highlighted by white box in the heat plot) versus group 2 (with generally low CD4+CTLA-4+ T cell rates). The number of patients and the corresponding median survival for each group are given
Discussion
Phenotypic T cell profiling of patients with CRPC receiving Prostate GVAX and ipilimumab revealed that in particular CD4+ T cell differentiation and activation were associated with clinical benefit. A pre-treatment profile of low frequencies of differentiated CD4+ T cells and of Tregs, but high frequencies of activated CTLA-4+ or PD-1+ Th cells, had predictive power for OS benefit on treatment. Importantly, unsupervised cluster analysis of these markers revealed cancer-related CTLA-4 expression in conventional CD4+ T cells as the dominant predictor for survival after Prostate GVAX/ipilimumab therapy. As this was only a small exploratory study of 28 patients, no multivariate analyses were performed. Moreover, the predictive value of the identified parameters should be validated prospectively in large randomized trials.
It has been reported recently that an ALC >1,000/μl blood after two ipilimumab infusions is strongly predictive of clinical benefit and OS in melanoma patients [33]. As most of our patients already exhibited ALC levels >1,000/μl blood prior to therapy, no such relation could be found in our study. Nevertheless, on-treatment increases of more than 25 % over baseline ALC levels were associated with prolonged OS, indicating that changes in ALC may be predictive of response to prostate GVAX/ipilimumab. These data complement the findings by Berman et al. of an association between ALC change and clinical activity in ipilimumab-treated melanoma patients [34].
In keeping with previous findings by others for anti-CTLA-4 treatment [12, 35–38], Prostate GVAX/ipilimumab immunotherapy also resulted in increases in circulating HLA-DR- and CD45RO-expressing CD4+ and CD8+ T cells. Moreover, and in contrast to the aforementioned publications, the magnitude of early HLA-DR up-regulation correlated with on-treatment PSA declines and stabilizations [20], and treatment-induced increases in CD4+CD45RO+ memory T cell rates were associated with improved OS. Interestingly, patients who did not show increases in non-naive CD4+ T cells already exhibited high pre-treatment levels (not shown). This observation suggests that the presence of a large pool of pre-treatment non-naive CD4+ cells, primed and differentiated under cancer-associated immune suppressive conditions, precludes the on-treatment de novo priming of a sizeable pool of CD4+ T helper cells with more favorable effector characteristics and/or antitumor specificity. This hypothesis is further supported by co-clustering of pre-treatment Treg frequencies with non-naive CD4+ rates in an unsupervised analysis (see Fig. 4a).
Sustained increases in the frequency of CD4+ICOS+ cells (defined as >twofold increase in CD4+ICOShi cells over baseline that was sustained over 12 weeks after therapy) have been described to serve as a biomarker of anti-CTLA-4 activity and/or of clinical benefit in melanoma patients [39]. No such relation with clinical outcome was observed in this study. This may be explained by lower doses of antibody administered, resulting in lower ICOS increases in CD4+ T cells—on average 2.4-fold, versus the previously reported five- to tenfold [39]. As a result, only few patients adhered to the predefined (persistent) twofold increase in CD4+ICOS+ cells over baseline in our study. In addition, we cannot rule out that the Prostate GVAX vaccine also may play a role in the observed differences.
nTregs have been described to constitutively express CTLA-4 [40, 41], thereby making them potential targets for CTLA-4 blockade therapy. Data on the effects of CTLA-4 antibody therapy on nTreg frequency and function are not consistent, with groups claiming increases [42], early decreases [43], or variable results [31] in nTreg frequencies and/or function after CTLA-4 blockade therapy. Furthermore, multiple vaccinations with a GM-CSF-producing tumor cell vaccine have also been shown to induce Treg expansion in mice [44]. Our data demonstrated gradual increases in Treg frequencies following multiple Prostate GVAX/ipilimumab doses in a subgroup of patients, suggesting that Prostate GVAX/ipilimumab treatment induces expansion rather than depletion of nTregs. Moreover, these treatment-induced increases in Treg frequencies were associated with reduced OS, in keeping with the notion that indeed these cells play an important role in dampening antitumor immune responses. Nevertheless, increases in T cell activation were more profound than increases in nTreg frequencies, resulting in an increased Teff/Treg ratio. In keeping with previous findings, this increased Teff/Treg ratio was associated with clinical benefit [31, 42].
Although FoxP3 expression is generally associated with natural Tregs (nTregs), it has also been described to be transiently upregulated in activated effector T cells, and importantly, this up-regulation has not been associated with suppressive function [26–28]. In our study, FoxP3 was significantly upregulated in CD4+CD25int (activated, effector-like) cells, but not in CD4+CD25hi nTregs, which was consistent with the observed association with prolonged survival. Similarly, on-treatment increases in CTLA-4 and PD-1 were observed in CD4+ T cells, confirming previously reported findings for CTLA-4 [38]. Neither of these were associated with clinical benefit. Co-clustering of pre-treatment CD4+PD-1+ and CD4+CTLA4+ T cell frequencies (Fig. 4a) suggests coordinated activation of a co-inhibitory program in CRPC. This notion, combined with our observation that the majority of CTLA-4 and PD-1 is expressed in different subpopulations of CD4+ T cells (not shown) and that PD-1 is upregulated upon treatment, supports the rationale for combinatorial co-inhibitory (CTLA-4 and PD-1) blockade therapy.
Due to IRAE risk, there is a widely recognized and urgent need for biomarkers to predict clinical benefit from CTLA-4 blockade therapy. Our findings suggest that cancer-related high pre-treatment CD4+CTLA-4+ T cell frequencies may provide such a biomarker, enabling the identification of “ipilimumab-sensitive” patients. To our knowledge, this is the first time that such a pre-treatment marker has been described. Previous studies by Zhou et al. [38] also demonstrated higher CTLA-4 levels on CD25lo/neg cells compared with healthy volunteers; however, so far no association for this biomarker with prognosis or treatment outcome has been reported. Our observation of a relation to prolonged survival for frequencies of CTLA4+CD4+ T cells, both with or without inclusion of Treg frequencies (see Fig. 3f, Fig. S3B), is in line with a previous report by Peggs et al. [45], showing that, when combined with a GVAX vaccine in the B16 melanoma model, anti-CTLA4 modulation of conventional Teff cells had a more profound effect on tumor growth control than modulation of Tregs, but that modulation of both was even more effective.
In summary, our profiling data are consistent with the clinical benefit and survival advantage for patients with high levels of effector T cell activation and differentiation and low Treg levels. Moreover, they suggest that cancer-related expression of CTLA-4 in Th cells may provide a simple, robust, and much-needed tool for patient selection for GVAX and/or ipilimumab treatment. It is tempting to hypothesize that the survival advantage for patients with CD4+CTLA-4+ T cells is due to the specific use of antibodies against CTLA-4, yet, it may also be a mere reflection of a more activated T cell state, leading to better outcome in CRPC patients regardless of the applied (immuno)therapy. As Prostate GVAX is not readily available at this time, the relevance of this biomarker should therefore first be validated in a larger group of mCRPC patients receiving ipilimumab monotherapy in the context of randomized phase III trials. This potential biomarker for patient selection may also be of interest for other tumor types, especially after the recent FDA approval of ipilimumab (Yervoy) for the treatment of late-stage unresectable or metastatic melanoma.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Acknowledgments
This research was financially supported by awards and grants from the Prostate Cancer Foundation (PCF to T.D.G.), Stichting VUmc-CCA, and the Dutch Cancer Society (KFW; VU 2006-3697). The authors thank Dr. S.A.G.M. Cillessen for her assistance with the unsupervised clustering analysis.
Conflict of interest
J. M. C. is a Bristol-Myers Squibb employee, and J. M. C and I. L. own stock and/or stock options from Bristol-Myers Squibb. A.J.M.v.d.E. and W.R.G. have served as consultants and received honoraria from Bristol-Myers Squibb. T.D.G and W.R.G. received an educational grant from Cell Genesys Inc. All other authors declare that they have no conflict of interest.
Abbreviations
- CTLA-4
CTL antigen-4
- CRCP
Castration-resistant prostate cancer
- HPS
Halabi predicted survival
- IRAE
Immune-related adverse events
- OS
Overall survival
- PD-1
Programmed death-1
- PD
Progressive disease
- PR
Partial response
- SD
Stable disease
- Tregs
Regulatory T cells
Contributor Information
Saskia J. A. M. Santegoets, Email: s.santegoets@vumc.nl
Tanja D. de Gruijl, Email: td.degruijl@vumc.nl
References
- 1.Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;60:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
- 2.Drake CG. Prostate cancer as a model for tumour immunotherapy. Nat Rev Immunol. 2010;10:580–593. doi: 10.1038/nri2817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, Dahut WL, Arlen PM, Gulley JL, Godfrey WR. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol. 2010;28:1099–1105. doi: 10.1200/JCO.2009.25.0597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 2010;363:411–422. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
- 5.Tannock IF, De WR, Berry WR, Horti J, Pluzanska A, Chi KN, Oudard S, Theodore C, James ND, Turesson I, Rosenthal MA, Eisenberger MA. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351:1502–1512. doi: 10.1056/NEJMoa040720. [DOI] [PubMed] [Google Scholar]
- 6.Vishnu P, Tan WW. Update on options for treatment of metastatic castration-resistant prostate cancer. Onco Targets Ther. 2010;3:39–51. doi: 10.2147/ott.s5818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Greenwald RJ, Freeman GJ, Sharpe AH. The B7 family revisited. Annu Rev Immunol. 2005;23:515–548. doi: 10.1146/annurev.immunol.23.021704.115611. [DOI] [PubMed] [Google Scholar]
- 8.Weber J. Immune checkpoint proteins: a new therapeutic paradigm for cancer–preclinical background: CTLA-4 and PD-1 blockade. Semin Oncol. 2010;37:430–439. doi: 10.1053/j.seminoncol.2010.09.005. [DOI] [PubMed] [Google Scholar]
- 9.Korman AJ, Peggs KS, Allison JP. Checkpoint blockade in cancer immunotherapy. Adv Immunol. 2006;90:297–339. doi: 10.1016/S0065-2776(06)90008-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Peggs KS, Quezada SA, Allison JP. Cell intrinsic mechanisms of T-cell inhibition and application to cancer therapy. Immunol Rev. 2008;224:141–165. doi: 10.1111/j.1600-065X.2008.00649.x. [DOI] [PubMed] [Google Scholar]
- 11.Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbe C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711–723. doi: 10.1056/NEJMoa1003466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Small EJ, Tchekmedyian NS, Rini BI, Fong L, Lowy I, Allison JP. A pilot trial of CTLA-4 blockade with human anti-CTLA-4 in patients with hormone-refractory prostate cancer. Clin Cancer Res. 2007;13:1810–1815. doi: 10.1158/1078-0432.CCR-06-2318. [DOI] [PubMed] [Google Scholar]
- 13.Yang JC, Hughes M, Kammula U, Royal R, Sherry RM, Topalian SL, Suri KB, Levy C, Allen T, Mavroukakis S, Lowy I, White DE, Rosenberg SA. Ipilimumab (anti-CTLA4 antibody) causes regression of metastatic renal cell cancer associated with enteritis and hypophysitis. J Immunother. 2007;30:825–830. doi: 10.1097/CJI.0b013e318156e47e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Fong L, Kwek SS, O’Brien S, Kavanagh B, McNeel DG, Weinberg V, Lin AM, Rosenberg J, Ryan CJ, Rini BI, Small EJ. Potentiating endogenous antitumor immunity to prostate cancer through combination immunotherapy with CTLA4 blockade and GM-CSF. Cancer Res. 2009;69:609–615. doi: 10.1158/0008-5472.CAN-08-3529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, Davis T, Henry-Spires R, Macrae S, Willman A, Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman A, Allison JP, Dranoff G. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci USA. 2003;100:4712–4717. doi: 10.1073/pnas.0830997100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hurwitz AA, Yu TF, Leach DR, Allison JP. CTLA-4 blockade synergizes with tumor-derived granulocyte-macrophage colony-stimulating factor for treatment of an experimental mammary carcinoma. Proc Natl Acad Sci USA. 1998;95:10067–10071. doi: 10.1073/pnas.95.17.10067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Weber J. Ipilimumab: controversies in its development, utility and autoimmune adverse events. Cancer Immunol Immunother. 2009;58:823–830. doi: 10.1007/s00262-008-0653-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bouwhuis MG, Ten Hagen TL, Suciu S, Eggermont AM. Autoimmunity and treatment outcome in melanoma. Curr Opin Oncol. 2011;23:170–176. doi: 10.1097/CCO.0b013e328341edff. [DOI] [PubMed] [Google Scholar]
- 19.Weber J, Thompson JA, Hamid O, Minor D, Amin A, Ron I, Ridolfi R, Assi H, Maraveyas A, Berman D, Siegel J, O’Day SJ. A randomized, double-blind, placebo-controlled, phase II study comparing the tolerability and efficacy of ipilimumab administered with or without prophylactic budesonide in patients with unresectable stage III or IV melanoma. Clin Cancer Res. 2009;15:5591–5598. doi: 10.1158/1078-0432.CCR-09-1024. [DOI] [PubMed] [Google Scholar]
- 20.van den Eertwegh AJ, Versluis J, van den Berg HP, Santegoets SJ, van Moorselaar RJ, van der Sluis TM, Gall HE, Harding TC, Jooss K, Lowy I, Pinedo HM, Scheper RJ, Stam AG, von Blomberg BM, de Gruijl TD, Hege K, Sacks N, Gerritsen WR. Combined immunotherapy with granulocyte-macrophage colony-stimulating factor-transduced allogeneic prostate cancer cells and ipilimumab in patients with metastatic castration-resistant prostate cancer: a phase 1 dose-escalation trial. Lancet Oncol. 2012;13:509–517. doi: 10.1016/S1470-2045(12)70007-4. [DOI] [PubMed] [Google Scholar]
- 21.Simons JW, Carducci MA, Mikhak B, Lim M, Biedrzycki B, Borellini F, Clift SM, Hege KM, Ando DG, Piantadosi S, Mulligan R, Nelson WG. Phase I/II trial of an allogeneic cellular immunotherapy in hormone-naive prostate cancer. Clin Cancer Res. 2006;12:3394–3401. doi: 10.1158/1078-0432.CCR-06-0145. [DOI] [PubMed] [Google Scholar]
- 22.Small EJ, Sacks N, Nemunaitis J, Urba WJ, Dula E, Centeno AS, Nelson WG, Ando D, Howard C, Borellini F, Nguyen M, Hege K, Simons JW. Granulocyte macrophage colony-stimulating factor–secreting allogeneic cellular immunotherapy for hormone-refractory prostate cancer. Clin Cancer Res. 2007;13:3883–3891. doi: 10.1158/1078-0432.CCR-06-2937. [DOI] [PubMed] [Google Scholar]
- 23.Scher HI, Halabi S, Tannock I, Morris M, Sternberg CN, Carducci MA, Eisenberger MA, Higano C, Bubley GJ, Dreicer R, Petrylak D, Kantoff P, Basch E, Kelly WK, Figg WD, Small EJ, Beer TM, Wilding G, Martin A, Hussain M. Design and end points of clinical trials for patients with progressive prostate cancer and castrate levels of testosterone: recommendations of the Prostate Cancer Clinical Trials Working Group. J Clin Oncol. 2008;26:1148–1159. doi: 10.1200/JCO.2007.12.4487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Molling JW, Langius JA, Langendijk JA, Leemans CR, Bontkes HJ, von Blomberg BM, Scheper RJ, van den Eertwegh AJ. Low levels of circulating invariant natural killer T cells predict poor clinical outcome in patients with head and neck squamous cell carcinoma. J Clin Oncol. 2007;25:862–868. doi: 10.1200/JCO.2006.08.5787. [DOI] [PubMed] [Google Scholar]
- 25.Sallusto F, Lenig D, Forster R, Lipp M, Lanzavecchia A. Two subsets of memory T lymphocytes with distinct homing potentials and effector functions. Nature. 1999;401:708–712. doi: 10.1038/44385. [DOI] [PubMed] [Google Scholar]
- 26.Allan SE, Crome SQ, Crellin NK, Passerini L, Steiner TS, Bacchetta R, Roncarolo MG, Levings MK. Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production. Int Immunol. 2007;19:345–354. doi: 10.1093/intimm/dxm014. [DOI] [PubMed] [Google Scholar]
- 27.Wang J, Ioan-Facsinay A, van d V, Huizinga TW, Toes RE. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol. 2007;37:129–138. doi: 10.1002/eji.200636435. [DOI] [PubMed] [Google Scholar]
- 28.Gavin MA, Torgerson TR, Houston E, DeRoos P, Ho WY, Stray-Pedersen A, Ocheltree EL, Greenberg PD, Ochs HD, Rudensky AY. Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc Natl Acad Sci USA. 2006;103:6659–6664. doi: 10.1073/pnas.0509484103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Molling JW, de Gruijl TD, Glim J, Moreno M, Rozendaal L, Meijer CJ, van d V, van den Eertwegh AJ, Scheper RJ, von Blomberg ME, Bontkes HJ. CD4(+)CD25hi regulatory T-cell frequency correlates with persistence of human papillomavirus type 16 and T helper cell responses in patients with cervical intraepithelial neoplasia. Int J Cancer. 2007;121:1749–1755. doi: 10.1002/ijc.22894. [DOI] [PubMed] [Google Scholar]
- 30.Halabi S, Small EJ, Kantoff PW, Kattan MW, Kaplan EB, Dawson NA, Levine EG, Blumenstein BA, Vogelzang NJ. Prognostic model for predicting survival in men with hormone-refractory metastatic prostate cancer. J Clin Oncol. 2003;21:1232–1237. doi: 10.1200/JCO.2003.06.100. [DOI] [PubMed] [Google Scholar]
- 31.Liakou CI, Kamat A, Tang DN, Chen H, Sun J, Troncoso P, Logothetis C, Sharma P. CTLA-4 blockade increases IFNgamma-producing CD4 + ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc Natl Acad Sci USA. 2008;105:14987–14992. doi: 10.1073/pnas.0806075105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest. 2006;116:1935–1945. doi: 10.1172/JCI27745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ku GY, Yuan J, Page DB, Schroeder SE, Panageas KS, Carvajal RD, Chapman PB, Schwartz GK, Allison JP, Wolchok JD. Single-institution experience with ipilimumab in advanced melanoma patients in the compassionate use setting: lymphocyte count after 2 doses correlates with survival. Cancer. 2010;116:1767–1775. doi: 10.1002/cncr.24951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Berman DM, Wolchok J, Weber J, Hamid O, O’Day S, Chasalow SD. Association of peripheral blood absolute lymphocyte count (ALC) and clinical activity in patients (pts) with advanced melanoma treated with ipilimumab. ASCO Meeting Abstract J Clin Oncol. 2009;27:15s. [Google Scholar]
- 35.Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, Restifo NP, Haworth LR, Seipp CA, Freezer LJ, Morton KE, Mavroukakis SA, Duray PH, Steinberg SM, Allison JP, Davis TA, Rosenberg SA. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci USA. 2003;100:8372–8377. doi: 10.1073/pnas.1533209100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Attia P, Phan GQ, Maker AV, Robinson MR, Quezado MM, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Restifo NP, Haworth LR, Levy C, Mavroukakis SA, Nichol G, Yellin MJ, Rosenberg SA. Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J Clin Oncol. 2005;23:6043–6053. doi: 10.1200/JCO.2005.06.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Maker AV, Phan GQ, Attia P, Yang JC, Sherry RM, Topalian SL, Kammula US, Royal RE, Haworth LR, Levy C, Kleiner D, Mavroukakis SA, Yellin M, Rosenberg SA. Tumor regression and autoimmunity in patients treated with cytotoxic T lymphocyte-associated antigen 4 blockade and interleukin 2: a phase I/II study. Ann Surg Oncol. 2005;12:1005–1016. doi: 10.1245/ASO.2005.03.536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhou J, Bashey A, Zhong R, Corringham S, Messer K, Pu M, Ma W, Soiffer R, Mitrovich RC, Lowy I, Ball ED. CTLA-4 blockade following relapse of malignancy after allogeneic stem cell transplantation is associated with T cell activation but not increased levels of T regulatory cells. Biol Blood Marrow Transplant. 2011;17:682–692. doi: 10.1016/j.bbmt.2010.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Carthon BC, Wolchok JD, Yuan J, Kamat A, Ng Tang DS, Sun J, Ku G, Troncoso P, Logothetis CJ, Allison JP, Sharma P. Preoperative CTLA-4 blockade: tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin Cancer Res. 2010;16:2861–2871. doi: 10.1158/1078-0432.CCR-10-0569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Takahashi T, Tagami T, Yamazaki S, Uede T, Shimizu J, Sakaguchi N, Mak TW, Sakaguchi S. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med. 2000;192:303–310. doi: 10.1084/jem.192.2.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. J Exp Med. 2000;192:295–302. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kavanagh B, O’Brien S, Lee D, Hou Y, Weinberg V, Rini B, Allison JP, Small EJ, Fong L. CTLA4 blockade expands FoxP3 + regulatory and activated effector CD4 + T cells in a dose-dependent fashion. Blood. 2008;112:1175–1183. doi: 10.1182/blood-2007-11-125435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.O’Mahony D, Morris JC, Quinn C, Gao W, Wilson WH, Gause B, Pittaluga S, Neelapu S, Brown M, Fleisher TA, Gulley JL, Schlom J, Nussenblatt R, Albert P, Davis TA, Lowy I, Petrus M, Waldmann TA, Janik JE. A pilot study of CTLA-4 blockade after cancer vaccine failure in patients with advanced malignancy. Clin Cancer Res. 2007;13:958–964. doi: 10.1158/1078-0432.CCR-06-1974. [DOI] [PubMed] [Google Scholar]
- 44.Lacelle MG, Jensen SM, Fox BA. Partial CD4 depletion reduces regulatory T cells induced by multiple vaccinations and restores therapeutic efficacy. Clin Cancer Res. 2009;15:6881–6890. doi: 10.1158/1078-0432.CCR-09-1113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Peggs KS, Quezada SA, Chambers CA, Korman AJ, Allison JP. Blockade of CTLA-4 on both effector and regulatory T cell compartments contributes to the antitumor activity of anti-CTLA-4 antibodies. J Exp Med. 2009;206:1717–1725. doi: 10.1084/jem.20082492. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.