Purpose:

Tumors with high mutation load are thought to engender stronger immune responses, which in turn promote prolonged patient survival. To investigate this, we assessed tumor-infiltrating lymphocytes (TILs) and immunosuppressive factors across the 4 molecular subtypes of endometrial cancer, which have characteristic mutation rates ranging from low to ultra-high.

Experimental Design:

A total of 460 endometrial cancers were stratified by ProMisE (Proactive Molecular Risk Classifier in Endometrial cancer) into 4 molecular subtypes: mismatch repair-deficient (MMRd), POLE mutant (POLE), p53 abnormal (p53abn), and p53 wild-type (p53wt). Immune markers (CD3, CD8, CD79a, CD138, PD-1, PD-L1, FoxP3, IDO-1) were quantified by multiplex IHC and tested for associations with ProMisE subtype, survival, and other clinicopathologic parameters.

Results:

Two major TIL patterns were observed. TILhigh tumors harbored dense T- and B-lineage infiltrates and multiple immunosuppressive features and were common in molecular subtypes associated with high mutation load (MMRd and POLE); however, equally strong responses were seen in significant numbers of p53abn and p53wt tumors, which have characteristically low mutation loads. TILlow tumors were generally devoid of immunologic features and were more prevalent in p53abn and p53wt endometrial cancers, yet were also seen in MMRd and POLE subtypes. In multivariable models involving ProMisE subtype, T-cell markers, and TIL clusters, only ProMisE showed independent prognostic significance.

Conclusions:

Immune response correlates with endometrial cancer molecular subtype but does not carry independent prognostic significance. Profound variation in immune response is seen across and within endometrial cancer molecular subtypes, suggesting that assessment of immune response rather than molecular subtype may better predict response to immunotherapy.

See related commentary by Mullen and Mutch, p. 2366

Translational Relevance

We have entered a new era in endometrial carcinoma research and clinical care where pragmatic molecular classification tools enable consistent categorization/stratification of endometrial cancers and provide prognostic information to inform management. However, the ability of molecular classification to predict response to immunotherapy has not been defined, nor has the impact of immune environment on outcomes within this new framework of molecular subtypes. Herein, we characterize the tumor-infiltrating lymphocytes and immunosuppressive factors across the 4 major molecular subtypes of endometrial cancer, demonstrating diversity even among characteristically “high” and “low” mutation load tumors. Molecular subtype is demonstrated to be the parameter most strongly associated with clinical outcomes (more than immune response); however, assessment of immune response (rather than molecular subtype) may better predict response to immunotherapy.

The recent success of immunotherapy has prompted a need to better understand the features of cancers that impart susceptibility to immune recognition and control. Among the best evidence for immunologic control is the phenomenon of tumor-infiltrating lymphocytes (TILs), which are strongly associated with patient survival in almost all human cancers (1–4). Moreover, the presence of TIL is associated, albeit imperfectly, with clinical response to immunotherapies targeting the CTLA-4 and PD-1 pathways, referred to as “checkpoint blockade” (5–7). TIL responses generally show a positive association with mutation load, presumably due to an increased number of mutated antigens (neoantigens) available for T-cell recognition (8, 9). In addition, DNA damage can promote TIL responses by activating mechanisms such as the cGAS/STING pathway (10), leading to an immunologically permissive tumor microenvironment. Conversely, TIL responses can be inhibited by oncogenic signaling pathways, including the Wnt/β-catenin and PI3K signaling pathways (11–14). For a given patient, the magnitude of the TIL response likely reflects a balance of these factors, which in turn strongly influences the patient's likelihood of survival and response to checkpoint blockade. A better understanding of the interplay between these factors is required for the development of cancer immunotherapies that benefit a broader spectrum of patients.

Endometrial cancer provides a unique opportunity to study the influence of the tumor genome on antitumor immunity while also presenting an urgent need for improved therapies. Endometrial cancer is the most common gynecologic malignancy in North America, and the fourth most common cancer in women overall, with 150,000 new cases in Europe and North America each year and an increasing trend in incidence and mortality (15, 16). A woman diagnosed with endometrial cancer first undergoes surgical staging, after which histomorphologic classification and risk group stratification directs further adjuvant therapies or observation only. While the majority of women with endometrial cancer have early-stage disease and excellent outcomes, those with metastases or aggressive subtypes typically experience a modest and/or short-lived response to conventional therapies and frequently succumb to their disease (16).

Endometrial cancer pathologic classification and risk stratification has advanced with the recent development of novel molecular classification tools. These tools are objective, biologically based, and clinically relevant, with implications for prognosis, treatment prediction, and genetic heritability. The Cancer Genome Atlas (TCGA) characterized the transcriptional, proteomic, and genomic landscape of over 370 serous and endometrioid cancers and identified 4 genomic subgroups with distinct prognostic outcomes (17). From this, pared down, pragmatic classifiers have emerged, that can use standard formalin-fixed paraffin embedded (FFPE) tissues obtained from hysterectomy (18–22) or diagnostic biopsies (22, 23). ProMisE (Proactive Molecular Risk Classifier in Endometrial cancer) has been developed (18), confirmed (19) and validated (22) according to the Institute of Medicine Guidelines for the development of new biomarker tests (24). ProMisE stratifies endometrial cancer into 4 groups: (i) a mismatch repair–deficient subtype (MMRd, analogous to TCGA and TransPORTEC “MSI”); (ii) a subtype with mutations in the exonuclease domain of polymerase epsilon, which is responsible for DNA proofreading repair and characterized by excellent outcomes (POLE, analogous to TCGA “ultramutated” and “POLE-mutant” TransPORTEC subgroups (20–22, 25); (iii) a poor-prognosis subtype characterized by p53 abnormalities [p53abn, analogous to TCGA “copy-number high” and TransPORTEC “p53-mutant” subgroups (17, 20, 21)]; and (iv) an intermediate prognosis subtype, with wild-type p53 and no specific mutational profile (p53wt, analogous to TCGA “copy-number low,” TransPORTEC “NSMP” or no specific molecular profile; refs. 18–22).

The molecular features of these subtypes influence the immune landscape of endometrial cancer. Owing to intrinsic DNA repair deficiencies, the POLE and MMRd subtypes give rise to ultramutated (mean = 232 × 10−6 mutations/Mb) and hypermutated (18 × 10−6 mutations/Mb) tumors, respectively (17). Accordingly, these subtypes have been associated with high neoantigen loads and robust TIL responses (9, 26–29). Given the general association between TIL and favorable prognosis across cancers, it has been suggested that the good to excellent prognosis associated with MMRd and POLE tumors is attributable to the antitumor immune response (30, 31). However, prior studies of TILs in endometrial cancer have not had sufficient sample size to test this hypothesis. Moreover, relatively less is known about the immune response within the p53wt and p53abn molecular subtypes, which encompass the majority of endometrial cancer cases (12, 31). Notably, p53wt cases have a similar prognosis as MMRd cases (18), despite an almost 10-fold lower mutation rate (2.9 vs. 18 × 10−6 mutations/Mb; ref. 17). Indeed, with mutation rates spanning 2 orders of magnitude, endometrial cancer provides a powerful setting to investigate the paradigm that high mutation load drives favorable prognosis via the antitumor immune response.

As with other cancers, there is intense interest in the use of checkpoint blockade to treat recurrent endometrial cancer (6, 32, 35). Although relapse is exceedingly rare among POLE cases, there are anecdotal reports of objective responses to anti-PD-1 antibody (36–38), which fits the paradigm that high mutation load predicts response to such agents. Anti-PD-1 antibody is an FDA-approved treatment option for MMRd cases (39), again based on the predictive value of high mutation load. However, it remains unclear whether the p53wt and p53abn subtypes of endometrial cancer might also benefit from checkpoint blockade, as we have a relatively poor understanding of how the immune system responds to these characteristically low mutation load tumors.

To define the relationship between molecular subtype, TIL response, and prognosis in endometrial cancer, we profiled the major TIL subsets (including CD4 and CD8 T cells and B-lineage cells) across a 460-case, molecularly subtyped endometrial cancer cohort with full clinical data. To inform future immunotherapy strategies, we also evaluated immunosuppressive factors, including regulatory T cells (Tregs), IDO-1, and the PD-1/PD-L1 pathway. We report that TIL responses and immunosuppressive features vary in frequency but not magnitude across the molecular subtypes of endometrial cancer. Moreover, the prognostic effect of molecular subtype outweighs TIL response, challenging the paradigm that high mutation load drives prognosis by promoting antitumor immunity. Our findings further suggest that molecular subtype alone may be inadequate to stratify patients with endometrial cancer for immunotherapy.

Patient cohort and ProMisE classification

Our cohort consisted of 460 endometrial cancer cases from 2 previously published series that were used to develop and confirm the ProMisE molecular classifier (18, 19). Clinical, pathologic, molecular, and outcome parameters have previously been described (18, 19) and are detailed in the Supplementary Methods. The study was approved by the Research Ethics Board of BC Cancer and the University of British Columbia.

Tissue microarray and IHC

Representative samples of pathology-reviewed endometrial cancer from our Vancouver center were cored (0.6 mm) in duplicate and arrayed as described previously (18, 19). TMAs were cut at 4-μm thickness onto Superfrost+ glass slides for IHC analysis. Single and multiplex IHC was used to profile immune cells and associated factors in both the epithelial and stromal compartments of tumors. Four antibody panels were used: (i) CD3/CD8/CD79a, (ii) CD138/CD79a, (iii) PD-1/CD8/PD-L1, and (iv) FoxP3/PD-1/CD3. IDO-1 was stained separately (Supplementary Methods). Slides were scanned in brightfield using the Vectra Multispectral Imaging System (PerkinElmer). InForm 2.1 software (PerkinElmer) was used to quantify signals. PD-L1 and IDO-1 positivity was scored visually as a binary variable (whereby >5% staining of immune cells and/or tumor cells was considered positive) or by image analysis as a continuous variable. Additional information is provided in Supplementary Methods.

Statistical analyses

For each tumor core, raw cell counts and marker values were normalized to epithelial and stromal area to yield a density value. For several markers, a substantial proportion of samples had scores of zero (i.e., zero-inflated/right-skewed data). When log (base 2) transformed, the data followed a delta log normal distribution (Supplementary Fig. S1) and were analyzed using a 2-step approach (40, 41). In the first step, the proportion of cases having no marker expression was modeled separately using a χ2 test. Conditional on a nonzero value, the second step followed a lognormal distribution and was assessed using a 1-way ANOVA.

Immune markers were further investigated using heatmaps, overlaying the previously validated ProMisE subtypes. Immune signatures were inferred using an unsupervised ensemble clustering approach implemented in the R package diceR (42). Briefly, several algorithms (Supplementary Methods) were used to cluster 100 random subsets of the data, each of which included 80% of samples. The results were combined to form a final consensus clustering using the k-modes method (43). Several numbers of clusters were tested, and the proportion of ambiguously clustered cases (PAC; ref. 44) within the ensemble was used to determine the optimal number of clusters. Univariable associations between immune clusters, ProMisE subtypes, and other clinicopathologic parameters were evaluated. Associations between immune clusters and clinical outcomes (overall, disease-specific, and progression-free survival) were assessed individually in univariable analyses using log-rank tests and Kaplan–Meier analyses and collectively by adjusting for known risk factors in multivariable Cox proportional hazard models. Survival was also evaluated within each ProMisE subtype. Multivariate analysis was performed, correcting for the immune clusters, ProMisE subtype, age, BMI, grade, histotype, stage, lymph vascular space invasion (LVSI), myometrial invasion, nodal status, or treatment (see Supplementary Statistical Methods). All statistical tests were 2-sided, and statistical significance was set at 0.05 with no adjustment for multiple comparisons.

Cohort and IHC analysis

ProMisE subtype distribution within the 460 endometrial cancer cases included 105 MMRd, 42 POLE, 202 p53wt, and 111 p53abn cases. Clinicopathologic parameters across ProMisE subgroups are shown in Supplementary Table S1. Patients were diagnosed between 1983 and 2013 and received treatment according to standard of care at our center over this time period (18, 19). Adjuvant treatment, when given, consisted primarily of standard radiotherapy (most commonly pelvic external beam radiotherapy or vaginal brachytherapy) with or without platinum/taxane chemotherapy. No patients were known to have received immune checkpoint inhibitors or other immunotherapies.

The densities of various TIL subsets and associated factors were assessed by multiplex IHC and automated image analysis with visual confirmation. Representative images are shown in Fig. 1. The number of missing values (due to missing or uninterpretable cores) ranged from 10 to 28 for individual markers and were not significantly associated with subtype (Supplementary Table S2).

Figure 1.

Representative images of multiplex IHC for TIL subsets (A–D). Specific marker combinations and color legends are as shown, with left column images magnified at 20×.

Figure 1.

Representative images of multiplex IHC for TIL subsets (A–D). Specific marker combinations and color legends are as shown, with left column images magnified at 20×.

Close modal

POLE and MMRd subtypes show increased infiltration by both T cells and B-lineage cells

The epithelial and stromal densities of immune markers are shown in Fig. 2 and Supplementary Table S3. Consistent with prior reports (2, 31), the density of epithelial and stromal CD3+CD8+ TIL (putative cytotoxic T cells) was highest in POLE tumors, followed closely by MMRd tumors, with p53abn and p53wt tumors showing the lowest levels (P < 0.01; Supplementary Table S2). A similar, statistically significant pattern was seen for CD3+CD8 TIL (putative CD4+ TIL), although the differences between molecular subtypes were less pronounced (P < 0.01). Similar patterns were seen in the stromal compartment (P < 0.01), although the densities of CD3+CD8+ and CD3+CD8 T cells were generally higher compared with epithelium.

Figure 2.

The epithelial (A) and stromal (B) densities of immune markers. The density of epithelial and stromal CD3+CD8+ TIL (putative cytotoxic T cells) and CD3+CD8 TIL (putative CD4+ TIL) was highest in POLE tumors, followed closely by MMRd tumors. For B cells (CD138CD79a+) and plasma cells (CD138+CD79a+) markedly higher densities were seen in tumor stroma (not epithelium) and varied by ProMisE subtype (P < 0.01; see also Supplementary Table S2).

Figure 2.

The epithelial (A) and stromal (B) densities of immune markers. The density of epithelial and stromal CD3+CD8+ TIL (putative cytotoxic T cells) and CD3+CD8 TIL (putative CD4+ TIL) was highest in POLE tumors, followed closely by MMRd tumors. For B cells (CD138CD79a+) and plasma cells (CD138+CD79a+) markedly higher densities were seen in tumor stroma (not epithelium) and varied by ProMisE subtype (P < 0.01; see also Supplementary Table S2).

Close modal

In contrast to T cells, neither B cells (CD138CD79a+) nor plasma cells (CD138+CD79a+) were prevalent in tumor epithelium, and neither subset showed a significant difference across ProMisE subtypes (P = 0.74 and P = 0.22, respectively). However, in tumor stroma, higher densities of B cells and plasma cells were seen and varied by ProMisE subtype (P < 0.01 for both), with POLE tumors showing the highest density. Generally, elevated densities of epithelial and stromal T cells in POLE and MMRd tumors were accompanied by higher levels of B cells and plasma cells in the stromal compartment, with the next highest expression levels seen in MMRd (B cells) or p53abn (plasma cells) tumors (Fig. 2; Supplementary Table S2).

Immunosuppressive factors across the ProMisE subtypes

We also evaluated PD-1+ cells, PD-L1+ cells, and Tregs (defined as FoxP3+PD-1+CD3+ cells; Fig. 2; Supplementary Table S2). Consistent with overall TIL densities, POLE tumors expressed the highest densities of epithelial PD-1+CD8 (putative CD4+) and PD-1+CD8+ TILs, followed by MMRd tumors; significantly lower levels of these cells were seen in p53abn and p53wt tumors (P = 0.01 and P < 0.01, respectively). The stromal compartment showed a similar ranking, except that the p53abn subtype ranked second for PD-1+CD8 TILs (P < 0.01). The proportion of PD-1+CD8+ cells of all PD-1+ cells was similar in POLE, MMRd, and p53abn tumors (78.4%, 74.7%, and 77.9%, respectively; Supplementary Fig. S2); however, this value dropped to 33.1% in p53wt tumors. A similar though less-pronounced pattern was seen in stroma. This suggests that CD8+ TILs have a lower level of activation and/or exhaustion in p53wt tumors compared with the other 3 subtypes.

To compare the extent of PD-L1 expression across subtypes, we measured the total area of PD-L1 positivity in tumor cores (i.e., irrespective of tumor versus lymphocyte expression or stromal vs epithelial location). Using a binary scoring method (cores with >5% of cells expressing PD-L1 were considered positive), the proportion of PD-L1+ cases was highest for POLE tumors (70%), followed by MMRd (56%), p53abn (32%), and p53wt tumors (16%; P < 0.01). Likewise, when evaluated as a continuous variable, PD-L1 expression differed significantly across ProMisE subtype (P < 0.01) and followed a similar ranking as seen with binarized data.

Similar to other immune markers, Tregs were highest in the stromal compartment in POLE and MMRd tumors, followed in order by p53abn and p53wt tumors (P < 0.01). When the 4 subtypes were considered together, the ratio of CD3+CD8+ T cells to Tregs was approximately 4:1 in epithelium and 2:1 in stroma (Supplementary Table S4). Within subtypes, these ratios (in epithelium/stroma, respectively) were 5.25/2.72 for POLE, 4.46/2.49 for MMRd, 3.80/1.72 for p53abn, and 3.46/2.28 for p53wt. Thus, POLE cases had the highest ratio of CD3+CD8+ T cells to Tregs.

Lymphocyte infiltration and immunosuppressive factors within the MMRd subtype

Within the MMRd group, we explored differences between patients with loss of MLH1/PMS2 versus MSH2/MSH6. MLH1 loss is usually somatic or epigenetic and generally occurs in older women, raising the possibility it may be associated with a weaker immune response. Of the 105 MMRd cases in our cohort, 71 showed MLH1/PMS2 loss, and 13 showed MSH2/MSH6 loss. Although we were insufficiently powered to make strong conclusions, within the stromal compartment there was a trend toward decreased density of plasma cells in MLH1/PMS2-loss versus MSH2/MSH6-loss tumors (P = 0.07). Moreover, MLH1/PMS2-loss tumors showed significantly lower stromal density of Tregs (FOXP3+PD-1+CD3+ cells; P = 0.05) compared with MSH2/MSH6-loss tumors (Supplementary Fig. S3).

Ensemble clustering reveals 2 major TIL patterns

An ensemble clustering approach was used to identify TIL patterns present in endometrial cancer based on the log-transformed IHC results for all TIL subsets (i.e., CD8+ T cells, putative CD4+ T cells, Tregs, B cells, and plasma cells) and the PD-1/PD-L1 pathway in both the epithelial and stromal compartments. We tested different cluster sizes and found that PAC was lowest in the 2-cluster solution (Supplementary Table S5); nonetheless, we also explored the 3-cluster solution. The 2-cluster solution separated tumors into 2 major groups: TILhigh versus TILlow (Fig. 3A). TILhigh tumors showed significantly higher densities of all 5 TIL subsets, including CD8+ and CD4+ T cells, B cells, plasma cells, and Tregs. High levels of PD-1+ and PD-L1+ cells also characterized this group. In contrast, TILlow tumors showed low or negligible levels of all 5 TIL subsets, as well as PD-1+ and PD-L1+ cells. In the 3-cluster solution (Fig. 3B), the TILhigh cluster remained largely the same, whereas the TILlow cluster split into 2 subgroups referred to as TILmedium and TILnegative. Relative to TILhigh tumors, TILmedium tumors exhibited lower levels of all 5 TIL subsets and notably lower levels of PD-1+ and PD-L1+ cells. The majority of TILmedium tumors (75%) belonged to the p53wt subtype. TILnegative tumors exhibited negligible levels of all 5 TIL subsets and PD-1+ and PD-L1+ cells.

Figure 3.

Cluster analysis of TIL patterns in endometrial cancer based on log-transformed IHC results for all TIL subsets and the PD-1/PD-L1 pathway in both epithelial (A) and stromal compartments (B). TILhigh tumors showed significantly higher densities of all 5 TIL subsets; CD8+ and CD4+ T cells, B cells, plasma cells, and Tregs and high levels of PD-1+ and PD-L1+ cells also characterized this group.

Figure 3.

Cluster analysis of TIL patterns in endometrial cancer based on log-transformed IHC results for all TIL subsets and the PD-1/PD-L1 pathway in both epithelial (A) and stromal compartments (B). TILhigh tumors showed significantly higher densities of all 5 TIL subsets; CD8+ and CD4+ T cells, B cells, plasma cells, and Tregs and high levels of PD-1+ and PD-L1+ cells also characterized this group.

Close modal

None of the ProMisE subtypes exhibited a TIL pattern that was not seen in the other subtypes; rather, they differed with respect to the prevalence of TILhigh tumors: POLE (89.5%), MMRd (78%), p53abn (60%), and p53wt (37%). Associations were noted between TIL patterns and certain clinicopathologic parameters such as BMI, LVSI, nodal status, grade, and treatment (Supplementary Table S6). Further investigation revealed that there were no statistical differences in clinical parameters between POLE and MMRd cases with TILhigh and TILlow immune patterns.

Patient outcomes are more strongly associated with ProMisE subtype than immune pattern

We have previously reported on progression-free and overall survival in this cohort, finding highly favorable outcomes in women with POLE mutated ECs, intermediate outcomes in MMRd and p53wt cases, and poor outcomes in p53abn tumors (18, 19). Cox regression survival analysis with log-rank test was performed across all 4 molecular subtypes to assess the association of individual or combined immune markers with clinical outcomes (Supplementary Table S6). High densities of epithelial CD3+CD8 or CD3+CD8+ TILs were associated with increased patient survival [overall survival (OS) P = 0.05; disease-specific survival (DSS) P = 0.02; progression-free survival (PFS) P = 0.06 and OS P < 0.01, DSS P < 0.01, PFS P < 0.01, respectively]. Likewise, FOXP3PD-1CD3+ cells were prognostically favorable in stroma (OS P < 0.01, DSS P = 0.01, PFS P = 0.01) and epithelium (PFS P < 0.01). Conversely, epithelial PD-1+CD8+ TILs were correlated with decreased OS (P = 0.01), and epithelial PD-1+CD8 TILs were correlated with decreased PFS (P < 0.01). Stromal CD138CD79a+ and CD138+CD79a+ TILs lacked prognostic significance as did PD-L1+ cells and epithelial or stromal Tregs. Expression of IDO-1 correlated with increased PFS (PFS P < 0.01, OS P = 0.16, DSS P = 0.29). TILhigh versus TILlow immune clusters showed no association with OS, DSS, or PFS (Fig. 4A–C), even in multivariable analysis after adjusting for ProMisE subtype and other clinicopathologic parameters associated with survival, which have been assessed previously (18, 19) and include age, stage, LVSI, and myometrial invasion (Table 1). However, ProMisE molecular classifier maintained a statistically significant association with outcomes for OS, DSS, and PFS (P = 0.04, P = 0.04, and P < 0.01 respectively; Table 1).

Figure 4.

Survival analyses. TILhigh versus TILlow immune clusters showed no association with OS (A), DSS (B), or PFS (C) across the full cohort. Within ProMisE subsets, there was a trend toward inferior outcomes in TILlow MMRd endometrial cancers (P = ns, data not shown).

Figure 4.

Survival analyses. TILhigh versus TILlow immune clusters showed no association with OS (A), DSS (B), or PFS (C) across the full cohort. Within ProMisE subsets, there was a trend toward inferior outcomes in TILlow MMRd endometrial cancers (P = ns, data not shown).

Close modal
Table 1.

Multivariable survival

OSDSSPFS
94/36368/35275/312
No. of events/nHR (95% CI)LRT PHR (95% CI)LRT PHR (95% CI)LRT P
Immune cluster (ref: Low immune) High immune 0.80 (0.51–1.26)a 0.346 0.71 (0.42–1.22)a 0.217 0.83 (0.49–1.39)a 0.496 
ProMisE molecular subgroup (ref: p53wt) MMRd 1.71 (0.90–3.26)a 0.036 1.36 (0.62–2.93)a 0.040 0.79 (0.37–1.64)a 0.007 
 POLE 0.87 (0.22–2.59)a  0.37 (0.04–1.61)a  0.16 (0.02–0.67)a  
 p53abn 2.53 (1.26–5.18)a  2.19 (0.99–5.02)a  1.45 (0.70–3.14)a  
Age  1.02 (1.00–1.04)a 0.036 1.00 (0.97–1.02)a 0.724 0.98 (0.96–1.00)a 0.120 
BMI  0.98 (0.96–1.01)a 0.198 0.98 (0.95–1.01)a 0.278 1.00 (0.97–1.03)a 0.888 
Grade (ref grade 1/2) Grade 3 1.13 (0.62–2.10)a 0.706 1.92 (0.88–4.43)a 0.104 3.59 (1.55–9.07)a 0.002 
Histology (ref: endometrioid) Nonendometrioid 1.03 (0.54–1.95)a 0.932 1.36 (0.66–2.80)a 0.406 1.28 (0.66–2.52)a 0.469 
Stage (reference group: I) II–IV 2.44 (1.42–4.24)a 0.001 2.62 (1.38–5.07)a 0.003 3.31 (1.72–6.50)a <0.001 
LVSI (ref: no) Yes 1.89 (1.11–3.29)a 0.018 1.92 (0.99–3.85)a 0.048 1.33 (0.69–2.64)a 0.379 
Myometrial invasion (ref: none) <50 0.81 (0.38–1.92)a 0.022 0.80 (0.29–2.70)a 0.001 0.64 (0.24–1.91)a 0.002 
 >50 1.63 (0.75–3.93)a  2.40 (0.87–8.08)a  1.72 (0.65–5.30)a  
Nodal status (ref: negative) Not tested 1.41 (0.81–2.41)a 0.163 1.85 (0.96–3.49)a 0.136 0.44 (0.17–0.99)a 0.041 
 Positive 0.66 (0.35–1.21)a  0.80 (0.40–1.54)a  0.62 (0.33–1.14)a  
Treatment (ref: none) Any 0.73 (0.43–1.26)a 0.261 0.65 (0.34–1.29)a 0.220 0.60 (0.32–1.17)a 0.136 
OSDSSPFS
94/36368/35275/312
No. of events/nHR (95% CI)LRT PHR (95% CI)LRT PHR (95% CI)LRT P
Immune cluster (ref: Low immune) High immune 0.80 (0.51–1.26)a 0.346 0.71 (0.42–1.22)a 0.217 0.83 (0.49–1.39)a 0.496 
ProMisE molecular subgroup (ref: p53wt) MMRd 1.71 (0.90–3.26)a 0.036 1.36 (0.62–2.93)a 0.040 0.79 (0.37–1.64)a 0.007 
 POLE 0.87 (0.22–2.59)a  0.37 (0.04–1.61)a  0.16 (0.02–0.67)a  
 p53abn 2.53 (1.26–5.18)a  2.19 (0.99–5.02)a  1.45 (0.70–3.14)a  
Age  1.02 (1.00–1.04)a 0.036 1.00 (0.97–1.02)a 0.724 0.98 (0.96–1.00)a 0.120 
BMI  0.98 (0.96–1.01)a 0.198 0.98 (0.95–1.01)a 0.278 1.00 (0.97–1.03)a 0.888 
Grade (ref grade 1/2) Grade 3 1.13 (0.62–2.10)a 0.706 1.92 (0.88–4.43)a 0.104 3.59 (1.55–9.07)a 0.002 
Histology (ref: endometrioid) Nonendometrioid 1.03 (0.54–1.95)a 0.932 1.36 (0.66–2.80)a 0.406 1.28 (0.66–2.52)a 0.469 
Stage (reference group: I) II–IV 2.44 (1.42–4.24)a 0.001 2.62 (1.38–5.07)a 0.003 3.31 (1.72–6.50)a <0.001 
LVSI (ref: no) Yes 1.89 (1.11–3.29)a 0.018 1.92 (0.99–3.85)a 0.048 1.33 (0.69–2.64)a 0.379 
Myometrial invasion (ref: none) <50 0.81 (0.38–1.92)a 0.022 0.80 (0.29–2.70)a 0.001 0.64 (0.24–1.91)a 0.002 
 >50 1.63 (0.75–3.93)a  2.40 (0.87–8.08)a  1.72 (0.65–5.30)a  
Nodal status (ref: negative) Not tested 1.41 (0.81–2.41)a 0.163 1.85 (0.96–3.49)a 0.136 0.44 (0.17–0.99)a 0.041 
 Positive 0.66 (0.35–1.21)a  0.80 (0.40–1.54)a  0.62 (0.33–1.14)a  
Treatment (ref: none) Any 0.73 (0.43–1.26)a 0.261 0.65 (0.34–1.29)a 0.220 0.60 (0.32–1.17)a 0.136 

NOTE: Multivariable survival analysis, including immune clusters (K = 2), ProMisE subtype, and other known risk factors and using Cox proportional hazard to estimate HRs on OS, DSS, and PFS. P values were evaluated using likelihood ratio test (LRT). Boldface refers to P values <0.05.

aFirth confidence intervals were computed using the profile likelihood and may not match the LRT P value.

We assessed the prognostic effect of epithelial CD3+CD8+ TILs in a multivariable model that included ProMisE subtype and these other clinicopathologic parameters (Table 2). Although both ProMisE subtype and epithelial CD8+ TILs showed a trend toward increased OS and DSS, only ProMisE subtype retained a significant independent prognostic effect (PFS P = 0.02).

Table 2

Multivariable survival

OSDSSPFS
94/36368/35275/312
No. of events/nHR (95% CI)LRT PHR (95% CI)LRT PHR (95% CI)LRT P
Epith CD3+ CD8+ (cytotoxic T cells)  0.99 (0.98–1.00)a 0.067 0.99 (0.98–1.00)a 0.080 1.00 (0.99–1.01)a 0.429 
ProMisE molecular subgroup (ref: p53wt) MMRd 1.80 (0.95–3.40)a 0.062 1.35 (0.63–2.87)a 0.094 0.78 (0.37–1.62)a 0.017 
 POLE 1.04 (0.26–3.15)a  0.42 (0.04–1.84)a  0.17 (0.02–0.72)a  
 p53abn 2.38 (1.18–4.89)a  2.02 (0.92–4.62)a  1.40 (0.67–3.02)a  
Age  1.02 (1.00–1.05)a 0.029 1.00 (0.97–1.02)a 0.745 0.98 (0.96–1.00)a 0.100 
BMI  0.98 (0.96–1.01)a 0.169 0.99 (0.96–1.01)a 0.284 1.00 (0.97–1.03)a 0.928 
Grade (ref grade 1/2) Grade 3 1.17 (0.64–2.18)a 0.645 2.06 (0.94–4.78)a 0.078 3.88 (1.68–9.68)a 0.001 
Histology (ref: endometrioid) Nonendometrioid 1.07 (0.56–2.03)a 0.841 1.38 (0.67–2.83)a 0.481 1.28 (0.66–2.51)a 0.467 
Stage (ref: stage: I) II–IV 2.32 (1.34–4.04)a 0.002 2.52 (1.32–4.87)a 0.005 3.28 (1.70–6.45)a <0.001 
LVSI (ref: no) Yes 1.82 (1.06–3.16)a 0.028 1.82 (0.94–3.66)a 0.070 1.28 (0.66–2.52)a 0.445 
Myometrial invasion (ref: none) <50 0.81 (0.38–1.91)a 0.018 0.80 (0.29–2.70)a 0.001 0.64 (0.25–1.92)a 0.002 
 >50 1.66 (0.76–3.97)a  2.43 (0.89–8.15)a  1.72 (0.65–5.30)a  
Nodal status (ref: negative) Not tested 1.45 (0.83–2.46)a 0.205 1.91 (0.99–3.59)a 0.126 0.45 (0.17–1.00)a 0.046 
 Positive 0.70 (0.37–1.29)a  0.81 (0.41–1.56)a  0.62 (0.33–1.14)a  
Treatment (ref: none) Any Rx 0.73 (0.43–1.25)a 0.247 0.66 (0.34–1.29)a 0.223 0.60 (0.31–1.16)a 0.129 
OSDSSPFS
94/36368/35275/312
No. of events/nHR (95% CI)LRT PHR (95% CI)LRT PHR (95% CI)LRT P
Epith CD3+ CD8+ (cytotoxic T cells)  0.99 (0.98–1.00)a 0.067 0.99 (0.98–1.00)a 0.080 1.00 (0.99–1.01)a 0.429 
ProMisE molecular subgroup (ref: p53wt) MMRd 1.80 (0.95–3.40)a 0.062 1.35 (0.63–2.87)a 0.094 0.78 (0.37–1.62)a 0.017 
 POLE 1.04 (0.26–3.15)a  0.42 (0.04–1.84)a  0.17 (0.02–0.72)a  
 p53abn 2.38 (1.18–4.89)a  2.02 (0.92–4.62)a  1.40 (0.67–3.02)a  
Age  1.02 (1.00–1.05)a 0.029 1.00 (0.97–1.02)a 0.745 0.98 (0.96–1.00)a 0.100 
BMI  0.98 (0.96–1.01)a 0.169 0.99 (0.96–1.01)a 0.284 1.00 (0.97–1.03)a 0.928 
Grade (ref grade 1/2) Grade 3 1.17 (0.64–2.18)a 0.645 2.06 (0.94–4.78)a 0.078 3.88 (1.68–9.68)a 0.001 
Histology (ref: endometrioid) Nonendometrioid 1.07 (0.56–2.03)a 0.841 1.38 (0.67–2.83)a 0.481 1.28 (0.66–2.51)a 0.467 
Stage (ref: stage: I) II–IV 2.32 (1.34–4.04)a 0.002 2.52 (1.32–4.87)a 0.005 3.28 (1.70–6.45)a <0.001 
LVSI (ref: no) Yes 1.82 (1.06–3.16)a 0.028 1.82 (0.94–3.66)a 0.070 1.28 (0.66–2.52)a 0.445 
Myometrial invasion (ref: none) <50 0.81 (0.38–1.91)a 0.018 0.80 (0.29–2.70)a 0.001 0.64 (0.25–1.92)a 0.002 
 >50 1.66 (0.76–3.97)a  2.43 (0.89–8.15)a  1.72 (0.65–5.30)a  
Nodal status (ref: negative) Not tested 1.45 (0.83–2.46)a 0.205 1.91 (0.99–3.59)a 0.126 0.45 (0.17–1.00)a 0.046 
 Positive 0.70 (0.37–1.29)a  0.81 (0.41–1.56)a  0.62 (0.33–1.14)a  
Treatment (ref: none) Any Rx 0.73 (0.43–1.25)a 0.247 0.66 (0.34–1.29)a 0.223 0.60 (0.31–1.16)a 0.129 

NOTE: Multivariable survival analysis, including epithelial cytotoxic T cells, ProMisE subtype, and other known risk factors using Cox proportional hazard to estimate HRs on OS, DSS, and PFS. P values were evaluated using likelihood ratio test (LRT). Boldface refers to P values <0.05.

aFirth confidence intervals were computed using the profile likelihood and may not match the LRT P value.

Prognostic effect of immune markers within ProMisE subtypes

Next, we evaluated the prognostic significance of individual immune markers within ProMisE subtypes. In general, most immune markers failed to show prognostic significance, with a few exceptions (Table 3). Within MMRd cases, CD8+CD3+ and CD3+CD8 TILs were positively associated with DSS (P = 0.04 and 0.02, respectively), and within p53wt cases, stromal B cells (CD138CD79a+) were associated with increased DSS (P = 0.04; Table 3). However, even when statistical significance was reached, differences in outcomes were small.

Table 3.

Univariable survival of selected immune markers within ProMisE subtype

OSDSSPFS
MarkerHR (95% CI)PHR (95% CI)PHR (95% CI)P
MMRd 
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.482 (0.222–1.046) 0.059 0.393 (0.145–0.954) 0.037 0.536 (0.203–1.329) 0.173 
  S 0.487 (0.225–1.055) 0.062 0.489 (0.190–1.167) 0.103 0.884 (0.351–2.227) 0.793 
 CD3+CD8 (putative CD4+
  E 0.531 (0.245–1.150) 0.103 0.338 (0.116–0.845) 0.019 0.500 (0.190–1.239) 0.130 
  S 0.762 (0.361–1.612) 0.476 0.713 (0.287–1.687) 0.430 0.724 (0.284–1.791) 0.479 
 CD138CD79a+ (B cells) 
  E 0.816 (0.388–1.716) 0.590 1.078 (0.456–2.612) 0.850 0.881 (0.349–2.220) 0.788 
  S 0.421 (0.185–0.892) 0.023 0.306 (0.105–0.766) 0.010 0.740 (0.296–1.847) 0.521 
 CD8+PD-1+ 
  E 0.808 (0.382–1.708) 0.575 0.707 (0.284–1.672) 0.419 0.801 (0.315–1.982) 0.622 
  S 0.791 (0.374–1.672) 0.538 0.840 (0.347–1.987) 0.682 0.829 (0.326–2.050) 0.674 
POLE 
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.965 (0.149–6.249) 0.971 0.995 (0.081–12.263) 0.997 0.333 (0.002–6.248) 0.317 
  S 2.404 (0.396–24.806) 0.303 1.061 (0.086–13.065) 0.967 3.000 (0.160–437.761) 0.317 
 CD3+CD8 (putative CD4+
  E 1.003 (0.155–6.490) 0.998 0.212 (0.002–2.608) 0.170 3.000 (0.160–437.761) 0.317 
  S 0.104 (0.001–0.976) 0.039 0.188 (0.001–2.316) 0.145 0.296 (0.002–5.554) 0.289 
 CD138CD79a+ (B cells) 
  E 0.429 (0.042–2.611) 0.318 0.212 (0.002–2.608) 0.170 0.333 (0.002–6.248) 0.317 
  S 0.402 (0.039–2.445) 0.287 0.199 (0.001–2.444) 0.156 0.269 (0.002–5.554) 0.289 
 CD8+PD-1+ 
  E 0.402 (0.039–2.445) 0.287 0.199 (0.001–2.444) 0.156 0.296 (0.002–5.554) 0.289 
  S 1.030 (0.159–6.666) 0.976 1.061 (0,086–13.065) 0.967 0.333 (0.002–6.248) 0.317 
P53wt       
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.706 (0.353–1.374) 0.301 0.455 (0.180–1.043) 0.061 0.748 (0.329–1.644) 0.460 
  S 1.558 (0.800–3.113) 0.189 1.409 (0.630–3.236) 0.398 1.178 (0.532–2.607) 0.689 
 CD3+CD8 (putative CD4+
  E 1.098 (0.565–2.156) 0.777 0.783 (0.341–1.750) 0.543 0.793 (0.348–1.743) 0.552 
  S 0.715 (0.361–1.391) 0.320 0.704 (0.306–1.574) 0.387 0.604 (0.259–1.334) 0.207 
 CD138CD79a+ (B cells) 
  E 0.574 (0.283–1.122) 0.102 0.421 (0.167–0.967) 0.040 0.605 (0.259–1.338) 0.209 
  S 0.827 (0.421–1.609) 0.573 0.628 (0.268–1.408) 0.253 0.764 (0.336–1.679) 0.492 
 CD8+PD-1+ 
  E 1.585 (0.770–3.115) 0.214 1.276 (0.506–2.926) 0.650 1.307 (0.521–2.969) 0.611 
  S 0.708 (0.354–1.377) 0.304 0.643 (0.274–1.439) 0.276 0.671 (0.295–1.474) 0.315 
P53abn       
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.929 (0.533–1.619) 0.796 1.066 (0.577–1.971) 0.838 1.136 (0.627–2.057) 0.676 
  S 0.851 (0.488–1.483) 0.570 1.039 (0.562–1.921) 0.903 0.768 (0.424–1.391) 0.381 
 CD3+CD8 (putative CD4+
  E 1.126 (0.644–1.966) 0.677 1.431 (0.763–2.686) 0.262 1.165 (0.643–2.111) 0.615 
  S 1.065 (0.610–1.860) 0.824 1.198 (0.643–2.233) 0.568 0.770 (0.425–1.394) 0.385 
 CD138CD79a+ (B cells) 
  E 1.558 (0.888–2.733) 0.120 1.678 (0.895–3.144) 0.102 1.301 (0.719–2.357) 0.384 
  S 0.800 (0.458–1.395) 0.430 0.936 (0.507–1.728) 0.833 0.971 (0.537–1.754) 0.920 
 CD8+PD-1+ 
  E 1.253 (0.719–2.184) 0.424 1.540 (0.826–2.869) 0.717 1.256 (0.695–2.270) 0.451 
  S 1.102 (0.632–1.919) 0.733 1.358 (0.732–2.518) 0.330 0.741 (0.409–1.344) 0.321 
OSDSSPFS
MarkerHR (95% CI)PHR (95% CI)PHR (95% CI)P
MMRd 
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.482 (0.222–1.046) 0.059 0.393 (0.145–0.954) 0.037 0.536 (0.203–1.329) 0.173 
  S 0.487 (0.225–1.055) 0.062 0.489 (0.190–1.167) 0.103 0.884 (0.351–2.227) 0.793 
 CD3+CD8 (putative CD4+
  E 0.531 (0.245–1.150) 0.103 0.338 (0.116–0.845) 0.019 0.500 (0.190–1.239) 0.130 
  S 0.762 (0.361–1.612) 0.476 0.713 (0.287–1.687) 0.430 0.724 (0.284–1.791) 0.479 
 CD138CD79a+ (B cells) 
  E 0.816 (0.388–1.716) 0.590 1.078 (0.456–2.612) 0.850 0.881 (0.349–2.220) 0.788 
  S 0.421 (0.185–0.892) 0.023 0.306 (0.105–0.766) 0.010 0.740 (0.296–1.847) 0.521 
 CD8+PD-1+ 
  E 0.808 (0.382–1.708) 0.575 0.707 (0.284–1.672) 0.419 0.801 (0.315–1.982) 0.622 
  S 0.791 (0.374–1.672) 0.538 0.840 (0.347–1.987) 0.682 0.829 (0.326–2.050) 0.674 
POLE 
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.965 (0.149–6.249) 0.971 0.995 (0.081–12.263) 0.997 0.333 (0.002–6.248) 0.317 
  S 2.404 (0.396–24.806) 0.303 1.061 (0.086–13.065) 0.967 3.000 (0.160–437.761) 0.317 
 CD3+CD8 (putative CD4+
  E 1.003 (0.155–6.490) 0.998 0.212 (0.002–2.608) 0.170 3.000 (0.160–437.761) 0.317 
  S 0.104 (0.001–0.976) 0.039 0.188 (0.001–2.316) 0.145 0.296 (0.002–5.554) 0.289 
 CD138CD79a+ (B cells) 
  E 0.429 (0.042–2.611) 0.318 0.212 (0.002–2.608) 0.170 0.333 (0.002–6.248) 0.317 
  S 0.402 (0.039–2.445) 0.287 0.199 (0.001–2.444) 0.156 0.269 (0.002–5.554) 0.289 
 CD8+PD-1+ 
  E 0.402 (0.039–2.445) 0.287 0.199 (0.001–2.444) 0.156 0.296 (0.002–5.554) 0.289 
  S 1.030 (0.159–6.666) 0.976 1.061 (0,086–13.065) 0.967 0.333 (0.002–6.248) 0.317 
P53wt       
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.706 (0.353–1.374) 0.301 0.455 (0.180–1.043) 0.061 0.748 (0.329–1.644) 0.460 
  S 1.558 (0.800–3.113) 0.189 1.409 (0.630–3.236) 0.398 1.178 (0.532–2.607) 0.689 
 CD3+CD8 (putative CD4+
  E 1.098 (0.565–2.156) 0.777 0.783 (0.341–1.750) 0.543 0.793 (0.348–1.743) 0.552 
  S 0.715 (0.361–1.391) 0.320 0.704 (0.306–1.574) 0.387 0.604 (0.259–1.334) 0.207 
 CD138CD79a+ (B cells) 
  E 0.574 (0.283–1.122) 0.102 0.421 (0.167–0.967) 0.040 0.605 (0.259–1.338) 0.209 
  S 0.827 (0.421–1.609) 0.573 0.628 (0.268–1.408) 0.253 0.764 (0.336–1.679) 0.492 
 CD8+PD-1+ 
  E 1.585 (0.770–3.115) 0.214 1.276 (0.506–2.926) 0.650 1.307 (0.521–2.969) 0.611 
  S 0.708 (0.354–1.377) 0.304 0.643 (0.274–1.439) 0.276 0.671 (0.295–1.474) 0.315 
P53abn       
 CD3+CD8+ (putative cytotoxic T cells) 
  E 0.929 (0.533–1.619) 0.796 1.066 (0.577–1.971) 0.838 1.136 (0.627–2.057) 0.676 
  S 0.851 (0.488–1.483) 0.570 1.039 (0.562–1.921) 0.903 0.768 (0.424–1.391) 0.381 
 CD3+CD8 (putative CD4+
  E 1.126 (0.644–1.966) 0.677 1.431 (0.763–2.686) 0.262 1.165 (0.643–2.111) 0.615 
  S 1.065 (0.610–1.860) 0.824 1.198 (0.643–2.233) 0.568 0.770 (0.425–1.394) 0.385 
 CD138CD79a+ (B cells) 
  E 1.558 (0.888–2.733) 0.120 1.678 (0.895–3.144) 0.102 1.301 (0.719–2.357) 0.384 
  S 0.800 (0.458–1.395) 0.430 0.936 (0.507–1.728) 0.833 0.971 (0.537–1.754) 0.920 
 CD8+PD-1+ 
  E 1.253 (0.719–2.184) 0.424 1.540 (0.826–2.869) 0.717 1.256 (0.695–2.270) 0.451 
  S 1.102 (0.632–1.919) 0.733 1.358 (0.732–2.518) 0.330 0.741 (0.409–1.344) 0.321 

NOTE: The within-subtype association between select immune markers binarized at the median with clinical outcome were assessed by Cox proportion hazard to compute HRs of OS, DSS, and PFS using a Kaplan–Meier survival analysis. E refers to markers assessed in the epithelium, and S in the stroma.

Finally, we evaluated the prognostic significance of the TILhigh versus TILlow clusters within each of the 4 ProMisE subtypes. This analysis was not meaningful in the POLE subtype, as only 4 cases were TILlow. Similarly, we were challenged to see a difference within p53wt where events were rare. In the remaining ProMisE subtypes, there was a nonsignificant trend toward inferior outcomes in TILlow tumors within MMRd tumors. Collectively, these findings indicate that patient survival is more strongly influenced by ProMisE subtype than TILs.

There is increasing appreciation of the importance of the tumor microenvironment in defining outcomes and therapeutic opportunities in cancer. In particular, TILs have been shown to be both prognostic and predictive in that their presence is generally associated with improved outcomes and favorable response to conventional chemotherapies and immune-targeting agents (1–4, 45–49). We investigated the relationships between immune response, molecular subtype, and patient survival in endometrial cancer. Multiplexed, IHC-based analysis of 460 tumors spanning the 4 molecular subtypes of endometrial cancer (MMRd, POLE, p53wt, and p53abn) identified 2 major TIL patterns: TILhigh and TILlow. As expected, POLE and MMRd subtypes were enriched for TILhigh tumors; however, 22% of MMRd tumors were TILlow. Conversely, p53abn and p53wt cases were enriched for TILlow tumors, yet also contained significant proportions of TILhigh tumors. Therefore, all 4 molecular subtypes of endometrial cancer can engender both immunologically “hot” and “cold” tumors, suggesting it may be inadequate to use molecular subtype alone to stratify patients for immunotherapy. As observed across many cancers, CD8+ TILs were associated with improved survival, but in multivariate analysis, individual immune markers and immune clusters were not prognostic. Rather, ProMisE molecular subtype retained prognostic value, demonstrating an association with both DSS (P = 0.04) and PFS (<0.01) and a trend toward association with OS (P = 0.08). Thus, molecular subtype, not immune response, was the most significant prognostic factor in this series, suggesting that the molecular/genomic features of tumors influence outcome through mechanisms apart from the immune response.

Immune markers have been assessed in endometrial cancer in both the pre- (2, 50) and post-TCGA eras (12, 30, 31, 46, 51). The latter have primarily focused on the newly recognized POLE ultramutated and MSI/MMRd-hypermutated subtypes. Particular attention has been paid to the notion that the exceptionally high mutation rates in these subtypes drive the generation of neoantigens, making tumors more immunogenic. Indeed, POLE and MMRd tumors have been shown to harbor the highest levels of not only CD8+ T cells but also CD4+ T cells, stromal CD20+ B cells, FOXP3+ cells, PD-1+ cells, and PD-L1+ cells (26, 27, 30). Model systems have further demonstrated that POLE ultramutated tumors are able to stimulate greater cytotoxic CD8+ T-cell responses compared with POLE-wt tumors (27). These findings are consistent with a mechanistic link between mutation load and TILs. On the other hand, our findings demonstrate that not all POLE and MMRd tumors engender a significant immune response. Conversely, significant numbers of p53abn and p53wt cases exhibited TIL patterns that, by the clustering analyses performed here, were indistinguishable from TILhigh POLE and MMRd cases. Although we did not directly assess mutation load in this study, the TCGA study (17) demonstrated minimal overlap in the mutation load between the POLE/MSI groups compared with the copy-number high (p53abn) and low (p53wt) groups. Thus, neoantigen load cannot be the only factor influencing the strength of TIL responses. Other factors that merit consideration include DNA damage–induced activation of the cGAS/STING, as well as inhibitory factors such as activation of the Wnt/β-catenin and PTEN/PI3K pathways (52–54). Notably, recurrent mutations in the PTEN, PIK3CA, PIK3R1, and CTNNB1 genes are common in endometrial cancer (55) and hence could play an important role in sculpting the immune response.

This series represents the most extensive assessment to date of B-cell and plasma cell responses in endometrial cancer. Consistent with prior reports in high-grade serous ovarian cancer (HGSC) and other malignancies, B cells and plasma cells were positively associated with CD8 and CD4 TILs (48, 56). Given our use of TMA cores, we were unable to assess the extent to which B-cell and plasma cell infiltrates were associated with tertiary lymphoid structures (57); this could be evaluated in the future by analyzing whole-tumor sections. We found B-lineage cells in both high and low mutation load molecular subtypes, consistent with their lack of association with mutation load in HGSCs (48). Given the extraordinarily favorable prognosis associated with POLE tumors, the presence of dense B-cell and plasma cell infiltrates in this subset challenges the view advanced by some researchers that B-cell/plasma cell responses are detrimental to antitumor immunity. Instead, our findings are in line with the majority of studies showing a positive prognostic effect of B-lineage cells across human cancers (58).

On the surface, the generally higher TIL levels in POLE and MMRd tumors would support these subtypes as the most appropriate candidates for checkpoint blockade. Indeed, objective responses to PD-1 blockade have been demonstrated in patients harboring POLE and MMRd tumors (32, 36, 38). However, recent preclinical data as well as meta-analysis of all published cohorts of POLE endometrial cancers indicates that these tumors may not require any adjuvant therapy (59, 60), let alone costly immunotherapies. Thus, the need for checkpoint blockade in POLE endometrial cancers would be restricted to rare recurrences. The use of checkpoint blockade for relapsed MMRd tumors is well founded and indeed has recently received FDA approval (32, 39). The major outstanding question is whether checkpoint blockade has a place in the treatment of p53abn and p53wt cases. Our findings indicate that significant proportions of these cases exhibit TIL patterns that are equally robust to those seen in POLE and MMRd tumors. Indeed, in our clustering analyses, POLE and MMRd tumors did not demonstrate categorically distinct TIL patterns but rather were part of a continuum shared with the p53wt and p53abn subtypes. This provides justification for evaluating the efficacy of checkpoint blockade in the p53wt and p53abn subtypes.

A major distinguishing feature of TIL clusters was the presence of PD-1+ and PD-L1+ cells. In particular, our 3-cluster model (Fig. 2B) revealed that the TILlow cluster contained a subgroup with intermediate levels of the major TIL subsets (i.e., T cells, B cells, plasma cells, Tregs) but negligible levels of PD-1+ T cells and PD-L1+ macrophages/tumor cells. This subgroup was dominated by p53wt tumors, which as corroborating evidence also demonstrated markedly lower percentages of PD-1+ T cells. Thus, with either 2- or 3-cluster statistical models, only the TILhigh subset had the PD-1+/PD-L1+ pattern that is associated (albeit imperfectly) with response to checkpoint blockade in other cancers. This TILhigh group contained the majority of POLE and MMRd tumors but also 28% of p53wt and 25% of p53abn tumors. Thus, there may be merit in comparing the TILhigh pattern versus molecular subtype as predictive biomarkers for checkpoint blockade in endometrial cancer.

With the MMRd subtype, we investigated whether the underlying genetic defect had an influence on TIL pattern. Somatic or epigenetic loss of PMS2/MLH1 is more common and usually arises in older individuals, who could conceivably mount less robust antitumor immune responses. In contrast, loss of MSH6/MSH2 is often seen in germline/Lynch Syndrome–associated endometrial cancers, which arise in younger individuals. Although our study was underpowered to address this issue definitively, cases with PMS2 loss showed a trend towards lower levels of several immune cells or markers (Tregs, B cells, plasma cells, PD-1, PD-L1), particularly in tumor stroma (Supplementary Fig. S2). Sloan and colleagues showed higher PD-L1 expression in Lynch syndrome–associated endometrial cancers as compared with endometrial cancers with MLH1 promoter methylation (61). However, as in our series, these observations were not accompanied by data on actual response rates to immune blockade, thus the predictive value of these differences within molecular subtype awaits verification through clinical trials.

In addition to the PD-1/PD-L1 pathway, we assessed the expression of IDO-1, which represents another pharmacologically targetable mechanism of immune suppression in cancer (62–66). Similar to PD-L1, IDO-1 expression was significantly higher in TILhigh tumors and spanned the 4 molecular subtypes. Prior publications have suggested high prevalence of IDO-1 expression in MMRd tumors, particularly Lynch syndrome–associated endometrial cancer (67, 68). Our series confirmed this and revealed even higher expression in the POLE subtype. Given that IDO-1 is frequently coexpressed with PD-L1 (not specifically evaluated in this series), dual immunotherapy could be considered (69, 70).

Strengths of this study include evaluation of a large, well-annotated cohort of endometrial cancers and a wide panel of immune markers. Moreover, we used a molecular classification tool that has undergone rigorous validation according to the Institute of Medicine criteria (70). Limitations of this study include the use of tissue microarrays instead of whole sections; however, we assessed cores in duplicates and the number of uninterpretable cases was very low in this series. Moreover, prior literature supports the use of tissue microarrays for immune studies of this type (71, 72). Nonetheless, if TIL patterns were to be used as clinical biomarkers, additional studies would be required to determine whether whole sections provide more robust data for clinical decision making. A second limitation is that we evaluated only a single tumor site from each patient and hence were unable to assess the extent of intratumoral heterogeneity of TIL patterns, as we reported recently in high-grade serous ovarian cancer (56). Importantly, however, this phenomenon does not negate the clinicopathologic significance of TIL when measured across large patient cohorts. A third limitation is that mutation load and other molecular/genomic features of tumors were inferred by ProMisE subtype rather than direct measurement. Finally, given that this cohort was accrued prior to the widespread use of immune-modulatory agents in endometrial cancer, we were unable to assess whether any immune cells or markers were predictive of response to PD-1/PD-L1 or IDO-1 inhibitors.

In summary, we have profiled the immune response in endometrial cancer through the modern lens of a pragmatic molecular classification tool. Immune markers, alone or in combination, failed to match the prognostic ability of ProMisE molecular classification. We demonstrate the diversity of immune marker expression across and within molecular subtypes. Candidates for immunotherapy extend beyond the POLE and MMRd subtypes, and assessment of immune response rather than molecular subtype may be more helpful in patient selection. Continued work is needed to identify the most accurate biomarkers for stratification of patients with endometrial cancer for immunotherapy, so as to minimize toxicity and cost for women who are unlikely to respond while ensuring that opportunities for treatment are not missed.

No potential conflicts of interest were disclosed.

Conception and design: A. Talhouk, H. Derocher, B. Gilks, B.H. Nelson, J.N. McAlpine

Development of methodology: H. Derocher, K. Milne, B. Gilks, B.H. Nelson, J.N. McAlpine

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): H. Derocher, K. Milne, B. Gilks, B.H. Nelson, J.N. McAlpine

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Talhouk, S. Leung, M.S. Anglesio, B.H. Nelson, J.N. McAlpine

Writing, review, and/or revision of the manuscript: A. Talhouk, H. Derocher, P. Schmidt, S. Leung, K. Milne, B. Gilks, M.S. Anglesio, B.H. Nelson, J.N. McAlpine

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Talhouk, S. Leung, K. Milne, B.H. Nelson, J.N. McAlpine

Study supervision: A. Talhouk, B.H. Nelson

We thank Dr. Phineas Hamilton for advice. This work was supported by grants from the BC Cancer Foundation (to J.N. McAlpine, B.H. Nelson), Terry Fox Research Institute (to B.H. Nelson), Canadian Institutes of Health Research (to A. Talhouk, B.H. Nelson, J.N. McAlpine), Canada's Networks of Centres of Excellence (to B.H. Nelson), and Canada Foundation for Innovation (to B.H. Nelson).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Zhang
L
,
Conejo-Garcia
JR
,
Katsaros
D
,
Gimotty
PA
,
Massobrio
M
,
Regnani
G
, et al
Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer
.
N Engl J Med
2003
;
348
:
203
13
.
2.
de Jong
RA
,
Leffers
N
,
Boezen
HM
,
ten Hoor
KA
,
van der Zee
AG
,
Hollema
H
, et al
Presence of tumor-infiltrating lymphocytes is an independent prognostic factor in type I and II endometrial cancer
.
Gynecol Oncol
2009
;
114
:
105
10
.
3.
Clemente
CG
,
Mihm
MC
 Jr
,
Bufalino
R
,
Zurrida
S
,
Collini
P
,
Cascinelli
N
. 
Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma
.
Cancer
1996
;
77
:
1303
10
.
4.
Gooden
MJ
,
de Bock
GH
,
Leffers
N
,
Daemen
T
,
Nijman
HW
. 
The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis
.
Br J Cancer
2011
;
105
:
93
103
.
5.
Rizvi
NA
,
Hellmann
MD
,
Snyder
A
,
Kvistborg
P
,
Makarov
V
,
Havel
JJ
, et al
Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer
.
Science
2015
;
348
:
124
8
.
6.
Tumeh
PC
,
Harview
CL
,
Yearley
JH
,
Shintaku
IP
,
Taylor
EJ
,
Robert
L
, et al
PD-1 blockade induces responses by inhibiting adaptive immune resistance
.
Nature
2014
;
515
:
568
71
.
7.
Topalian
SL
,
Hodi
FS
,
Brahmer
JR
,
Gettinger
SN
,
Smith
DC
,
McDermott
DF
, et al
Safety, activity, and immune correlates of anti-PD-1 antibody in cancer
.
N Engl J Med
2012
;
366
:
2443
54
.
8.
Brown
SD
,
Warren
RL
,
Gibb
EA
,
Martin
SD
,
Spinelli
JJ
,
Nelson
BH
, et al
Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival
.
Genome Res
2014
;
24
:
743
50
.
9.
Temko
D
,
Van Gool
IC
,
Rayner
E
,
Glaire
M
,
Makino
S
,
Brown
M
, et al
Somatic POLE exonuclease domain mutations are early events in sporadic endometrial and colorectal carcinogenesis, determining driver mutational landscape, clonal neoantigen burden and immune response
.
J Pathol
2018
;
245
:
283
96
.
10.
Parkes
EE
,
Walker
SM
,
Taggart
LE
,
McCabe
N
,
Knight
LA
,
Wilkinson
R
, et al
Activation of STING-dependent innate immune signaling by S-phase-specific DNA damage in breast cancer
.
J Natl Cancer Inst
2017
;
109
.
11.
Blanco
B
,
Herrero-Sanchez
C
,
Rodriguez-Serrano
C
,
Sanchez-Barba
M
,
Del Canizo
MC
. 
Comparative effect of two pan-class I PI3K inhibitors used as anticancer drugs on human T cell function
.
Int Immunopharmacol
2015
;
28
:
675
85
.
12.
Shukla
SA
,
Howitt
BE
,
Wu
CJ
,
Konstantinopoulos
PA
. 
Predicted neoantigen load in non-hypermutated endometrial cancers: correlation with outcome and tumor-specific genomic alterations
.
Gynecol Oncol Rep
2017
;
19
:
42
5
.
13.
Kurnit
KC
,
Munsell
MF
,
Broaddus
RR
. 
Molecular modifiers of the tumor immune microenvironment: activation of the Wnt/β-catenin pathway is associated with local immunosuppression in endometrial cancer
.
The United States and Canadian Academy of Pathology Annual Meeting. San Antonio, TX
. 
2017
.
14.
Abu-Eid
R
,
Samara
RN
,
Ozbun
L
,
Abdalla
MY
,
Berzofsky
JA
,
Friedman
KM
, et al
Selective inhibition of regulatory T cells by targeting the PI3K-Akt pathway
.
Cancer Immunol Res
2014
;
2
:
1080
9
.
15.
Surveillance Epidemiology and End Results
. 
SEER Stat Fact Sheets: endometrial cancer
; 
2015
.
Available from:
https://seer.cancer.gov/statfacts/html/corp.html.
Accessed December 17, 2018.
17.
Cancer Genome Atlas Research Network
,
Kandoth
C
,
Schultz
N
,
Cherniack
AD
,
Akbani
R
,
Liu
Y
, et al
Integrated genomic characterization of endometrial carcinoma
.
Nature
2013
;
497
:
67
73
.
18.
Talhouk
A
,
McConechy
MK
,
Leung
S
,
Li-Chang
HH
,
Kwon
JS
,
Melnyk
N
, et al
A clinically applicable molecular-based classification for endometrial cancers
.
Br J Cancer
2015
;
113
:
299
310
.
19.
Talhouk
A
,
McConechy
MK
,
Leung
S
,
Yang
W
,
Lum
A
,
Senz
J
, et al
Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer
.
Cancer
2017
;
123
:
802
13
.
20.
Stelloo
E
,
Bosse
T
,
Nout
RA
,
MacKay
HJ
,
Church
DN
,
Nijman
HW
, et al
Refining prognosis and identifying targetable pathways for high-risk endometrial cancer; a TransPORTEC initiative
.
Mod Pathol
2015
;
28
:
836
44
.
21.
Stelloo
E
,
Nout
RA
,
Osse
EM
,
Jurgenliemk-Schulz
IJ
,
Jobsen
JJ
,
Lutgens
LC
, et al
Improved risk assessment by integrating molecular and clinicopathological factors in early-stage endometrial cancer-combined analysis of the PORTEC cohorts
.
Clin Cancer Res
2016
;
22
:
4215
24
.
22.
Kommoss
S
,
McConechy
MK
,
Kommoss
F
,
Leung
S
,
Bunz
A
,
Magrill
J
, et al
Final validation of the ProMisE molecular classifier for endometrial carcinoma in a large population-based case series
.
Ann Oncol
2018
;
29
:
1180
8
.
23.
Talhouk
A
,
Hoang
LN
,
McConechy
MK
,
Nakonechny
Q
,
Leo
J
,
Cheng
A
, et al
Molecular classification of endometrial carcinoma on diagnostic specimens is highly concordant with final hysterectomy: earlier prognostic information to guide treatment
.
Gynecol Oncol
2016
;
143
:
46
53
.
24.
Micheel
CM
,
Nass
SJ
,
Omenn
GS
.
Evolution of translational omics: lessons learned and the path forward
.
Washington, DC
:
National Academies Press
; 
2012
.
Available from
: https://www.ncbi.nlm.nih.gov/pubmed/24872966.
25.
McConechy
MK
,
Talhouk
A
,
Leung
S
,
Chiu
D
,
Yang
W
,
Senz
J
, et al
Endometrial carcinomas with POLE exonuclease domain mutations have a favorable prognosis
.
Clin Cancer Res
2016
;
22
:
2865
73
.
26.
Howitt
BE
,
Shukla
SA
,
Sholl
LM
,
Ritterhouse
LL
,
Watkins
JC
,
Rodig
S
, et al
Association of polymerase e-mutated and microsatellite-instable endometrial cancers with neoantigen load, number of tumor-infiltrating lymphocytes, and expression of PD-1 and PD-L1
.
JAMA Oncol
2015
;
1
:
1319
23
.
27.
Bellone
S
,
Centritto
F
,
Black
J
,
Schwab
C
,
English
D
,
Cocco
E
, et al
Polymerase epsilon (POLE) ultra-mutated tumors induce robust tumor-specific CD4+ T cell responses in endometrial cancer patients
.
Gynecol Oncol
2015
;
138
:
11
7
.
28.
Piulats
JM
,
Matias-Guiu
X
. 
Immunotherapy in endometrial cancer: in the nick of time
.
Clin Cancer Res
2016
;
22
:
5623
5
.
29.
van Gool
IC
,
Bosse
T
,
Church
DN
. 
POLE proofreading mutation, immune response and prognosis in endometrial cancer
.
Oncoimmunology
2016
;
5
:
e1072675
.
30.
van Gool
IC
,
Eggink
FA
,
Freeman-Mills
L
,
Stelloo
E
,
Marchi
E
,
de Bruyn
M
, et al
POLE proofreading mutations elicit an antitumor immune response in endometrial cancer
.
Clin Cancer Res
2015
;
21
:
3347
55
.
31.
Eggink
FA
,
Van Gool
IC
,
Leary
A
,
Pollock
PM
,
Crosbie
EJ
,
Mileshkin
L
, et al
Immunological profiling of molecularly classified high-risk endometrial cancers identifies POLE-mutant and microsatellite unstable carcinomas as candidates for checkpoint inhibition
.
Oncoimmunology
2017
;
6
:
e1264565
.
32.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
33.
Overman
MJ
,
McDermott
R
,
Leach
JL
,
Lonardi
S
,
Lenz
HJ
,
Morse
MA
, et al
Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study
.
Lancet Oncol
2017
;
18
:
1182
91
.
34.
Nanda
R
,
Chow
LQ
,
Dees
EC
,
Berger
R
,
Gupta
S
,
Geva
R
, et al
Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study
.
J Clin Oncol
2016
;
34
:
2460
7
.
35.
Hui
R
,
Garon
EB
,
Goldman
JW
,
Leighl
NB
,
Hellmann
MD
,
Patnaik
A
, et al
Pembrolizumab as first-line therapy for patients with PD-L1-positive advanced non-small cell lung cancer: a phase 1 trial
.
Ann Oncol
2017
;
28
:
874
81
.
36.
Santin
AD
,
Bellone
S
,
Buza
N
,
Choi
J
,
Schwartz
PE
,
Schlessinger
J
, et al
Regression of chemotherapy-resistant polymerase epsilon (POLE) ultra-mutated and MSH6 hyper-mutated endometrial tumors with nivolumab
.
Clin Cancer Res
2016
;
22
:
5682
7
.
37.
Mittica
G
,
Ghisoni
E
,
Giannone
G
,
Aglietta
M
,
Genta
S
,
Valabrega
G
. 
Checkpoint inhibitors in endometrial cancer: preclinical rationale and clinical activity
.
Oncotarget
2017
;
8
:
90532
44
.
38.
Mehnert
JM
,
Panda
A
,
Zhong
H
,
Hirshfield
K
,
Damare
S
,
Lane
K
, et al
Immune activation and response to pembrolizumab in POLE-mutant endometrial cancer
.
J Clin Invest
2016
;
126
:
2334
40
.
39.
U.S. Food and Drug Administration/U.S. Department of Health and Human Services
. 
FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication
; 
2017
.
Available from
: https://www.fda.gov/drugs/informationondrugs/approveddrugs/ucm560040.htm.
40.
Duan
N
,
Manning
WG
,
Morris
CN
,
Newhouse
JP
. 
A comparison of alternative models for the demand for medical care
.
J Bus Econ Stat
1983
;
1
:
115
26
.
41.
Min
Y
,
Agresti
A
. 
Modeling nonnegative data with clumping at zero: a survey
.
JIRSS
2002
;
1
:
7
33
.
42.
Chiu
DS
,
Talhouk
A
. 
diceR: an R package for class discovery using an ensemble driven approach
.
BMC Bioinformatics
2018
;
19
:
11
.
43.
Huang
Z
. 
Extensions to the k-means algorithm for clustering large data sets with categorical values
.
Data Min Knowl Discov
1998
;
2
:
283
304
.
44.
Senbabaoglu
Y
,
Michailidis
G
,
Li
JZ
. 
Critical limitations of consensus clustering in class discovery
.
Sci Rep
2014
;
4
:
6207
.
45.
McGranahan
N
,
Furness
AJ
,
Rosenthal
R
,
Ramskov
S
,
Lyngaa
R
,
Saini
SK
, et al
Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade
.
Science
2016
;
351
:
1463
9
.
46.
Bellone
S
,
Bignotti
E
,
Lonardi
S
,
Ferrari
F
,
Centritto
F
,
Masserdotti
A
, et al
Polymerase epsilon (POLE) ultra-mutation in uterine tumors correlates with T lymphocyte infiltration and increased resistance to platinum-based chemotherapy in vitro
.
Gynecol Oncol
2017
;
144
:
146
52
.
47.
Clarke
B
,
Tinker
AV
,
Lee
CH
,
Subramanian
S
,
van de Rijn
M
,
Turbin
D
, et al
Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss
.
Mod Pathol
2009
;
22
:
393
402
.
48.
Kroeger
DR
,
Milne
K
,
Nelson
BH
. 
Tumor-infiltrating plasma cells are associated with tertiary lymphoid structures, cytolytic T-cell responses, and superior prognosis in ovarian cancer
.
Clin Cancer Res
2016
;
22
:
3005
15
.
49.
Leon-Ferre
RA
,
Polley
MY
,
Liu
H
,
Gilbert
JA
,
Cafourek
V
,
Hillman
DW
, et al
Impact of histopathology, tumor-infiltrating lymphocytes, and adjuvant chemotherapy on prognosis of triple-negative breast cancer
.
Breast Cancer Res Treat
2018
;
167
:
89
99
.
50.
Kondratiev
S
,
Sabo
E
,
Yakirevich
E
,
Lavie
O
,
Resnick
MB
. 
Intratumoral CD8+ T lymphocytes as a prognostic factor of survival in endometrial carcinoma
.
Clin Cancer Res
2004
;
10
:
4450
6
.
51.
Gargiulo
P
,
Della Pepa
C
,
Berardi
S
,
Califano
D
,
Scala
S
,
Buonaguro
L
, et al
Tumor genotype and immune microenvironment in POLE-ultramutated and MSI-hypermutated endometrial cancers: new candidates for checkpoint blockade immunotherapy?
Cancer Treat Rev
2016
;
48
:
61
8
.
52.
Pai
SG
,
Carneiro
BA
,
Mota
JM
,
Costa
R
,
Leite
CA
,
Barroso-Sousa
R
, et al
Wnt/beta-catenin pathway: modulating anticancer immune response
.
J Hematol Oncol
2017
;
10
:
101
.
53.
Peng
W
,
Chen
JQ
,
Liu
C
,
Malu
S
,
Creasy
C
,
Tetzlaff
MT
, et al
Loss of PTEN promotes resistance to T cell-mediated immunotherapy
.
Cancer Discov
2016
;
6
:
202
16
.
54.
Pepin
G
,
Gantier
MP
. 
cGAS-STING activation in the tumor microenvironment and its role in cancer immunity
.
Adv Exp Med Biol
2017
;
1024
:
175
94
.
55.
Kandoth
C
,
McLellan
MD
,
Vandin
F
,
Ye
K
,
Niu
B
,
Lu
C
, et al
Mutational landscape and significance across 12 major cancer types
.
Nature
2013
;
502
:
333
9
.
56.
Zhang
AW
,
McPherson
A
,
Milne
K
,
Kroeger
DR
,
Hamilton
PT
,
Miranda
A
, et al
Interfaces of malignant and immunologic clonal dynamics in ovarian cancer
.
Cell
2018
;
173
:
1755
69
.
57.
Teillaud
JL
,
Dieu-Nosjean
MC
. 
Tertiary lymphoid structures: an anti-tumor school for adaptive immune cells and an antibody factory to fight cancer?
Front Immunol
2017
;
8
:
830
.
58.
Wouters
MC
,
Nelson
BH
. 
Prognostic significance of tumor-infiltrating B cells and plasma cells in human cancer
.
Clin Cancer Res
2018 Jul 26
[Epub ahead of print]
.
59.
Van Gool
IC
,
Rayner
E
,
Osse
EM
,
Nout
RA
,
Creutzberg
CL
,
Tomlinson
I
, et al
Adjuvant treatment for POLE proofreading domain-mutant cancers: sensitivity to radiotherapy, chemotherapy, and nucleoside analogs
.
Clin Cancer Res
2018
;
24
:
3197
3203
.
60.
McAlpine
JN
,
Nout
R
,
Kommoss
S
,
Church
D
,
Brucker
S
,
Koebel
M
, et al
Survival benefit in women with endometrial cancers harboring POLE may be independent of adjuvant therapy. International Gynecologic Cancer Society. Kyoto, Japan
:
Int J Gyne Cancer
2018
.
61.
Sloan
EA
,
Ring
KL
,
Willis
BC
,
Modesitt
SC
,
Mills
AM
. 
PD-L1 expression in mismatch repair-deficient endometrial carcinomas, including lynch syndrome-associated and MLH1 promoter hypermethylated tumors
.
Am J Surg Pathol
2017
;
41
:
326
33
.
62.
Andersen
MH
. 
The specific targeting of immune regulation: T-cell responses against Indoleamine 2,3-dioxygenase
.
Cancer Immunol Immunother
2012
;
61
:
1289
97
.
63.
Asghar
K
,
Farooq
A
,
Zulfiqar
B
,
Rashid
MU
. 
Indoleamine 2,3-dioxygenase: as a potential prognostic marker and immunotherapeutic target for hepatocellular carcinoma
.
World J Gastroenterol
2017
;
23
:
2286
93
.
64.
Bjoern
J
,
Iversen
TZ
,
Nitschke
NJ
,
Andersen
MH
,
Svane
IM
. 
Safety, immune and clinical responses in metastatic melanoma patients vaccinated with a long peptide derived from indoleamine 2,3-dioxygenase in combination with ipilimumab
.
Cytotherapy
2016
;
18
:
1043
55
.
65.
Ino
K
,
Yoshida
N
,
Kajiyama
H
,
Shibata
K
,
Yamamoto
E
,
Kidokoro
K
, et al
Indoleamine 2,3-dioxygenase is a novel prognostic indicator for endometrial cancer
.
Br J Cancer
2006
;
95
:
1555
61
.
66.
Moon
YW
,
Hajjar
J
,
Hwu
P
,
Naing
A
. 
Targeting the indoleamine 2,3-dioxygenase pathway in cancer
.
J Immunother Cancer
2015
;
3
:
51
.
67.
Mills
A
,
Zadeh
S
,
Sloan
E
,
Chinn
Z
,
Modesitt
SC
,
Ring
KL
. 
Indoleamine 2,3-dioxygenase in endometrial cancer: a targetable mechanism of immune resistance in mismatch repair-deficient and intact endometrial carcinomas
.
Mod Pathol
2018
;
31
:
1282
90
.
68.
Meireson
A
,
Chevolet
I
,
Hulstaert
E
,
Ferdinande
L
,
Ost
P
,
Geboes
K
, et al
Peritumoral endothelial indoleamine 2, 3-dioxygenase expression is an early independent marker of disease relapse in colorectal cancer and is influenced by DNA mismatch repair profile
.
Oncotarget
2018
;
9
:
25216
24
.
69.
Volaric
A
,
Gentzler
R
,
Hall
R
,
Mehaffey
JH
,
Stelow
EB
,
Bullock
TN
, et al
Indoleamine-2,3-dioxygenase in non-small cell lung cancer: a targetable mechanism of immune resistance frequently coexpressed with PD-L1
.
Am J Surg Pathol
2018
;
42
:
1216
23
.
70.
Mills
AM
,
Peres
LC
,
Meiss
A
,
Ring
KL
,
Modesitt
SC
,
Abbott
SE
, et al
Targetable immune regulatory molecule expression in high-grade serous ovarian carcinomas in African American women: a study of PD-L1 and IDO in 112 cases from the African American Cancer Epidemiology Study (AACES)
.
Int J Gynecol Pathol
2018 Feb 26
[Epub ahead of print]
.
71.
Camp
RL
,
Neumeister
V
,
Rimm
DL
. 
A decade of tissue microarrays: progress in the discovery and validation of cancer biomarkers
.
J Clin Oncol
2008
;
26
:
5630
7
.
72.
Koshiol
J
,
Lin
SW
. 
Can tissue-based immune markers be used for studying the natural history of cancer?
Ann Epidemiol
2012
;
22
:
520
30
.