Background: The presence of CD8+ tumor-infiltrating lymphocytes (TIL) is associated with prolonged survival in high-grade serous ovarian cancer (HGSC) and other epithelial cancers. Survival is most strongly associated with intraepithelial versus intrastromal CD8+ TILs; however, the mechanisms that promote the intraepithelial localization of TILs remain poorly understood. We hypothesized that intraepithelial CD8+ TILs, like normal mucosal intraepithelial lymphocytes, might express CD103, a subunit of αE7 integrin, which binds E-cadherin on epithelial cells.

Methods: A large collection of primary ovarian tumors (HGSC, endometrioid, mucinous, and clear cell) was analyzed by immunohistochemistry for the presence of TIL-expressing CD103. The activation and differentiation status of CD103+ TILs were assessed by flow cytometry. The prognostic significance of TIL subsets was evaluated by Kaplan–Meier analysis.

Results: CD103+ TILs were present in all major ovarian cancer subtypes and were most abundant in HGSC. CD103+ TILs were preferentially localized to epithelial regions of tumors and were comprised predominantly of CD8+ T cells expressing activation (HLA-DR, Ki-67, PD-1) and cytolytic (TIA-1) markers, as well as CD56+ NK cells. Tumor infiltration by CD103+ TILs was strongly associated with patient survival in HGSC. Tumors containing CD8+ TILs that were CD103 showed poor prognosis equivalent to tumors lacking CD8+ TILs altogether.

Conclusions: CD103+ TILs comprise intraepithelial, activated CD8+ T cells, and NK cells and are strongly associated with patient survival in HGSC. CD103 may serve as a useful marker for enriching the most beneficial subsets of TILs for immunotherapy. Clin Cancer Res; 20(2); 434–44. ©2013 AACR.

Translational Relevance

The immune system has a strong influence on clinical outcomes in cancer, as evidenced by the association between tumor-infiltrating lymphocytes (TIL) and patient survival. TILs actively enter the malignant epithelium of tumors, where they are thought to mount cytolytic responses against tumor cells. The mechanisms that promote the intraepithelial localization of TILs remain poorly understood. We addressed this issue in ovarian cancer, an aggressive malignancy in which CD8+ TILs correlate with survival. We show that the subset of CD8+ TILs localized to tumor epithelium expresses CD103 (αE7 integrin), a molecule commonly found on normal mucosal intraepithelial lymphocytes and tissue-resident memory T cells. CD8+ TILs that express CD103 have a highly activated and cytolytic phenotype and are strongly associated with patient survival. Thus, CD103 demarcates the subset of CD8+ TILs that mediates protective tumor immunity. CD103 may serve as a useful marker to enrich these cells for cancer immunotherapy.

Epithelial ovarian cancer (EOC) is the fifth most frequent cause of death due to cancer in women worldwide (1). The overall 5-year survival rate is 44% (2) and depends on stage at diagnosis, histologic subtype, and extent of surgical debulking. In addition, a number of studies have demonstrated that the presence of tumor-infiltrating lymphocytes (TIL) in surgically resected tumor tissue correlates with a favorable prognosis, particularly in patients with high-grade serous ovarian cancer (HGSC). For example, in a landmark study by Coukos and colleagues, retrospective analysis of 186 stage III–IV EOC patients revealed that the 5-year overall survival rate was 38% for patients with CD3+ tumor-infiltrating lymphocyte (TIL)-positive tumors, compared with only 4.5% for CD3+ TIL-negative tumors (3). Follow-up studies revealed that the CD8+ subset of TILs correlates most strongly with survival (4). Moreover, although TILs are found in all EOC subtypes, their beneficial effects are most pronounced in HGSC (5). Importantly, the prognostic effect is strongest for CD8+ TILs that are localized within the epithelial component of tumors rather than the associated stroma (5). Despite this association, the mechanisms that promote the localization of TILs to tumor epithelium remain poorly understood.

Retention of T cells within epithelial tissues under a variety of normal and pathologic conditions is known to be mediated, at least in part, through expression of the αE(CD103)/β7 integrin molecule (referred to hereafter as CD103; ref. 6). In healthy individuals, CD103 is expressed on less than 2% of circulating peripheral blood lymphocytes but widely expressed on intraepithelial lymphocytes (IEL) of the gut mucosa and skin (7, 8). The only known ligand for CD103 is the epithelial cell surface molecule E-cadherin and adhesive interactions between CD103 and E-cadherin are thought to be responsible for retention of antigen-specific lymphocytes within epithelial tissues (6). Interestingly, CD103 is also expressed by tissue-infiltrating, allo-reactive CD8+ T cells during transplant rejection (9) and graft-versus-host disease (10). In the infectious disease setting, CD103 is increasingly being recognized as a definitive marker of “tissue resident memory” CD8+ T cells (CD8+ TRM; ref. 11). Virus-specific CD8+ TRM have been found in tissues such as skin (12), sensory ganglia (13), brain (14), lung (15), and tonsils (16). CD8+ TRM are stably retained at the site of primary infection for long periods of time after disease resolution, even in the complete absence of persisting antigen (17) and are thought to play a pivotal role in conferring immunity against re-exposure to the same pathogen (13, 18).

Recent evidence suggests that CD103 also plays an important role in immunity against cancer, and in particular, cancers of epithelial origin (19–23). For example, CD103:E-cadherin interactions have been implicated in the recognition of tumor cells by cytotoxic T cells in colon (19), lung (20), and pancreatic cancer (21). We recently reported that the ascites compartment of some patients with HGSC contains large numbers of CD8+CD103+ T cells with an effector memory phenotype (24). CD103 is reported to be recruited to the immunologic synapse during tumor cell recognition, where it mediates adhesive interactions with E-cadherin on the target tumor cell, resulting in polarization of lytic granules and potentiated cytotoxic activity (20). In a lung cancer xenograft model, CD103 surface expression was upregulated on TILs after migration to the tumor and coengagement of TCR and TGF-β receptors at that site (25). Together, these findings lead us to hypothesize that CD103 expression might demarcate intraepithelial, tumor-reactive TILs that confer favorable prognosis in epithelial cancers.

Using a new anti-CD103 antibody that works with formalin-fixed, paraffin-embedded tissues (26), we have performed the first analysis of the intratumoral location and prognostic influence of CD103+ TILs in human cancer. We show herein that CD103 is widely expressed by TILs in human ovarian tumors and that it is preferentially expressed by intraepithelial versus stromal CD8+ TILs in both ovarian and breast tumors. In some patients, CD103 is also expressed by tumor-infiltrating NK cells. Importantly, CD103+ TILs are strongly associated with survival in patients with HGSC, indicating that CD103 demarcates a clinically beneficial TIL subset that warrants further analysis as a potential effector cell population for immunotherapy.

Patients

All specimens and clinical data were obtained with either informed written consent or a formal waiver of consent under protocols approved by the Research Ethics Board of the BC Cancer Agency (BCCA) and the University of British Columbia (Vancouver, British Columbia, Canada). The prospective cohort was comprised of HGSC patients admitted to the BCCA between August 2007 and August 2011. Patients were treated with primary cytoreductive surgery followed by chemotherapy with carboplatin with or without paclitaxel. Tumor tissue obtained during primary cytoreductive surgery was fixed in formalin and embedded in paraffin using standard methodologies. A tissue microarray was constructed with duplicate 1.0 mm cores from representative tumor areas using a manual tissue arrayer (Beecher Instruments). Immunohistochemical analyses were performed on either whole tumor sections or on tissue microarrays, as indicated.

The retrospective cohort used for survival analysis has been previously described (5). In brief, tumor samples were obtained from EOC cases of various histologic subtypes seen at the BC Cancer Agency from 1984 to 2000 (OvCaRe Ovarian Tumour Bank). All tumor tissues were obtained at the time of primary surgery and patients had no macroscopic residual disease following surgical cytoreduction. Table 1 describes this cohort in accordance with REMARK criteria. The breast tumor microarray utilized has also been previously described (27) and contained 24 primary breast tumors with no specific selection criteria.

Table 1.

CD103+ TILs in relation to clinical characteristics of the retrospective patient cohort (according to REMARK criteria)

Histologic subtypeCD103 TILs present (%)aMean number of CD103+ TILs per core (when present)b
Serous (N = 210)  172 (81.9) 46.4 
 DSS (y), median (95% CI) 6.9 (4.6–9.2)   
 Age (y), median (range) 60 (37.6–86)   
 Grade 
  1 12 6 (50.0) 4.2 
  2 56 41 (73.2) 42.9 
  3 142 125 (88.0) 49.6 
 FIGO stage 
  I 50 44 (88.0) 43.4 
  II 91 72 (79.1) 55.0 
  III 69 56 (81.2) 37.7 
Mucinous (N = 31)  19 (61.3) 8.6 
 DSS (y), median (95% CI) Not reached   
 Age (y), median (range) 55.0 (25.4–82.5)   
 Grade 
  1 11 6 (54.5) 11.3 
  2 18 11 (61.1) 8.3 
  3 2 (100) 2.8 
 FIGO stage 
  I 18 11 (61.1) 10.8 
  II 12 7 (58.3) 6.0 
  III 1 (100.0) 3.8 
Endometrioid (N = 125)  87 (69.6) 21.6 
 DSS (y), median (95% CI) Not reached   
 Age (y), median (range) 53.9 (29.4–88.1)   
 Grade 
  1 82 60 (73.2) 16.3 
  2 35 20 (57.1) 21.2 
  3 7 (87.5) 67.9 
 FIGO stage 
  I 69 53 (76.8) 20.5 
  II 50 32 (64) 24.4 
  III 2 (33.3) 5.5 
Clear cell (N = 131)  61 (46.6) 16.0 
 DSS (y), median (95% CI) Not reached   
 Age (y), median (range) 54.8 (28.1–89.0)   
 Grade 
  1  —  
  2  —  
  3 131 61 (46.6) 16.0 
 FIGO stage 
  I 67 30 (44.8) 11.8 
  II 56 28 (50.0) 21.3 
  III 2 (25.0) 2.4 
Histologic subtypeCD103 TILs present (%)aMean number of CD103+ TILs per core (when present)b
Serous (N = 210)  172 (81.9) 46.4 
 DSS (y), median (95% CI) 6.9 (4.6–9.2)   
 Age (y), median (range) 60 (37.6–86)   
 Grade 
  1 12 6 (50.0) 4.2 
  2 56 41 (73.2) 42.9 
  3 142 125 (88.0) 49.6 
 FIGO stage 
  I 50 44 (88.0) 43.4 
  II 91 72 (79.1) 55.0 
  III 69 56 (81.2) 37.7 
Mucinous (N = 31)  19 (61.3) 8.6 
 DSS (y), median (95% CI) Not reached   
 Age (y), median (range) 55.0 (25.4–82.5)   
 Grade 
  1 11 6 (54.5) 11.3 
  2 18 11 (61.1) 8.3 
  3 2 (100) 2.8 
 FIGO stage 
  I 18 11 (61.1) 10.8 
  II 12 7 (58.3) 6.0 
  III 1 (100.0) 3.8 
Endometrioid (N = 125)  87 (69.6) 21.6 
 DSS (y), median (95% CI) Not reached   
 Age (y), median (range) 53.9 (29.4–88.1)   
 Grade 
  1 82 60 (73.2) 16.3 
  2 35 20 (57.1) 21.2 
  3 7 (87.5) 67.9 
 FIGO stage 
  I 69 53 (76.8) 20.5 
  II 50 32 (64) 24.4 
  III 2 (33.3) 5.5 
Clear cell (N = 131)  61 (46.6) 16.0 
 DSS (y), median (95% CI) Not reached   
 Age (y), median (range) 54.8 (28.1–89.0)   
 Grade 
  1  —  
  2  —  
  3 131 61 (46.6) 16.0 
 FIGO stage 
  I 67 30 (44.8) 11.8 
  II 56 28 (50.0) 21.3 
  III 2 (25.0) 2.4 

Abbreviations: DSS, disease-specific survival; 95% CI, confidence interval.

aTumors with a mean of ≥1 CD103-positive cells per 0.6 mm core (2 duplicate cores, each counted once by two independent investigators).

bMean number of CD103-positive cells per 0.6 mm core in those tumors scored as positive in the previous column.

Immunohistochemistry

To detect CD103+ cells, 4 μm sections of either whole tumor tissue or tissue microarrays were deparaffinized and subjected to heat-induced epitope retrieval using a decloaking chamber and Diva decloaking reagent (Biocare Medical). Anti-CD103 rabbit monoclonal antibody (Epitomics clone EPR4166(2)) was applied at a dilution of 1/1,000 for 30 minutes at room temperature. Following washing, anti-rabbit-HRP polymer reagent was applied for 30 minutes at room temperature followed by 3,3′-diaminobenzidine (DAB) reagent. The slides were counterstained with hematoxylin, dehydrated, and cover-slipped using Cytoseal 60.

For two-color immunohistochemical staining, sections were stained with rabbit anti-CD103 as described above in combination with a murine primary antibody against either CD8 (C8/144B) or CD56 (123C3.D5). Slides were incubated with Mach 2 Double Stain #1 polymer reagent (Biocare) for 30 minutes at room temperature, and the horseradish peroxidase (HRP), and alkaline phospatase (AP) enzyme tags were detected using Vina Green and Warp Red chromogens (Biocare), respectively. Slides were counterstained with hematoxylin.

For the CD103/CD8/cytokeratin triple stain, rabbit anti-CD103 and murine anti-CD8 antibodies were added for 30 minutes at room temperature followed by Mach 2 Double Stain 2 polymer for 30 minutes. CD103 was detected with DAB chromogen and CD8 was detected with Warp Red. Slides were subjected to a denaturation protocol followed by a second round of staining with pan-Cytokeratin antibody (clone PCK-26, Sigma-Aldrich). Slides were incubated with Mach 2 Mouse-AP polymer followed by Perma-Green Plus AP chromogen (Diagnostic Biosystems).

Slides were analyzed using the Nuance Multispectral Imaging System (CRi) with a BX-53 microscope (Olympus). Single-stained slides were used to create spectral profiles for each chromogen and these profiles were then used to discriminate the individual colours on multistained slides. False color images were generated using Metamorph software (Molecular Devices).

Flow cytometry

Single-cell suspensions were prepared from freshly excised tumor specimens and cryopreserved, as described (28). After thawing, cells were stained with the following fluorescently conjugated antibodies: CD3-BV510, PD-1-PECy7 (Biolegend), CD103-PECy7 (Ebioscience), CD8-BV421, CD103-PE, HLA-DR-PE, CD127-AF647, CD56-PE/APC, CD4 FITC, and Ki67-FITC (BD Biosciences). All flow cytometry was performed using a FACS Calibur that had been upgraded to 8-color capability by Cytek (DxP8). Analyses were performed using FlowJo software (Tree Star).

Scoring and statistical analysis

Scoring for epithelial localization of CD8 and CD103 markers was undertaken by an experienced histopathologist (P.H. Watson) blinded to results of other markers or case identity. Positively stained cells that had morphologic features compatible with lymphocytes were counted according to two localization patterns: (i) intraepithelial infiltrates, defined as TILs within cancer cell nests or in direct contact with cancer cells; and (ii) stromal infiltrates, defined as TILs within stromal areas that lack direct contact with epithelial cells. For prognostic analyses, absolute numbers of CD103+ TILs (independent of location) in cores of the retrospective cohort TMA were enumerated by visual inspection. Duplicate cores were counted independently by two different investigators and the average cell count per core was used for statistical analyses. Statistical analyses and HR calculations were performed using GraphPad software. Univariate survival analysis was performed using the Kaplan–Meier method and P values were determined using log-rank test. Multivariate survival analysis was performed using a Cox regression model. P values <0.05 were regarded as significant.

CD103 is preferentially expressed by intraepithelial TILs

To assess the frequency and distribution of CD103+ TILs in primary ovarian tumor samples, we used a recently developed anti-CD103 antibody (26) to stain a previously described tissue microarray (5) containing cores from a cohort of 497 EOC solid tumor specimens. CD103+ TILs were detectable in tumors of all stages and grades and in all four major EOC subtypes (Fig. 1; Table 1). In all tumor subtypes, CD103+ TILs seemed to be preferentially localized to the epithelial regions of tumors (Fig. 1A). The density of CD103+ TILs within each subtype ranged from negligible to heavy infiltration (Fig. 1B) and the highest mean frequency of CD103+ TILs was seen in serous, followed by the endometrioid, clear cell, and mucinous subtypes, respectively (Fig. 1B; Table 2).

Figure 1.

CD103 is expressed by TILs in the major subtyes of ovarian cancer. A, a retrospective TMA containing 497 ovarian cancer cases comprising all four major ovarian cancer tumor types (serous, endometrioid, mucinous and clear cell) was stained with anti-CD103 antibody and counterstained with hematoxylin. Representative image of each tumor subtype is shown. B, cluster plot showing the distribution of CD103+ TILs within EOC tumors of the indicated histologic subtype. The mean CD103+ TILs density for each subtype is indicated by a horizontal line.

Figure 1.

CD103 is expressed by TILs in the major subtyes of ovarian cancer. A, a retrospective TMA containing 497 ovarian cancer cases comprising all four major ovarian cancer tumor types (serous, endometrioid, mucinous and clear cell) was stained with anti-CD103 antibody and counterstained with hematoxylin. Representative image of each tumor subtype is shown. B, cluster plot showing the distribution of CD103+ TILs within EOC tumors of the indicated histologic subtype. The mean CD103+ TILs density for each subtype is indicated by a horizontal line.

Close modal
Table 2.

Disease-specific survival and HR based upon the presence of CD103+ TILs

CD103 densityaMedian survival (days; absolute cutoff approach)Median survival (days; lowest tertile approach)
NMeanMedianRangeLowest tertile<5 CD103+≥5 CD103+HRb95% CIPcLowest tertileAll othersHRb95% CIPc
HGSC 198 40.19 20.8 0–310 7.5 1842 3928 0.4757 0.263–0.674 0.0003 1842 4432 0.4344 0.239–0.603 <0.0001 
Mucinous 31 5.34 2.0 0–36 0.5 ** ** 0.2245 0.051–0.988 0.0481 ** ** 0.7651 0.190–3.103 0.7122 
Endometrioid 125 15.07 3.8 0–391 1.0 ** ** 0.4912 0.212–1.231 0.1357 ** ** 0.4340 0.138–1.011 0.0542 
Clear cell 131 7.61 0.5 0–133 ** 6407 0.7682 0.368–1.657 0.5221 2825 6407 0.5704 0.281–1.040 0.0688 
CD103 densityaMedian survival (days; absolute cutoff approach)Median survival (days; lowest tertile approach)
NMeanMedianRangeLowest tertile<5 CD103+≥5 CD103+HRb95% CIPcLowest tertileAll othersHRb95% CIPc
HGSC 198 40.19 20.8 0–310 7.5 1842 3928 0.4757 0.263–0.674 0.0003 1842 4432 0.4344 0.239–0.603 <0.0001 
Mucinous 31 5.34 2.0 0–36 0.5 ** ** 0.2245 0.051–0.988 0.0481 ** ** 0.7651 0.190–3.103 0.7122 
Endometrioid 125 15.07 3.8 0–391 1.0 ** ** 0.4912 0.212–1.231 0.1357 ** ** 0.4340 0.138–1.011 0.0542 
Clear cell 131 7.61 0.5 0–133 ** 6407 0.7682 0.368–1.657 0.5221 2825 6407 0.5704 0.281–1.040 0.0688 

NOTE: Values shown in bold are statistically significant (P < 0.05).

aBased on the mean number of CD103+ TILs per 0.6 mm core (average of 2 cores), regardless of location within tumor.

bHR calculated using log-rank.

cP value using log-rank (Mantel–Cox) test.

**, Not reached.

Focusing on the HGSC (grades 2 and 3), we then quantified the intratumoral location of CD103+ TILs as epithelial or stromal. Serial sections of a prospective HGSC TMA were stained with hematoxylin and eosin (H&E), anti-CD8 or anti-CD103 and localization was evaluated by an experienced pathologist. There was a statistically significant bias in the localization of CD103+ versus CD8+ TILs to tumor epithelium with an average epithelial:stromal density ratio of 2.29 for CD103+ TILs versus 1.36 for CD8+ TILs (P = 0.008 by the Wilcoxon test, n = 34; Fig. 2A). The biased intraepithelial localization was particularly evident in those tumors with denser CD103+ TIL infiltration.

Figure 2.

CD103+ TILs are preferentially localized within the intraepithelial regions of HGSC tumors. A, serial sections of a HGSC prospective TMA were stained with (1) hematoxylin and eosin, (2) anti-CD103, or (3) anti-CD8 antibodies and the density of CD103+ and CD8+ cells within epithelial versus stromal regions was scored by a pathologist. The graph depicts a pairwise comparison of the density of CD103+ and CD8+ TILs in epithelial versus stromal regions of each tumor and statistical comparison using the Wilcoxon test. B, a representative HGSC sample was triple-stained with antibodies against CD8 (red), CD103 (green), and cytokeratin (blue). The image was false-colored to highlight cells that were double positive for CD8 and CD103 (yellow). The majority of CD103+ TILs were found in epithelial (cytokeratin-positive) regions. C, Representative example of human breast tumor tissue stained with anti-CD103 antibody showing localization of CD103+ TIL within epithelial regions.

Figure 2.

CD103+ TILs are preferentially localized within the intraepithelial regions of HGSC tumors. A, serial sections of a HGSC prospective TMA were stained with (1) hematoxylin and eosin, (2) anti-CD103, or (3) anti-CD8 antibodies and the density of CD103+ and CD8+ cells within epithelial versus stromal regions was scored by a pathologist. The graph depicts a pairwise comparison of the density of CD103+ and CD8+ TILs in epithelial versus stromal regions of each tumor and statistical comparison using the Wilcoxon test. B, a representative HGSC sample was triple-stained with antibodies against CD8 (red), CD103 (green), and cytokeratin (blue). The image was false-colored to highlight cells that were double positive for CD8 and CD103 (yellow). The majority of CD103+ TILs were found in epithelial (cytokeratin-positive) regions. C, Representative example of human breast tumor tissue stained with anti-CD103 antibody showing localization of CD103+ TIL within epithelial regions.

Close modal

As a complementary approach, we performed triple staining of HGSC tumors to simultaneously detect CD8, CD103, and cytokeratin, the latter being a well-established marker of tumor epithelium. Consistent with the above results, CD8+CD103+ TILs were preferentially localized to cytokeratin-positive regions of the tumor (Fig. 2B). In addition, we evaluated the localization of CD103+ TILs in breast cancer, where epithelial and stromal regions are typically more distinct than in HGSC. CD103+ TILs were present in 5 of 20 invasive breast cancer specimens analyzed, and they again showed preferential localization to tumor epithelium (Fig. 2C). Thus, as seen with normal mucosal IEL, CD103 is preferentially expressed by “intraepithelial” TILs in ovarian and breast cancer.

The only known ligand for CD103 is the epithelial surface molecule E-cadherin (6). Consistent with prior reports (29), E-cadherin expression was variable across HGSC tumors, ranging from negative to strong expression (Fig. 3). However, there was no obvious correlation between E-cadherin staining intensity and the presence of CD103+ TILs (P = 0.667, Pearson test, n = 60). Thus, infiltration by CD103+ TILs is determined by factors other than E-cadherin expression.

Figure 3.

CD103+ TILs do not correlate with E-cadherin expression by tumors. Serial sections of a HGSC prospective TMA were stained antibodies to E-cadherin (left) or CD103 (right). Representative examples are shown of tumors with low (top), intermediate (middle), and high (bottom) densities of CD103+ TILs.

Figure 3.

CD103+ TILs do not correlate with E-cadherin expression by tumors. Serial sections of a HGSC prospective TMA were stained antibodies to E-cadherin (left) or CD103 (right). Representative examples are shown of tumors with low (top), intermediate (middle), and high (bottom) densities of CD103+ TILs.

Close modal

CD103+ TILs are comprised of activated CD8+ T cells and CD56+ NK cells

To determine the lineage and activation status of CD103+ TILs, multiparameter flow cytometry was performed on disaggregated tumor preparations from 6 HGSC solid tumor specimens (Fig. 4A). In most patients, the majority of CD103+ TILs were CD3+CD8+ T lymphocytes. The second most common subset of CD103+ TILs was comprised of CD3CD56+ NK cells. In some tumors, we also observed small numbers of CD3+CD4+CD8- and CD3+CD8-CD4- T cells among CD103+ TILs (Fig. 4A). CD103 staining was not detected on CD20+ B cells, FOXP3+ lymphocytes or CD14+ monocytes; CD11c+ DC were too rare to assess (data not shown). The presence of tumor-infiltrating CD103+CD8+ and CD103+CD56+ lymphocytes was confirmed by multiparameter IHC using antibodies against CD103, CD8, and CD56 (Fig. 4B). Consistent with the flow cytometry results, both CD8+ and CD56+ cells expressing CD103 were readily detectable by IHC and were localized primarily to tumor epithelium.

Figure 4.

CD103 is expressed predominantly by intraepithelial CD8+ T cells and CD56+ NK cells in HGSC. A, FACS analysis of CD103+ TILs in primary ovarian tumors. Tumor tissue obtained from 6 HGSC patients at the time of initial debulking surgery was enzymatically dissociated into single-cell suspension and stained with antibodies to CD103, CD3, CD4, CD8, and CD56, followed by flow cytometry. Events were first gated on CD103+ TIL populations and then sequentially into the indicated subpopulations. B, immunohistochemical analysis of CD103+ TILs in primary ovarian tumors. Sections of two representative HGSC tumors were dual-stained with antibodies to CD8 (red) and CD103 (green; top), or CD103 (red), and CD56 (green; bottom). Images were false colored to highlight cells that are positive for both markers (yellow). Results are representative examples from over 50 HGSC tumors.

Figure 4.

CD103 is expressed predominantly by intraepithelial CD8+ T cells and CD56+ NK cells in HGSC. A, FACS analysis of CD103+ TILs in primary ovarian tumors. Tumor tissue obtained from 6 HGSC patients at the time of initial debulking surgery was enzymatically dissociated into single-cell suspension and stained with antibodies to CD103, CD3, CD4, CD8, and CD56, followed by flow cytometry. Events were first gated on CD103+ TIL populations and then sequentially into the indicated subpopulations. B, immunohistochemical analysis of CD103+ TILs in primary ovarian tumors. Sections of two representative HGSC tumors were dual-stained with antibodies to CD8 (red) and CD103 (green; top), or CD103 (red), and CD56 (green; bottom). Images were false colored to highlight cells that are positive for both markers (yellow). Results are representative examples from over 50 HGSC tumors.

Close modal

We also compared the activation and differentiation status of CD103 and CD103+ TILs by flow cytometry. Figure 5 shows representative plots from tumors in which the CD103+ TIL population was predominantly CD8+ T cells (Case #014) or a mixture of CD8+ T cells and NK cells (Case #042). Consistent with the above observation that most CD103+ TILs were either CD8+ T cells or CD56+ NK cells, CD103+ TIL cells universally expressed TIA-1, a marker of cytolytic potential. Compared with their CD103- counterparts, CD103+CD8+ TILs had markedly increased expression of the activation markers HLA-DR, Ki-67, and PD-1 and lower levels of IL-7 receptor α chain (CD127).

Figure 5.

Cell surface phenotype of CD103+ TILs in HGSC tumors. Tumor tissue obtained from two HGSC patients at the time of initial debulking surgery was enzymatically dissociated into single-cell suspension and stained with antibodies to CD103 plus the indicated markers, followed by flow cytometry. Top panels are gated on the FSC/SSC lymphocyte gate. Bottom panels are gated on CD8+ lymphocytes.

Figure 5.

Cell surface phenotype of CD103+ TILs in HGSC tumors. Tumor tissue obtained from two HGSC patients at the time of initial debulking surgery was enzymatically dissociated into single-cell suspension and stained with antibodies to CD103 plus the indicated markers, followed by flow cytometry. Top panels are gated on the FSC/SSC lymphocyte gate. Bottom panels are gated on CD8+ lymphocytes.

Close modal

CD103+ TILs are associated with increased survival in HGSC

We next assessed the relationship between CD103+ TILs and patient survival, using two distinct statistical approaches, both of which have previously been used to demonstrate the prognostic significance of TILs in EOC. The first approach used an absolute value (>5 CD103+ TILs per 0.6 mm core) to stratify the cohort into infiltrated versus noninfiltrated subsets, similar to as has been described previously (3, 4). Using this strategy, the presence of CD103+ TILs correlated with increased disease-specific survival in HGSC (P = 0.0003) and mucinous EOC (P = 0.0481), but not endometrioid or clear cell cancers (Table 2; Fig. 6A). The second approach relied upon the quantitative characteristics of the cohort to determine the cutoff value. As shown in Table 2, the distribution of CD103+ TIL infiltration was positively skewed (i.e., the mean was significantly higher than the median) for all 4 histologic subtypes. Similar patterns of positive skewing have been observed previously for other TIL subsets and has led to the use of the lowest tertile as an approach for stratifying cohorts into infiltrated versus noninfiltrated subsets (4). When the lowest tertile approach was used, the presence of CD103+ TILs again correlated with increased disease-specific survival in HGSC (P < 0.0001). However, using this strategy, CD103+ TIL infiltration did not reach statistical significance in mucinous, endometrioid, or clear cell EOC, likely due to the disproportionately low tertile values obtained in each of these subtypes (Table 2). CD103+ TIL infiltration remained a significant factor in multivariate survival analysis when age, stage, and grade were used as covariants (HR = 0.509; 95% confidence interval = 0.331–0.783; P = 0.002). Importantly, while previous studies have shown that only “intraepithelial” CD8+ TILs are associated with survival in HGSC, herein, TILs were scored solely on the basis of CD103 expression, which obviated the need to precisely account for their localization within the tumor.

Figure 6.

Prognostic value of CD103+ TILs in the four major subtypes of ovarian cancer. A, a retrospective TMA containing samples from 497 cases comprising the four major histologic subtypes of EOC was stained with anti-CD103 antibody. Total numbers of CD103+ TILs were determined without regard to their epithelial or stromal location. Data were binarized on the basis of a threshold of ≤5 or >5 CD103+ cells per core (average of 2 cores/case) followed by Kaplan–Meier analysis. The log-rank test was used to compare curves and P values less than 0.05 were considered significant. B, sequential sections from the TMA described in (A) were stained separately with antibodies to CD8 and CD103. Total numbers of CD8+ and CD103+ TILs (irrespective of location) were determined using a cutoff of ≤5 or >5 cells per core (average of 2 cores/case). Kaplan–Meier analysis was used to compare outcomes for patients with tumors that were positive for one, both, or neither marker.

Figure 6.

Prognostic value of CD103+ TILs in the four major subtypes of ovarian cancer. A, a retrospective TMA containing samples from 497 cases comprising the four major histologic subtypes of EOC was stained with anti-CD103 antibody. Total numbers of CD103+ TILs were determined without regard to their epithelial or stromal location. Data were binarized on the basis of a threshold of ≤5 or >5 CD103+ cells per core (average of 2 cores/case) followed by Kaplan–Meier analysis. The log-rank test was used to compare curves and P values less than 0.05 were considered significant. B, sequential sections from the TMA described in (A) were stained separately with antibodies to CD8 and CD103. Total numbers of CD8+ and CD103+ TILs (irrespective of location) were determined using a cutoff of ≤5 or >5 cells per core (average of 2 cores/case). Kaplan–Meier analysis was used to compare outcomes for patients with tumors that were positive for one, both, or neither marker.

Close modal

Finally, we attempted to determine whether CD103+ CD8 cells or CD103+ NK cells were primarily responsible for the observed prognostic effect in HGSC. Although it is technically feasible to perform three-color IHC with antibodies to CD8, CD56, and CD103, the resulting images are difficult to score reliably on the scale of a large TMA. Therefore, as an alternate approach, we stained serial sections of the TMA with antibodies to CD8 and CD103 and then scored tumors as positive for CD8+ and/or CD103+ TILs using a threshold of more than 5 cells per core. The majority of tumors were positive for both CD103+ and CD8+ TILs and, accordingly, were associated with favorable prognosis (Fig. 6B). In contrast, tumors that contained CD8+ TILs that lacked CD103 expression were associated with inferior survival, similar to those tumors lacking CD8+ TILs altogether, implying that it is the CD103+CD8+ TIL cells that are primarily responsible for improved prognosis. Moreover, very few tumors contained CD103+ TILs without CD8+ TILs; therefore, we were unable to precisely infer the contribution of CD103+ NK cells to prognosis.

CD8+ TILs are strongly associated with patient survival in a broad range of epithelial cancers (30); however, the mechanisms that promote their localization to tumor epithelium remain poorly understood. We examined the hypothesis that intraepithelial CD8+ TILs, like normal mucosal IEL and TRM, might express the αE7 integrin subunit CD103. Indeed, we found that CD103 expression is a common feature of intraepithelial CD8+ TILs in the four major subtypes of EOC. CD103+ TILs were found to have a highly activated, cytolytic phenotype, and included both CD8+ T cells and CD56+ NK cells. Consistent with their cytolytic phenotype, CD103+ TILs were strongly associated with survival in HGSC as well as mucinous EOC. Although CD103+CD8+ TILs have been reported in other epithelial cancers (19–23), to the best of our knowledge this is the first demonstration of their prognostic significance in any cancer. Our data indicates that CD103 demarcates a subset of TILs that seem to mediate protective antitumor immunity in HGSC and warrant further investigation as effector cells for immunotherapy.

In our analysis of 497 EOC tumors, the density of CD103+ TILs ranged widely, from 0 to more than 300 CD103+ TILs per tumor core. Although the underlying cause of this variability is unknown, earlier, in vitro studies have demonstrated that upregulation of CD103 on the surface of CD8+ T cells is dependent upon the concurrent stimulation of the TCR and TGF-β receptors (19, 31). Based upon this dual signal model, variability in CD103+ TIL density could therefore reflect inter-patient differences in the levels of TGF-β, cognate antigens, or both. Although TGF-β is widely considered an immunosuppressive factor, it can facilitate immune function in some physiologic contexts. For example, gut mucosal tissue contains resident IELs that upregulate CD103 expression in response to stimulation with cognate microbial Ag and locally expressed TGF-β (32). This culminates in the retention of IEL through adhesive interactions between αE(CD103)/β7 integrin and E-cadherin on gut epithelial cells (11). Accordingly, CD103 knockout mice show substantially reduced numbers of IEL in gut mucosa (33). Furthermore, allograft rejection involves infiltration of grafted tissues by CD8+ T cells, which express CD103 upon exposure to TGF-β in the local microenvironment (9). Importantly, allogeneic islet cell transplants show increased in vivo persistence in CD103-deficient mice due to impaired infiltration of T cells into graft tissue (34). Ovarian cancers have long been known to express TGF-β (35), and indeed TGF-β expression has been linked to metastatic potential (36) and to the process of epithelial to mesenchymal transition (37). Moreover, we previously reported that the malignant ascites of some patients with HGSC contains CD103-expressing CD8+ T cells, which correlate with the amount of TGF-β in the ascites fluid (24). Thus, the density of CD103+ TILs in solid tumors might reflect local TGF-β levels.

The fact that T cells also require antigen stimulation to upregulate expression of CD103 implies that CD103+ TILs may be responding to antigens in the tumor microenvironment. Although there is currently a paucity of well-characterized tumor antigens to test this hypothesis comprehensively in EOC, we previously showed that ascites-derived CD8+ T cells specific for the cancer-testis antigen NY-ESO-1 were predominantly CD103+ (24). The notion that CD103 is a marker of antigen-specific T cells is further supported by recent studies demonstrating CD103 expression by influenza-specific CD8+ T cells in human lung (15) and EBV-specific cells in human tonsil (16). CD103 is also expressed by virus-specific CD8+ cells in mouse brain after experimental infection with vesicular stomatitis virus (VSV; refs. 14, 18). In the above examples, CD103 demarcates tissue resident memory CD8+ T cells (CD8+ TRM) that persist at sites of antigen encounter, even long after pathogen clearance (for review see refs. 11, 38, 39). Finally, in the present study, it is noteworthy that CD103+ TILs express high levels of PD-1 (Fig. 5), which has been shown to be marker of tumor-reactive TILs in melanoma (40).

At present, the antigens recognized by the vast majority of TILs in ovarian cancer remain poorly defined. Based upon the evidence described above, we speculate that CD103 may specifically demarcate TILs that are tumor-specific and, as such, may serve as a useful marker for enriching tumor-reactive TILs for antigen discovery efforts. On the other hand, given that CD103 is also a marker by TRM, it is also conceivable that CD103+CD8+ TILs are specific for pathogen antigens rather than tumor antigens. This possibility is consistent with the recent hypothesis that many “ovarian” cancers actually originate in the fallopian tubes (41), which constitute a barrier tissue that is continuously exposed to various microbial agents. Indeed, similar to other mucosal tissues, the mucosal surface of the female genital tract is known to harbor intraepithelial T cells that express CD103 (42). Thus, it is plausible that at least some of the CD103+ TILs in EOC may represent pathogen-specific TRM that are remnants of prior immune response(s) in the fallopian tubes or neighboring mucosal tissues. However, this alternative hypothesis needs to be reconciled with the strong prognostic effect of CD103+ TILs observed herein, which is indicative of tumor-specificity rather than pathogen specificity.

In addition to its potential utility as both a prognostic marker and a tool for antigen discovery, CD103 provides a convenient cell surface marker for investigating the functional phenotype of intraepithelial TILs. Previously, CD103+CD8+ T cells have been shown to have regulatory/immunosuppressive properties, at least in some settings (43). Moreover, CD103 is often expressed on conventional regulatory CD4 T cells (Treg; ref. 44). Although Tregs are known to influence outcomes in ovarian cancer (4), we did not detect significant numbers of CD103-expressing Tregs during our analyses. In contrast, we demonstrate herein that, compared with their CD103 counterparts, CD103+CD8+ TILs express higher levels of the activation markers HLA-DR, PD-1, and Ki-67 and lower levels of the naïve T-cell marker CD127. Most strikingly, virtually all CD103+ TILs expressed TIA-1, a marker of cytolytic potential that is strongly associated with survival in EOC (45) and breast cancer (46). The strong association between CD103+ TILs and patient survival, together with their universal expression of the cytolytic marker TIA-1, suggests that CD103 demarcates a subset of highly activated, cytolytic CD8+ TILs. However, the expression of PD-1 by CD103+CD8+ TILs could also signify functional exhaustion (47). Indeed, one can imagine that CD103+ TILs might be trapped within the tumor bed through interactions with E-cadherin expressed on tumor cells, and that they subsequently become exhausted due to chronic antigen stimulation. Indeed, the fact that these cells express high levels of PD-1 suggests that their antitumor activity might be further enhanced through blockade of PD-1 or other inhibitory pathways.

Our finding that CD103 is preferentially expressed by intraepithelial TILs also has practical implications for TILs scoring. In particular, a worldwide task force is being assembled to validate the prognostic significance of TILs as defined by the so-called Immunoscore (48). The ultimate goal of this effort is to bring the Immunoscore into standard clinical practice as a prognostic biomarker. Colorectal cancer is likely to be the first indication, but other tumor sites are being considered. It is often difficult to distinguish between epithelial versus stromal compartments of tumors, especially for nonexperts. Our results suggest that immunohistochemical detection of CD103+ TILs may provide an objective surrogate means to score intraepithelial TILs. Indeed, we showed a strong correlation between CD103+ TILs and survival in HGSC without the need to discriminate their epithelial versus stromal location (Fig. 6A).

Although the majority of CD103+ TILs observed herein were CD8+ T cells, the next largest subset was comprised of CD56+ NK cells. Like CD8+ T cells, NK cells express CD103 when exposed to TGF-β (49); however, it is unknown whether they require a second signal equivalent to antigen stimulation. In healthy individuals, CD103+CD56+ NK cells are found in microenvironments rich in TGF-β, including uterine decidua (50) and tonsil. Very little is known about the functional profile or prognostic significance of CD3CD56+ NK cells in HGSC. To date, greater attention has been focused on CD56+CD3+ NKT-like cells, which correlate with poor outcome as defined by platinum resistance (51). Because CD103+ NK cells were almost always found together with CD103+CD8+ TILs, we were unable to precisely define their independent prognostic significance. However, given the strong association between CD103+ TILs and survival, further study regarding the role of CD103+CD56+ TILs in tumor immunity is clearly warranted.

In closing, the strong association between intraepithelial CD8+ TILs and patient survival has spawned great interest in the use of TILs as predictive and prognostic biomarkers and as effector cells for immunotherapy. We propose that CD103, by serving as a definitive marker of these cells, has the potential to dramatically increase our understanding of TIL function, antigen specificity and immunotherapeutic potential, opening new avenues in our understanding of tumor immunity in EOC and other cancers.

No potential conflicts of interest were disclosed.

Conception and design: J.R. Webb, B.H. Nelson

Development of methodology: J.R. Webb, K. Milne, R.J. deLeeuw

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.R. Webb, K. Milne, R.J. deLeeuw, B.H. Nelson

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.R. Webb, K. Milne, P. Watson, R.J. deLeeuw, B.H. Nelson

Writing, review, and/or revision of the manuscript: J.R. Webb, K. Milne, P. Watson, R.J. deLeeuw, B.H. Nelson

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K. Milne

Study supervision: 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.
Jemal
A
,
Siegel
R
,
Ward
E
,
Murray
T
,
Xu
J
,
Smigal
C
, et al
Cancer statistics, 2006
.
CA Cancer J Clin
2006
;
56
:
106
30
.
2.
Cancer Facts & Figures 2013
.
Atlanta, GA
:
American Cancer Society
; 
2013
.
3.
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
.
4.
Sato
E
,
Olson
SH
,
Ahn
J
,
Bundy
B
,
Nishikawa
H
,
Qian
F
, et al
Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer
.
Proc Natl Acad Sci U S A
2005
;
102
:
18538
43
.
5.
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
.
6.
Gorfu
G
,
Rivera-Nieves
J
,
Ley
K
. 
Role of beta7 integrins in intestinal lymphocyte homing and retention
.
Curr Mol Med
2009
;
9
:
836
50
.
7.
Cerf-Bensussan
N
,
Jarry
A
,
Brousse
N
,
Lisowska-Grospierre
B
,
Guy-Grand
D
,
Griscelli
C
. 
A monoclonal antibody (HML-1) defining a novel membrane molecule present on human intestinal lymphocytes
.
Eur J Immunol
1987
;
17
:
1279
85
.
8.
Kruschwitz
M
,
Fritzsche
G
,
Schwarting
R
,
Micklem
K
,
Mason
DY
,
Falini
B
, et al
Ber-ACT8: new monoclonal antibody to the mucosa lymphocyte antigen
.
J Clin Pathol
1991
;
44
:
636
45
.
9.
Hadley
GA
,
Bartlett
ST
,
Via
CS
,
Rostapshova
EA
,
Moainie
S
. 
The epithelial cell-specific integrin, CD103 (alpha E integrin), defines a novel subset of alloreactive CD8 +CTL
.
J Immunol
1997
;
159
:
3748
56
.
10.
El-Asady
R
,
Yuan
R
,
Liu
K
,
Wang
D
,
Gress
RE
,
Lucas
PJ
, et al
TGF-{beta}-dependent CD103 expression by CD8(+) T cells promotes selective destruction of the host intestinal epithelium during graft-versus-host disease
.
J Exp Med
2005
;
201
:
1647
57
.
11.
Sheridan
BS
,
Lefrancois
L
. 
Regional and mucosal memory T cells
.
Nat Immunol
2011
;
12
:
485
91
.
12.
Jiang
X
,
Clark
RA
,
Liu
L
,
Wagers
AJ
,
Fuhlbrigge
RC
,
Kupper
TS
. 
Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity
.
Nature
2012
;
483
:
227
31
.
13.
Gebhardt
T
,
Wakim
LM
,
Eidsmo
L
,
Reading
PC
,
Heath
WR
,
Carbone
FR
. 
Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus
.
Nat Immunol
2009
;
10
:
524
30
.
14.
Wakim
LM
,
Woodward-Davis
A
,
Bevan
MJ
. 
Memory T cells persisting within the brain after local infection show functional adaptations to their tissue of residence
.
Proc Natl Acad Sci U S A
2010
;
107
:
17872
9
.
15.
Piet
B
,
de Bree
GJ
,
Smids-Dierdorp
BS
,
van der Loos
CM
,
Remmerswaal
EB
,
von der Thusen
JH
, et al
CD8(+) T cells with an intraepithelial phenotype upregulate cytotoxic function upon influenza infection in human lung
.
J Clin Invest
2011
;
121
:
2254
63
.
16.
Woodberry
T
,
Suscovich
TJ
,
Henry
LM
,
August
M
,
Waring
MT
,
Kaur
A
, et al
Alpha E beta 7 (CD103) expression identifies a highly active, tonsil-resident effector-memory CTL population
.
J Immunol
2005
;
175
:
4355
62
.
17.
Mackay
LK
,
Stock
AT
,
Ma
JZ
,
Jones
CM
,
Kent
SJ
,
Mueller
SN
, et al
Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation
.
Proc Natl Acad Sci U S A
2012
;
109
:
7037
42
.
18.
Wakim
LM
,
Woodward-Davis
A
,
Liu
R
,
Hu
Y
,
Villadangos
J
,
Smyth
G
, et al
The molecular signature of tissue resident memory CD8 T cells isolated from the brain
.
J Immunol
2012
;
189
:
3462
71
.
19.
Ling
KL
,
Dulphy
N
,
Bahl
P
,
Salio
M
,
Maskell
K
,
Piris
J
, et al
Modulation of CD103 expression on human colon carcinoma-specific CTL
.
J Immunol
2007
;
178
:
2908
15
.
20.
Le Floc'h
A
,
Jalil
A
,
Vergnon
I
,
Le Maux Chansac
B
,
Lazar
V
,
Bismuth
G
, et al
Alpha E beta 7 integrin interaction with E-cadherin promotes antitumor CTL activity by triggering lytic granule polarization and exocytosis
.
J Exp Med
2007
;
204
:
559
70
.
21.
French
JJ
,
Cresswell
J
,
Wong
WK
,
Seymour
K
,
Charnley
RM
,
Kirby
JA
. 
T cell adhesion and cytolysis of pancreatic cancer cells: a role for E-cadherin in immunotherapy
?
Br J Cancer
2002
;
87
:
1034
41
.
22.
Quinn
E
,
Hawkins
N
,
Yip
YL
,
Suter
C
,
Ward
R
. 
CD103+ intraepithelial lymphocytes–a unique population in microsatellite unstable sporadic colorectal cancer
.
Eur J Cancer
2003
;
39
:
469
75
.
23.
Cresswell
J
,
Robertson
H
,
Neal
DE
,
Griffiths
TR
,
Kirby
JA
. 
Distribution of lymphocytes of the alpha(E)beta(7) phenotype and E-cadherin in normal human urothelium and bladder carcinomas
.
Clin Exp Immunol
2001
;
126
:
397
402
.
24.
Webb
JR
,
Wick
DA
,
Nielsen
JS
,
Tran
E
,
Milne
K
,
McMurtrie
E
, et al
Profound elevation of CD8+ T cells expressing the intraepithelial lymphocyte marker CD103 (alphaE/beta7 Integrin) in high-grade serous ovarian cancer
.
Gynecol Oncol
2010
;
118
:
228
36
.
25.
Franciszkiewicz
K
,
Le Floc'h
A
,
Jalil
A
,
Vigant
F
,
Robert
T
,
Vergnon
I
, et al
Intratumoral induction of CD103 triggers tumor-specific CTL function and CCR5-dependent T-cell retention
.
Cancer Res
2009
;
69
:
6249
55
.
26.
Morgan
EA
,
Yu
H
,
Pinkus
JL
,
Pinkus
GS
. 
Immunohistochemical detection of hairy cell leukemia in paraffin sections using a highly effective CD103 rabbit monoclonal antibody
.
Am J Clin Pathol
2013
;
139
:
220
30
.
27.
West
NR
,
Panet-Raymond
V
,
Truong
PT
,
Alexander
C
,
Babinszky
S
,
Milne
K
, et al
Intratumoral Immune Responses Can Distinguish New Primary and True Recurrence Types of Ipsilateral Breast Tumor Recurrences (IBTR)
.
Breast Cancer
2011
;
5
:
105
15
.
28.
Milne
K
,
Barnes
RO
,
Girardin
A
,
Mawer
MA
,
Nesslinger
NJ
,
Ng
A
, et al
Tumor-infiltrating T cells correlate with NY-ESO-1-specific autoantibodies in ovarian cancer
.
PLoS ONE
2008
;
3
:
e3409
.
29.
Marques
FR
,
Fonsechi-Carvasan
GA
,
De Angelo Andrade
LA
,
Bottcher-Luiz
F
. 
Immunohistochemical patterns for alpha- and beta-catenin, E- and N-cadherin expression in ovarian epithelial tumors
.
Gynecol Oncol
2004
;
94
:
16
24
.
30.
Jochems
C
,
Schlom
J
. 
Tumor-infiltrating immune cells and prognosis: the potential link between conventional cancer therapy and immunity
.
Exp Biol Med (Maywood)
2011
;
236
:
567
79
.
31.
Hadley
GA
,
Rostapshova
EA
,
Gomolka
DM
,
Taylor
BM
,
Bartlett
ST
,
Drachenberg
CI
, et al
Regulation of the epithelial cell-specific integrin, CD103, by human CD8 +cytolytic T lymphocytes
.
Transplantation
1999
;
67
:
1418
25
.
32.
Sheridan
BS
,
Lefrancois
L
. 
Intraepithelial lymphocytes: to serve and protect
.
Current Gastroenterol Rep
2010
;
12
:
513
21
.
33.
Schon
MP
,
Arya
A
,
Murphy
EA
,
Adams
CM
,
Strauch
UG
,
Agace
WW
, et al
Mucosal T lymphocyte numbers are selectively reduced in integrin alpha E (CD103)-deficient mice
.
J Immunol
1999
;
162
:
6641
9
.
34.
Feng
Y
,
Wang
D
,
Yuan
R
,
Parker
CM
,
Farber
DL
,
Hadley
GA
. 
CD103 expression is required for destruction of pancreatic islet allografts by CD8(+) T cells
.
J Exp Med
2002
;
196
:
877
86
.
35.
Bartlett
JM
,
Langdon
SP
,
Scott
WN
,
Love
SB
,
Miller
EP
,
Katsaros
D
, et al
Transforming growth factor-beta isoform expression in human ovarian tumours
.
Eur J Cancer
1997
;
33
:
2397
403
.
36.
Rao
ZY
,
Cai
MY
,
Yang
GF
,
He
LR
,
Mai
SJ
,
Hua
WF
, et al
EZH2 supports ovarian carcinoma cell invasion and/or metastasis via regulation of TGF-beta1 and is a predictor of outcome in ovarian carcinoma patients
.
Carcinogenesis
2010
;
31
:
1576
83
.
37.
Vergara
D
,
Merlot
B
,
Lucot
JP
,
Collinet
P
,
Vinatier
D
,
Fournier
I
, et al
Epithelial-mesenchymal transition in ovarian cancer
.
Cancer Lett
2010
;
291
:
59
66
.
38.
Bevan
MJ
. 
Memory T cells as an occupying force
.
Eur J Immunol
2011
;
41
:
1192
5
.
39.
Gebhardt
T
,
Mueller
SN
,
Heath
WR
,
Carbone
FR
. 
Peripheral tissue surveillance and residency by memory T cells
.
Trends Immunol
2013
;
34
:
27
32
.
40.
Inozume
T
,
Hanada
K
,
Wang
QJ
,
Ahmadzadeh
M
,
Wunderlich
JR
,
Rosenberg
SA
, et al
Selection of CD8+PD-1+ lymphocytes in fresh human melanomas enriches for tumor-reactive T cells
.
J Immunother
2010
;
33
:
956
64
.
41.
Kurman
RJ
,
Shih Ie
M
. 
Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer–shifting the paradigm
.
Hum Pathol
2011
;
42
:
918
31
.
42.
Cuburu
N
,
Graham
BS
,
Buck
CB
,
Kines
RC
,
Pang
YY
,
Day
PM
, et al
Intravaginal immunization with HPV vectors induces tissue-resident CD8+ T cell responses
.
J Clin Invest
2012
;
122
:
4606
20
.
43.
Uss
E
,
Rowshani
AT
,
Hooibrink
B
,
Lardy
NM
,
van Lier
RA
,
ten Berge
IJ
. 
CD103 is a marker for alloantigen-induced regulatory CD8+ T cells
.
J Immunol
2006
;
177
:
2775
83
.
44.
Zhao
D
,
Zhang
C
,
Yi
T
,
Lin
CL
,
Todorov
I
,
Kandeel
F
, et al
In vivo-activated CD103+CD4+ regulatory T cells ameliorate ongoing chronic graft-versus-host disease
.
Blood
2008
;
112
:
2129
38
.
45.
Milne
K
,
Kobel
M
,
Kalloger
SE
,
Barnes
RO
,
Gao
D
,
Gilks
CB
, et al
Systematic analysis of immune infiltrates in high-grade serous ovarian cancer reveals CD20, FoxP3 and TIA-1 as positive prognostic factors
.
PLoS ONE
2009
;
4
:
e6412
.
46.
West
NR
,
Milne
K
,
Truong
PT
,
Macpherson
N
,
Nelson
BH
,
Watson
PH
. 
Tumor-infiltrating lymphocytes predict response to anthracycline-based chemotherapy in estrogen receptor-negative breast cancer
.
Breast Cancer Res
2011
;
13
:
R126
.
47.
Wherry
EJ
,
Ha
SJ
,
Kaech
SM
,
Haining
WN
,
Sarkar
S
,
Kalia
V
, et al
Molecular signature of CD8+ T cell exhaustion during chronic viral infection
.
Immunity
2007
;
27
:
670
84
.
48.
Galon
J
,
Pages
F
,
Marincola
FM
,
Angell
HK
,
Thurin
M
,
Lugli
A
, et al
Cancer classification using the Immunoscore: a worldwide task force
.
J Transl Med
2012
;
10
:
205
.
49.
Keskin
DB
,
Allan
DS
,
Rybalov
B
,
Andzelm
MM
,
Stern
JN
,
Kopcow
HD
, et al
TGFbeta promotes conversion of CD16+ peripheral blood NK cells into CD16- NK cells with similarities to decidual NK cells
.
Proc Natl Acad Sci U S A
2007
;
104
:
3378
83
.
50.
Burrows
TD
,
King
A
,
Loke
YW
. 
Expression of adhesion molecules by human decidual large granular lymphocytes
.
Cell Immunol
1993
;
147
:
81
94
.
51.
Bamias
A
,
Tsiatas
ML
,
Kafantari
E
,
Liakou
C
,
Rodolakis
A
,
Voulgaris
Z
, et al
Significant differences of lymphocytes isolated from ascites of patients with ovarian cancer compared to blood and tumor lymphocytes. Association of CD3+CD56 +cells with platinum resistance
.
Gynecol Oncol
2007
;
106
:
75
81
.