JCDR - Register at Journal of Clinical and Diagnostic Research
Journal of Clinical and Diagnostic Research, ISSN - 0973 - 709X
Pathology Section DOI : 10.7860/JCDR/2019/42239.13150
Year : 2019 | Month : Sep | Volume : 13 | Issue : 09 Full Version Page : EC11 - EC17

Investigation of Immunohistochemical Expression of Programmed Death-Ligand 1 (PD-L1) in Female Mammary Carcinoma and its Correlation with the Extent of Stromal Tumour Infiltrating Lymphocytes

Passant Essam Eldin Shibel1, Hanan Soliman Abdelhamid2, Somia Abdulatif Mahmoud Soliman3, Samia Mohamed Gabal4

1 Assistant Lecturer, Department of Pathology, Faculty of Medicine, Cairo University (kasr alainy), Cairo, Egypt.
2 Assistant Professor, Department of Pathology, Faculty of Medicine, Cairo University (kasr alainy), Cairo, Egypt.
3 Lecturer, Department of Pathology, Faculty of Medicine, Cairo University (kasr alainy), Cairo, Egypt.
4 Professor, Department of Pathology, Faculty of Medicine, Cairo University (kasr alainy), Cairo, Egypt.


NAME, ADDRESS, E-MAIL ID OF THE CORRESPONDING AUTHOR: Passant Essam Eldin Shibel, 6 El Sahaba Square, Mossadaq Street, Dokki, Giza, Egypt.
E-mail: bassant.shibel@kasralainy.edu.eg
Abstract

Introduction

Breast Cancer (BC) is the most common type of cancer among women worldwide and a leading cause of cancer related deaths. Based on insight into immune system/cancer interactions, cancer immunotherapy strategies have been developed. Of these strategies, immune checkpoints regulators are among the most important. Programmed Death Ligand 1 (PD-L1) is one of the 2 ligands of Programmed Death-1 (PD-1); an important immune checkpoint regulator. Clinical trials using PD-L1 antibodies have shown efficacy in various cancers. Some of these antibodies have received Food and Drug Administration (FDA) approval for clinical use.

Aim

Detection of Immunohistochemical (IHC) expression of PD-L1 by Tumour Cells (TC) and stromal Tumour Infiltrating Lymphocytes (TILs) in female BC, as well as, correlation of this expression with the pathologic parameters of the tumours.

Materials and Methods

One hundred BC tissue divtions were collected from mastectomy specimens. IHC expression of PD-L1 was investigated in TC and TILs. Correlation of PD-L1 expression with the tumours pathologic parameters was performed using Statistical Package of Social Science (SPSS) Software program, version 25.0.

Results

TC PD-L1 expression was detected in 61% of cases and showed statistically significant correlation with higher tumour grade, higher prognostic stage, Estrogen receptor negativity, Human Epidermal Growth Factor Receptor Type 2 (HER2) enriched molecular subtype and LVI (p-value <0.05). TILs PD-L1 expression was detected in 55% of cases and showed statistically significant correlation with higher tumour grade, higher prognostic stage, Estrogen and Progesterone receptor negativity, HER2 overexpression, higher Ki-67 index, triple negative molecular subtype and higher stromal TILs (p-value <0.05).

Conclusion

The present results supported PD-L1 expression in BC by both TC and TILs, with higher expression in subset of tumours that are high grade, TILs rich and lacking hormone receptors, highlighting them as candidates for anti-PD-1/PD-L1 therapy.

Keywords

Introduction

Breast Cancer (BC) is the most frequently diagnosed female cancer worldwide and is a leading cause of cancer-related deaths in women [1]. In Egypt, BC accounts for 38% of all female cancers according to the National Cancer Institute, Cairo University [2]. Currently, the prognosis and treatment of BC are dependent on tumour grade and stage, as well as 3 main protein markers: Oestrogen Receptor (ER), Progesterone Receptor (PR) and Human Epidermal Growth Factor Receptor 2 (HER2) [3]. To the best of authors knowledge, no immunotherapy is currently widely used, for treatment of BC, as BC hasn’t been generally considered an immunogenic cancer. However, some investigators have begun to consider novel immunotherapeutic strategies for treatment of BC [4].

The immune system recognises cancer cells as soon as they emerge. The interaction between T-cells and Antigen-presenting cells (APC) is complex and involves the T-cell receptor as well as, multiple co-regulatory receptors, which exert either activating or inhibitory stimuli to the T-cells [5]. Tumours can resist immune attack by expressing ligands that engage inhibitory receptors and dampen T-cell functions [6].

The programmed death pathway is one of the major immune response checkpoint and a target for cancer immunotherapy [7]. PD-1 is a transmembrane protein and member of the B7-CD28 family of co-regulatory molecules expressed by activated lymphocytes [8,9]. It has two known ligands, PD-L1 (B7-H1) and PD-L2 (B7-DC) [6]. The interaction between PD-1 and its ligands inhibits T-cells, blocking immune responses [10].

PD-L1 is the primary PD-1 ligand [6]. It is a cell surface glycoprotein [11] that is expressed on a variety of cell types, including B and T lymphocytes, dendritic cells, macrophages and mast cells [12]. Upregulated PD-L1 binds PD-1 on T-cells, contributing to the development of T-cell exhaustion. Tumour cells have co-opted this PD-1/PD-L1 regulatory mechanism, normally designed to prevent autoimmune attacks. They overexpress PD-L1 to escape immunologic attacks and permit tumour growth [13].

PD-L1 appears to be up-regulated in multiple solid malignancies [14], at rates of 20-70% including Non-Small Cell Lung Cancer (NSCLC) [15-17], urinary bladder cancer [18] and malignant melanoma [19]. PD-L1 is expressed in both tumour cells and immune cells [20-22]. Its expression in immune cells has also demonstrated clinical implication [23]. A report suggested that PD-L1 expression by immune cells rather than tumour cells is more predictive of response to PD-1 pathway blockade [15]. Therefore, the evaluation of PD-L1 should be performed in both cancer and immune cells [24].

These properties make PD-L1 a potentially promising cancer immunotherapy [6], where antibodies targeting either PD-1 or PD-L1 have shown durable responses in patients with highly immunogenic tumours such as melanoma, non-small-cell lung cancer and renal-cell carcinoma [25], with some of them receiving Food and Drug Administration (FDA) approval for clinical use [26].

Several studies investigated PD-L1 expression in BC. They showed variable rates of expression within the TC and Tumour Infiltrating Lymphocytes (TILs). The literature also showed contradictory results about the prognostic value of PD-L1 expression in BC. While some confirmed PD-L1 expression in BC as a marker of poor prognosis [27], others showed association of PD-L1 expression in BC with more favourable prognosis [9,28].

This study aimed at Investigation of immunohistochemical expression of PD-L1 in TC and TILs of female breast carcinoma, as well as, correlation of this expression with the BC subtypes, extent of tumour infiltrating lymphocytes and other available pathological aspects of the tumour, including tumour histological type, grade and stage.

Materials and Methods

This study was a retrospective observational cross-sectional study. A total of 100 formalin fixed, paraffin embedded full-face BC tumour tissue sections were collected from the archive of the Pathology Department at the Kasr El Aini Hospital (Cairo university hospital), in the time period between March 2015 and October 2017. The authors obtained the approval of Ethical Committee in the faculty of Medicine, Cairo University (N-115-2016).

Inclusion criteria included cases diagnosed as BC subjected to radical or conservative mastectomy with axillary dissection and has performed immunohistochemical evaluation for ER, PR, HER2 and Ki-67.

Exclusion criteria included: 1) Cases with missing data or no available IHC report; 2) Cases who received neo-adjuvant therapy; either hormonal or chemotherapy; 3) Patients subjected to lumpectomy or simple mastectomy without axillary sampllng; 4) Cases with HER2 Score 2 by IHC, with no available SISH/DISH report.

The data obtained from the pathology reports of the cases included age of patient, tumour size and Lymph Node (LN) status. An IHC report including the ER, PR, HER2 and Ki67 results was obtained for each case. A Dual In Situ Hybridization (DISH) report was obtained for cases with HER2 class 2+.

Haematoxylin and eosin stained sections were prepared for histopathological examination. The specimens were anonymous for confidentiality and replaced by numbers. The tumours were histologically typed according to the latest available World Health Organisation classification (WHO 2012) [29]. Histological grading was performed according to the Nottingham Grading System [30]. For further statistical evaluation, Grade 1 and 2 cases were considered as low grade, while Grade 3 cases were considered as high grade [31, 32]. Regarding lobular carcinomas, classic lobular carcinoma cases were considered as low grade and pleomorphic lobular carcinoma cases were considered as high grade [33]. Lympho-vascular invasion (LVI) was defined as presence of tumour cells within an endothelial lined space (lymphatic and/or blood vessel) outside the border of the tumour [34]. The slides were also examined for the presence of tumoural necrosis.

Regarding the TILs, they were scored following the recommendations of the International TILs Working Group 2014. Briefly, all stromal mononuclear cells within the borders of the invasive tumour were evaluated and reported as a percentage value of the stromal area (i.e., percentage of the stromal area occupied by mononuclear inflammatory cells) and not as a percentage of the stromal cells. TILs outside of the tumour border, around DCIS, ADH and normal breast tissue, as well as in areas of necrosis and hyalinosis, if any, were not included in the scoring. The working group recommended full assessment of average TILs in the examined tumour area; they don’t recommend focusing on ‘hot spots’ [35]. As the working group didn’t recommend a clinically relevant threshold[s] for TILs assessment [36], the present authors scored stromal TILs subjectively in 10% increments. Tumours were classified as high TILs (≥30%) or low TILs (<30%) [11,36].

Tumours staging was performed using the TNM staging system. The cases were further divided into anatomic stages and prognostic stages according to the 8th edition of the AJCC staging manual [37]. Regarding the prognostic stages, 15 cases were excluded as their prognostic stages were missing in the AJCC classification. The stages (whether anatomic or prognostic) were further subdivided into early; Stages I and II and advanced; stages III and IV, for statistical purposes [38].

Regarding the BC subtyping, the tumours were classified according to the St. Gallen International Expert Consensus 2013 recommendations, which stated that although identification of intrinsic subtypes is more accurate using molecular techniques, surrogate definitions of BC subtype can be obtained by IHC evaluation of ER, PgR, Ki-67 and HER2, in countries where routine molecular studies is not available for all cases [39]. Luminal cases having high histological grade were considered as luminal B rather than luminal A, regardless of their Ki-67 index, according to St. Gallen International Expert Consensus 2017 recommendations [40]. This subtyping was also supported by the 8th edition of the AJCC staging manual released in 2017 [37].

Paraffin sections were cut at 4 μm thickness on positively charged slides and stained manually for immunohistochemistry in the tumour marker unit of the Pathology Department at Kasr El Aini Hospital. The sections were deparaffinized in xylene, dehydrated in ethanol, then washed in Tris-buffer Physiological Saline (TBS). Antigen retrieval was performed by heating the sections in sodium citrate buffer. The sections were then washed in TBS buffer. Endogenous peroxide blocking was then achieved by 0.3% hydrogen peroxide in methanol, followed by washing in TBS for 15 minutes.

The primary antibody was anti-PD-L1 antibody (rabbit polyclonal, 0.2 mL concentrated, obtained from Novus Biologicals). The antibody was diluted with TBS, pH 7.4. The sections were incubated overnight with the diluted antibody. The sections were then incubated for 30 minutes with the secondary antibody (4.5 μL biotinylated anti-mouse antibody in 1 mL of 1% BSA), then washed in TBS buffer. The slides were finally stained with Diaminobenzidine tetrahydrochloride (DAB) solution, then in distilled water. Hematoxylin was used as a counterstain.

PD-L1 immunohistochemical staining was scored in the tumour cells and the stromal TILs separately:

Regarding the TC PD-L1 expression, although both cytoplasmic and membranous positivity was observed as reported by many other authors [11,23,41-44], only membranous staining was considered for further analysis [45] as PD-L1 localisation to the cell membrane is likely required for interaction with PD-1 [46].

Cases were considered as positive if 1% of tumour cells stained for PD-L1 with any staining intensity. This threshold was selected based on data demonstrating clinical response to PD-L1 inhibition at this expression level in some cancers. Further scoring was performed as follows: Score 1; 1% to 5%, Score 2; 6% to 10%, Score 3; 11% to 25%, Score 4; 26% to 50% and Score 5; >50% positive tumour cells [47].

Tumour infiltrating lymphocytes staining was considered as positive when >5% of stromal TILs showed PD-L1 reactivity with any intensity and was further scored as follows: Score 1; 5% to 10%, Score 2; 11% to 25%, Score 3; 26% to 50% and Score 4; >50% positive TILs [47].

Statistical Analysis

The previously mentioned histopathological and immuno-histochemical data were then transferred to the SPSS Software program, version 25.0 to be statistically analysed. Data were summarised using frequency and percentages. Comparison between groups was then performed using chi-square test. Statistically significant results were considered if p-value was ≤0.05.

Results

This study included 100 cases of BC obtained from modified radical mastectomy and conservative breast surgery specimens. The pathological data of the cases are summarised in [Table/Fig-1].

The pathological data of the collected cases.

ParameterNumber (%)
Histological typeIC-NST76 (76%)
ILC10 (10%)
Carcinoma with medullary features7 (7%)
Mixed duct and lobular2 (2%)
Mucinous carcinoma2 (2%)
Encysted papillary carcinoma with micro-invasion1 (1%)
Tubular carcinoma1 (1%)
Cribriform carcinoma1 (1%)
Histological gradeLow57 (57%)
High43 (43%)
Tumoural necrosisPositive29 (29%)
Negative71 (71%)
T stageT110 (10%)
T275 (75%)
T313 (13%)
T42 (2%)
N stage045 (45%)
126 (26%)
217 (17%)
312 (12%)
Anatomic stageEarly (I and II)66 (66%)
Late (III and IV)34 (34%)
Prognostic stageEarly (I and II)47 (55.3%)
Late (III and IV)38 (44.7%)
ERPositive74 (74%)
Negative26 (26%)
PRPositive70 (70%)
Negative30 (30%)
Her2-neuPositive15 (15%)
Negative85 (85%)
Ki-67 indexLow50 (50%)
High50 (50%)
BC subtypesLuminal A32 (32%)
Luminal B42 (42%)
HER2 enriched10 (10%)
Triple negative16 (16%)
LVIPositive49 (49%)
Negative51 (51%)
Stromal TILsLow78 (78%)
High22 (22%)

Regarding the extent of stromal TILs, 78 cases showed low TILs (78%) [Table/Fig-2a] and 22 cases showed high TILs (22%) [Table/Fig-2b,c].

H&E stained sections of the studied cases: a) Low stromal TILs IC-NST (<30% of the stroma) (x100); b) High stromal TILs IC-NST (≥30% of the stroma) (x100); c) High stromal TILs carcinoma with medullary features (≥30% of the stroma) (x100).

As regards TC PD-L1 expression, it was positive in 61 cases (61%) and negative in 39 cases (39%). Among the positive cases, 1 case (1.6%) was Score 1, 5 cases (8.2%) were Score 2, 7 cases (11.5%) were Score 3, 16 cases (26.2%) were Score 4 and 32 cases (52.5%) were Score 5 [Table/Fig-3a,b,4a,b].

Histogram showing Expression and scores of TC PD-L1: a) TC PD-L1. Expression; b) Positive TC PD-L1 cases scores.

Immunohistochemical expression of PD-L1 in TC showing mainly membranous staining: a) (×100); b) (×200).

Stromal TILs PD-L1 expression was positive in 55 cases (55%) and negative in 45 cases (45%). Among the positive cases, 6 case (10.9%) were Score 1, 11 cases (20%) were Score 2, 20 cases (36.4%) were Score 3 and 18 cases (32.7%) were Score 4 [Table/Fig-5a,b,6a,b].

Histogram showing Expression and scores of TILs PD-L1: a) TILs PD-L1 expression; b) Positive TILs PD-L1 cases scores.

Immunohistochemistry expressions of PD-L1 in TILs: a) (×200); b) (×400).

A highly statistically significant direct correlation was detected between TC and TILs PD-L1 expression (p-value≤0.001). The pathologic characteristics of the studied cases stratified by TC and TILs-PD-L1 expression were then summarised in [Table/Fig-7,8].

Immunohistochemical expression of both TC PD-L1 and TILs PD-L1 (×200).

The pathologic characteristics and PD-L1 protein expression status (Chi-square test).

ParameterTC PD-L1 Positive (% within parameter)TC PD-L1 Negative (% within parameter)p-valueTIL PD-L1 Positive (% within parameter)TIL PD-L1 Negative (% within parameter)p-value
Histological typeIC-NST47 (61.8%)29 (38.2%)0.72544 (57.9%)32 (42.1%)0.059
ILC6 (60%)4 (40%)3 (30%)7 (70%)
Carcinoma with medullary features5 (71.4%)2 (28.6%)6 (85.7%)1 (14.3%)
Others3 (42.9%)4 (57.1%)2 (28.6%)5 (71.4%)
Histological gradeLow27 (47.4%)30 (52.6%)0.01*23 (40.4%)34 (59.6%)0.001*
High34 (79.1%)9 (20.9%)32 (74.4%)11 (25.6%)
Tumoural necrosisPositive23 (79.3%)6 (20.7%)0.016*19 (65.5%)10 (34.5%)0.177
Negative38 (53.5%)33 (46.5%)36 (50.7%)35 (49.3%)
T stageT1+T249 (57.6%)36 (42.4%)0.10244 (51.8%)41 (48.2%)0.122
T3+T412 (80%)3 (20%)11 (73.3%)4 (26.7%)
N stage024 (53.3%)21 (46.7%)0.22124 (53.3%)21 (46.7%)0.153
115 (57.7%)11 (42.3%)14 (53.8%)12 (46.2%)
212 (70.6%)5 (29.4%)7 (41.2%)10 (58.8%)
310 (83.3%)2 (16.7%)10 (83.3%)2 (16.7%)
Anatomic stageEarly (I and II)36 (54.5%)30 (45.5%)0.06534 (51.5%)32 (48.5%)0.329
Late (III and IV)25 (73.5%)9 (26.5%)21 (61.8%)13 (38.2%)
Prognostic stageEarly (I and II)24 (51.1%)23 (48.9%)0.008*20 (42.6%)27 (57.4%)0.004*
Late (III and IV)30 (78.9%)8 (21.1%)28 (73.7%)10 (26.3%)
ERPositive40 (54.1%)34 (45.9%)0.016*34 (45.9%)40 (54.1%)0.002*
Negative21 (80.8%)5 (19.2%)21 (80.8%)5 (19.2%)
PRPositive39 (55.7%)31 (44.3%)0.09838 (54.3%)32 (45.7%)0.004*
Negative22 (73.3%)8 (26.7%)23 (76.7%)7 (23.3%)
Her2-neuPositive12 (80%)3 (20%)0.10213 (86.7%)2 (13.3%)0.007*
Negative49 (57.6%)36 (42.4%)42 (49.4%)43 (50.6%)
Ki-67 indexLow27 (54%)23 (46%)0.15122 (44%)28 (56%)0.027*
High34 (68%)16 (32%)33 (66%)17 (34%)
BC subtypesLuminal A13 (40.6%)19 (59.4%)0.006*10 (31.3%)22 (68.8%)0.002*
Luminal B27 (64.3%)15 (35.7%)24 (57.1%)18 (42.9%)
HER2 enriched10 (100%)0 (0%)8 (80%)2 (20%)
Triple negative11 (68.8%)5 (31.3%)13 (81.2)3 (18.8%)
LVIPositive37 (75.5%)12 (24.5%)0.004*31 (63.3%)18 (36.7%)0.103
Negative24 (47.1%)27 (52.9%)24 (47.1%)27 (52.9%)
Stromal TILsLow44 (56.4%)34 (43.6%)0.07636 (46.2%)42 (53.8%)0.001*
High17 (77.3%)5 (22.7%)19 (86.4%)3 (13.6%)

*Statistically significant


Discussion

Programmed death-ligand 1 is an immune check point regulator up-regulated in many solid malignancies [14]. Antibodies targeting PD-L1 have shown clinical responses in multiple tumours [6]. Some of these antibodies have received FDA approval for clinical use [26]. Several studies investigated PD-L1 expression in BC, showing variable rates of expression within the tumour cells and the tumour infiltrating lymphocytes [27]. In BC, the PD-1 inhibitor Pembrolizumab has shown promising effects and a good safety profile [48]. PD-L1 inhibitors, atezolizumab and avelumab appear to be particularly active in TNBC [49].

In the present study, TC PD-L1 expression was reported in 61% of the cases. The rates of TC PD-L1 expression varied greatly in the literature ranging from 1.6% [20] to 94.9% [23]. However, many studies reported rates lower than the present; 34% [28], 8.3% [50] and 19.6% [51]. This variation can be explained by the large number of antibodies used, the use of full face sections versus Tissue Micro-Array (TMA), the different cut-off values used for positivity, whether cytoplasmic or membranous staining were interpreted and the composition of the studied population itself.

In our study, the relatively high rate of expression can be explained by the use of full face sections and a polyclonal antibody, as well as, the low cut off of positivity (1%). A study using full-face sections and a polyclonal antibody, similar to us, reported TC PD-L1 expression in 94.9% [23]. Another study using polyclonal antibody, but on TMA reported 56.5% positive TC PD-L1 expression [10]. The present choice of this polyclonal antibody stemmed from the absence of a uniformly accepted clone among the different organs.

Concerning the TILs PD-L1 expression, it was positive in 55% of the present cases. Similar to TC PD-L1 expression, a wide range of TILs PD-L1 expression rates have been reported in the literature, ranging from 6% [20] to 78% [8]. The rate of TILs PD-L1 expression in the present study (55%) was slightly lower than TC PD-L1 expression (61%). Although this agreed with some studies [31], many other studies reported the reverse [20,47,50].

Upon scoring our PD-L1 positive cases, 52.5% of the TC PD-L1 positive cases showed diffuse positivity (>50% of cells) and 32.7% of the TILs PD-L1 positive cases showed diffuse positivity (>50% of cells). Dill E et al., using the same scoring system reported diffuse TC PD-L1 expression in 20% of TC PD-L1 positive cases and diffuse TILs PD-L1 expression in 8.5% TILs PD-L1 positive cases [47]. Park I et al., reported positive PD-L1 expression of H-score 3+ in 51.6% of their cases [23] and Tomioka N et al., reported that 22.7% of the positive cases showed PD-L1 in >50% of the tumour cells [36].

Despite these variable figures, most of the studies agreed that a significant proportion of the positive cases showed only focal positivity, with considerable intra-tumoural heterogeneity of PD-L1 expression. Although the clinically relevant threshold of PD-L1 positivity for BC has not been yet established, this finding may be of significance if low levels of PD-L1 expression proved to be clinically actionable [47]. Caution should be exercised when interpreting PD-L1 stain on core biopsies where at least the negative cases may need repeated staining on later excision biopsies.

Regarding the histological subtypes, carcinoma with medullary features showed the highest rates of both TC and TILs PD-L1 expression, followed by IC-NST cases then ILC cases and finally other types (including mostly low grade types; papillary, mucoid, tubular and cribriform). This agreed with the results of Dill E et al., who reported the highest rates of PD-L1 expression in both compartments in cases with medullary features [47]. Guo L et al., and Hou Y et al., also reported higher rates of overall PD-L1 expression in IC-NST cases than ILC cases [41,52]. In addition, a study which analysed PD-L1 micro RNA (mRNA) expression in BC cell lines reported highest rates of PD-L1 expression in cases with medullary features [53].

In the present study, high grade cases showed statistically significant higher rates of TC and TILs PD-L1 expression than low grade cases. This is consistent with most of the studies in the literature [41,52,53]. Also, cases associated with necrosis showed higher TC PD-L1 and TILs PD-L1 expression than those devoid of necrosis. This was consistent with the detected high PD-L1 expression in high grade cases and in cases with medullary features.

Regarding the T stage, larger tumour size cases (T3 and T4) showed higher rates of TC and TILs PD-L1 expression than smaller tumour size cases. This was consistent with the results of Baptista MZ et al., and Li F et al., who reported higher TC PD-L1 expression in cases ≥ T2 [10,51]. In contrast, Park IH et al., and Kitano A et al., reported higher TC PD-L1 expression in smaller size cases. This may be explained by the fact that both those studies were concerned more by early stage BC cases with larger number of T1 and T2 cases than T3 and T4 cases [23,28].

In this study, cases positive for LN metastasis showed higher rates of TC and TILs PD-L1 expression than LN negative cases. This agreed with the results of a meta-analysis involving five studies [4] and another study [54]. Among the LN positive cases in the present study, the rate of TC PD-L1 expression increased with the increase in the N stage; N1 (57.7%), N2 (70.6%) and N3 (83.3%). This was consistent with the results of Qin T et al., [27].

On grouping our cases into anatomic and prognostic stages, the rate of TC and TILs PD-L1 expression was higher in advanced than early stage cases. This was consistent with the results of Qin T et al., and Kim A et al., who reported higher rate of TC PD-L1 expression in advanced anatomic stage cases [27,31]. However, Kim A et al., reported a contradictory higher rate of TILs PD-L1 expression in early stage cases [31].

It is to be noted that none of these studies staged their cases according to the recently published AJCC prognostic staging system. In the present study, only the correlations of PD-L1 expression with the prognostic stages, incorporating the more important grade and hormone status, were statistically significant.

Concerning the hormone receptors, higher TC and TILs PD-L1 expression was detected in hormone (ER and PR) negative then hormone positive cases. This was consistent with the results of Kim A et al., and Wimberly H et al., who assessed PD-L1 expression using quantitative immunofluorescence [31,55]. In contrast to the present results, Park H et al., reported statistically significant higher TC PD-L1 expression in hormone positive cases [23].

In the present study, the rate of TC and TILs PD-L1 expression was higher in the HER2 positive cases than in the HER2 negative cases. This agreed with the results of many studies [10,11]. This finding may be clinically important, where; some pre-clinical studies suggest that combining anti-HER2 therapy with some immune checkpoint regulators, such as anti-PD-1, anti-PD-L1 or anti-CTLA4 antibodies, may have a synergistic effect, increasing their antitumour efficacy [43].

Regarding the Ki-67 proliferation index, cases with high index showed higher rate of TC and TILs PD-L1 expression than low Ki67 cases. This result was consistent with the wide agreement on the association of PD-L1 expression with the high grade as well as with the results of some studies [11,56]. This correlation might be explained by the fact that high mutation rate of hyper-proliferative tumour cells in high Ki67 cases, may be responsible for higher immunogenicity due to the rapid emergence of neo-antigens [53].

Some other studies reported contradictory higher PD-L1 expression in low Ki-67 cases [23,27]. Those contradictory results again can be explained by the nature of Ki-67 itself as a marker of proliferation and its weak reproducible results.

On grouping the present cases into BC subtypes, the highest rate of TC PD-L1 expression was noticed in HER2 enriched subtype, while highest rate of TILs PD-L1 expression was noticed in triple negative subtype. Although, many studies reported highest PD-L1 expression in TN cases, the present results agreed with others [47]. Also, Cimino-Mathews A et al., reported highest both TC and TILs PD-L1 expression in HER2 enriched subtype [8]. Jang BS et al., also reported highest PD-L1 mRNA expression in HER2 enriched subtype. In general, almost all of the studies agreed that PD-L1 expression was higher in non-luminal than luminal BC [57].

In the present study, cases with high stromal TILs (≥30% of the stroma) showed higher rate of TC and TILs PD-L1 expression than cases with low stromal TILs. This agreed with the results of many others [4,8,42,55].

Finally, a highly statistically significant direct correlation was detected between TC and TILs PD-L1 expression in the present study. This was consistent with the results of many studies [8,31,47].

Limitation

One important limitation of this study is the lack of correlation with the patient’s prognosis and survival. However, the present study agreed with many others that PD-L1 expression is higher in tumours showing poor prognostic factors such as the high grade and lack of hormone receptors.

Conclusion

The present results are in favour of targeting the PD-1/PD-L1 pathway as an immunotherapy in BC, where, PD-L1 expression was detected in both BC tumour cells and TILs. The present authors reported that the expression of PD-L1 in BC is higher in a subset of tumours that are high grade, rich in TILs and lacking hormone receptors, suggesting that they may be important candidates for anti-PD-1/PD-L1 therapy.

*Statistically significant

References

[1]Torre LA, Siegel RL, Ward EM, Jemal A, Global cancer incidence and mortality rates and trends-an update Cancer Epidemiol Biomarkers Prev 2016 25(1):16-27.10.1158/1055-9965.EPI-15-057826667886  [Google Scholar]  [CrossRef]  [PubMed]

[2]Mokhtar N, Salama A, Badawy O, Khorshed E, Mohamed G, Ibrahim M, Breast tumours. In: Gouda I, Badran A, editors Cancer pathology registry. A 12-year registry 2000-2011. department of pathology national cancer institute 2016 CairoCairo University:8-31.  [Google Scholar]

[3]Coates AS, Winer EP, Goldhirsch A, Gelber RD, Gnant M, Piccart-Gebhart M, Tailoring therapies-improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015 Ann Oncol 2015 26(8):1533-46.10.1093/annonc/mdv22125939896  [Google Scholar]  [CrossRef]  [PubMed]

[4]Guo Y, Yu P, Liu Z, Maimaiti Y, Wang S, Yin X, Prognostic and clinicopathological value of programmed death ligand-1 in breast cancer: a meta-analysis PloS one 2016 11(5):e015632310.1371/journal.pone.015632327227453  [Google Scholar]  [CrossRef]  [PubMed]

[5]Muenst S, Schaerli AR, Gao F, Däster S, Trella E, Droeser RA, Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer Breast Cancer Res Treat 2014 146(1):15-24.10.1007/s10549-014-2988-524842267  [Google Scholar]  [CrossRef]  [PubMed]

[6]Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Safety and activity of anti-PD-L1 antibody in patients with advanced cancer N Engl J Med 2012 366(26):2455-65.10.1056/NEJMoa120069422658128  [Google Scholar]  [CrossRef]  [PubMed]

[7]Cimino-Mathews A, Foote JB, Emens LA, Immune targeting in breast cancer Oncol 2015 29(5):375-85.  [Google Scholar]

[8]Cimino-Mathews A, Thompson E, Taube JM, Ye X, Lu Y, Meeker A, PD-L1 (B7-H1) expression and the immune tumour microenvironment in primary and metastatic breast carcinomas Hum Pathol 2016 47(1):52-63.10.1016/j.humpath.2015.09.00326527522  [Google Scholar]  [CrossRef]  [PubMed]

[9]Uhercik M, Sanders AJ, Owen S, Davies EL, Sharma AK, Jiang WG, Clinical significance of PD1 and PDL1 in human breast cancer Anticancer Res 2017 37(8):4249-54.10.21873/anticanres.11817  [Google Scholar]  [CrossRef]

[10]Baptista MZ, Sarian LO, Derchain SF, Pinto GA, Vassallo J, Prognostic significance of PD-L1 and PD-L2 in breast cancer Hum Pathol 2016 47(1):78-84.10.1016/j.humpath.2015.09.00626541326  [Google Scholar]  [CrossRef]  [PubMed]

[11]Polónia A, Pinto R, Cameselle-Teijeiro JF, Schmitt FC, Paredes J, Prognostic value of stromal tumour infiltrating lymphocytes and programmed cell death-ligand 1 expression in breast cancer J Clin Pathol 2017 70(10):860-67.10.1136/jclinpath-2016-20399028373294  [Google Scholar]  [CrossRef]  [PubMed]

[12]Ma W, Gilligan BM, Yuan J, Li T, Current status and perspectives in translational biomarker research for PD-1/PD-L1 immune checkpoint blockade therapy J Hematol Oncol 2016 9(1):4710.1186/s13045-016-0277-y27234522  [Google Scholar]  [CrossRef]  [PubMed]

[13]Patel SP, Kurzrock R, PD-L1 expression as a predictive biomarker in cancer immunotherapy Mol Cancer Ther 2015 14(4):847-56.10.1158/1535-7163.MCT-14-098325695955  [Google Scholar]  [CrossRef]  [PubMed]

[14]Schalper KA, Velcheti V, Carvajal D, Wimberly H, Brown J, Pusztai L, In situ tumour PD-L1 mRNA expression is associated with increased TILs and better outcome in breast carcinomas Clin Cancer Res 2014 20(10):2773-82.10.1158/1078-0432.CCR-13-270224647569  [Google Scholar]  [CrossRef]  [PubMed]

[15]Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients Nature 2014 515(7528):56310.1038/nature1401125428504  [Google Scholar]  [CrossRef]  [PubMed]

[16]D’incecco A, Andreozzi M, Ludovini V, Rossi E, Capodanno A, Landi L, PD-1 and PD-L1 expression in molecularly selected non-small-cell lung cancer patients Br J Cancer 2015 112(1):9510.1038/bjc.2014.55525349974  [Google Scholar]  [CrossRef]  [PubMed]

[17]Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, Eder JP, Pembrolizumab for the treatment of non–small-cell lung cancer N Engl J Med 2015 372(21):2018-28.10.1056/NEJMoa150182425891174  [Google Scholar]  [CrossRef]  [PubMed]

[18]Powles T, Eder JP, Fine GD, Braiteh FS, Loriot Y, Cruz C, MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer Nature 2014 515(7528):55810.1038/nature1390425428503  [Google Scholar]  [CrossRef]  [PubMed]

[19]Thierauf J, Veit JA, Affolter A, Bergmann C, Grünow J, Laban S, Identification and clinical relevance of PD-L1 expression in primary mucosal malignant melanoma of the head and neck Melanoma Res 2015 25(6):503-09.10.1097/CMR.000000000000019726352784  [Google Scholar]  [CrossRef]  [PubMed]

[20]Ali HR, Glont SE, Blows FM, Provenzano E, Dawson SJ, Liu B, PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes Ann Oncol 2015 26(7):1488-93.10.1093/annonc/mdv19225897014  [Google Scholar]  [CrossRef]  [PubMed]

[21]Zhang Y, Morgan R, Chen C, Cai Y, Clark E, Khan WN, Mammary-tumour-educated B cells acquire LAP/TGF-β and PD-L1 expression and suppress anti-tumour immune responses Int Immunol 2016 28(9):423-33.10.1093/intimm/dxw00726895637  [Google Scholar]  [CrossRef]  [PubMed]

[22]Meniawy TM, Lake RA, McDonnell AM, Millward MJ, Nowak AK, PD-L1 on peripheral blood T lymphocytes is prognostic in patients with non-small cell lung cancer (NSCLC) treated with EGFR inhibitors Lung Cancer 2016 93:09-16.10.1016/j.lungcan.2015.12.00626898608  [Google Scholar]  [CrossRef]  [PubMed]

[23]Park IH, Kong SY, Ro JY, Kwon Y, Kang JH, Mo HJ, Prognostic implications of tumour-infiltrating lymphocytes in association with programmed death ligand 1 expression in early-stage breast cancer Clin Breast Cancer 2016 16(1):51-58.10.1016/j.clbc.2015.07.00626364145  [Google Scholar]  [CrossRef]  [PubMed]

[24]Sun WY, Lee YK, Koo JS, Expression of PD-L1 in triple-negative breast cancer based on different immunohistochemical antibodies J Transl Med 2016 14(1):17310.1186/s12967-016-0925-627286842  [Google Scholar]  [CrossRef]  [PubMed]

[25]Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Safety, activity, and immune correlates of anti–PD-1 antibody in cancer N Engl J Med 2012 366(26):2443-54.10.1056/NEJMoa120069022658127  [Google Scholar]  [CrossRef]  [PubMed]

[26]Borne S. U.S. FDA approved immune-checkpoint inhibitors for cancer [document on the Internet]. Medi-paper online; 2017 November 9 [cited 2019 April 7]. Available from: https://medi-paper.com/us-fda-approved-immune-checkpoint-inhibitors-approved-immunotherapies  [Google Scholar]

[27]Qin T, Zeng YD, Qin G, Xu F, Lu JB, Fang WF, High PD-L1 expression was associated with poor prognosis in 870 Chinese patients with breast cancer Oncotarget 2015 6(32):3397210.18632/oncotarget.558326378017  [Google Scholar]  [CrossRef]  [PubMed]

[28]Kitano A, Ono M, Yoshida M, Noguchi E, Shimomura A, Shimoi T, Tumour-infiltrating lymphocytes are correlated with higher expression levels of PD-1 and PD-L1 in early breast cancer ESMO open 2017 2(2):e00015010.1136/esmoopen-2016-00015028761741  [Google Scholar]  [CrossRef]  [PubMed]

[29]Lakhani SR, Ellis IO, Schnitt SJ, Tan PH, van de Vijver MJ, WHO classification of tumours of the breast 2012 4th edLyonIARC Press  [Google Scholar]

[30]Elston CW, Ellis IO, Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up Histopathology 1991 19(5):403-10.10.1111/j.1365-2559.1991.tb00229.x1757079  [Google Scholar]  [CrossRef]  [PubMed]

[31]Kim A, Lee SJ, Kim YK, Park WY, Kim JY, Lee CH, Programmed death-ligand 1 (PD-L1) expression in tumour cell and tumour infiltrating lymphocytes of HER2-positive breast cancer and its prognostic value Sci Rep 2017 7(1):1167110.1038/s41598-017-11905-728916815  [Google Scholar]  [CrossRef]  [PubMed]

[32]Wang C, Ye Q, Cao Y, Tan J, Wang F, Jiang J, Downregulation of regulator of G protein signaling 2 expression in breast invasive carcinoma of no special type: Clinicopathological associations and prognostic relevance Oncol Lett 2018 15(1):213-20.10.3892/ol.2017.7351  [Google Scholar]  [CrossRef]

[33]Reed AE, Kutasovic JR, Lakhani SR, Simpson PT, Invasive lobular carcinoma of the breast: morphology, biomarkers and’omics Breast Cancer Res 2015 17(1):1210.1186/s13058-015-0519-x25849106  [Google Scholar]  [CrossRef]  [PubMed]

[34]Gujam FJ, Going JJ, Edwards J, Mohammed ZM, McMillan DC, The role of lymphatic and blood vessel invasion in predicting survival and methods of detection in patients with primary operable breast cancer Crit Rev Oncol Hematol 2014 89(2):231-41.10.1016/j.critrevonc.2013.08.01424075309  [Google Scholar]  [CrossRef]  [PubMed]

[35]Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, The evaluation of Tumour-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014 Ann Oncol 2014 26(2):259-71.10.1093/annonc/mdu45025214542  [Google Scholar]  [CrossRef]  [PubMed]

[36]Tomioka N, Azuma M, Ikarashi M, Yamamoto M, Sato M, Watanabe KI, The therapeutic candidate for immune checkpoint inhibitors elucidated by the status of tumour-infiltrating lymphocytes (TILs) and Programmed death ligand 1 (PD-L1) expression in Triple negative breast cancer (TNBC) Breast Cancer 2018 25(1):34-42.10.1007/s12282-017-0781-028488168  [Google Scholar]  [CrossRef]  [PubMed]

[37]Hortobagyi GN, Connolly JL, D’Orsi CJ, Edge SB, Mittendorf EA, Rugo HS, Breast. In: Edge SB, Greene FL, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, et al., editors AJCC (American Joint Committee on Cancer) cancer staging manual 2017 8th edNew YorkSpringer-Verlag:570-610.  [Google Scholar]

[38]Zhou SJ, Bi TQ, Qin CX, Yang XQ, Pang K, Expression of NAMPT is associated with breast invasive ductal carcinoma development and prognosis Oncol Lett 2018 15(5):6648-54.10.3892/ol.2018.8164  [Google Scholar]  [CrossRef]

[39]Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thürlimann B, Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013 Ann Oncol 2013 24(9):2206-23.10.1093/annonc/mdt30323917950  [Google Scholar]  [CrossRef]  [PubMed]

[40]Curigliano G, Burstein HJ, Winer E, Gnant M, Dubsky P, Loibl S, De-escalating and escalating treatments for early-stage breast cancer: the St. Gallen International Expert Consensus Conference on the Primary Therapy of Early Breast Cancer 2017 Ann Oncol 2017 28(8):1700-12.10.1093/annonc/mdx30828838210  [Google Scholar]  [CrossRef]  [PubMed]

[41]Guo L, Li W, Zhu X, Ling Y, Qiu T, Dong L, PD-L1 expression and CD274 gene alteration in triple-negative breast cancer: Implication for prognostic biomarker Springerplus 2016 5(1):80510.1186/s40064-016-2513-x27390646  [Google Scholar]  [CrossRef]  [PubMed]

[42]Beckers RK, Selinger CI, Vilain R, Madore J, Wilmott JS, Harvey K, Programmed death ligand 1 expression in triple-negative breast cancer is associated with tumour-infiltrating lymphocytes and improved outcome Histopathology 2016 69(1):25-34.10.1111/his.1290426588661  [Google Scholar]  [CrossRef]  [PubMed]

[43]Duchnowska R, Pęksa R, Radecka B, Mandat T, Trojanowski T, Jarosz B, Immune response in breast cancer brain metastases and their microenvironment: the role of the PD-1/PD-L axis Breast Cancer Res 2016 18(1):4310.1186/s13058-016-0702-827117582  [Google Scholar]  [CrossRef]  [PubMed]

[44]Joneja U, Vranic S, Swensen J, Feldman R, Chen W, Kimbrough J, Comprehensive profiling of metaplastic breast carcinomas reveals frequent overexpression of programmed death-ligand 1 J Clin Pathol 2017 70(3):255-59.10.1136/jclinpath-2016-20387427531819  [Google Scholar]  [CrossRef]  [PubMed]

[45]Xing X, Guo J, Ding G, Li B, Dong B, Feng Q, Analysis of PD1, PDL1, PDL2 expression and T-cells infiltration in 1014 gastric cancer patients Onco Immunology 2018 7(3):e135614410.1080/2162402X.2017.135614429399387  [Google Scholar]  [CrossRef]  [PubMed]

[46]Rebelatto MC, Midha A, Mistry A, Sabalos C, Schechter N, Li X, Development of a programmed cell death ligand-1 immunohistochemical assay validated for analysis of non-small cell lung cancer and head and neck squamous cell carcinoma Diagn Pathol 2016 11(1):9510.1186/s13000-016-0545-827717372  [Google Scholar]  [CrossRef]  [PubMed]

[47]Dill EA, Gru AA, Atkins KA, Friedman LA, Moore ME, Bullock TN, PD-L1 expression and intratumoural heterogeneity across breast cancer subtypes and stages Am J Surg Pathol 2017 41(3):334-42.10.1097/PAS.000000000000078028195880  [Google Scholar]  [CrossRef]  [PubMed]

[48]Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study J Clin Oncol 2016 34(21):2460-67.10.1200/JCO.2015.64.893127138582  [Google Scholar]  [CrossRef]  [PubMed]

[49]Dirix LY, Takacs I, Jerusalem G, Nikolinakos P, Arkenau HT, Forero-Torres A, Avelumab, an anti-PD-L1 antibody, in patients with locally advanced or metastatic breast cancer: a phase 1b JAVELIN Solid Tumour study Breast Cancer Res Treat 2018 167(3):671-86.10.1007/s10549-017-4537-529063313  [Google Scholar]  [CrossRef]  [PubMed]

[50]Burugu S, Gao D, Leung S, Chia SK, Nielsen TO, LAG-3+ tumour infiltrating lymphocytes in breast cancer: clinical correlates and association with PD-1/PD-L1+ tumours Ann Oncol 2017 28(12):2977-84.10.1093/annonc/mdx55729045526  [Google Scholar]  [CrossRef]  [PubMed]

[51]Li F, Ren Y, Wang Z, Programmed death 1 Ligand 1 expression in breast cancer and its association with patients’ clinical parameters J Cancer Res Ther 2018 14(1):15010.4103/jcrt.JCRT_602_1729516978  [Google Scholar]  [CrossRef]  [PubMed]

[52]Hou Y, Nitta H, Wei L, Banks PM, Parwani AV, Li Z, Evaluation of immune reaction and PD-L1 expression using multiplex immunohistochemistry in HER2-positive breast cancer: The association with response to Anti-HER2 neoadjuvant therapy Clin Breast Cancer 2018 18(2):e237-44.10.1016/j.clbc.2017.11.00129198959  [Google Scholar]  [CrossRef]  [PubMed]

[53]Sabatier R, Finetti P, Mamessier E, Adelaide J, Chaffanet M, Ali HR, Prognostic and predictive value of PDL1 expression in breast cancer Oncotarget 2015 6(7):544910.18632/oncotarget.321625669979  [Google Scholar]  [CrossRef]  [PubMed]

[54]Soliman H, Khalil F, Antonia S, PD-L1 expression is increased in a subset of basal type breast cancer cells PloS One 2014 9(2):e8855710.1371/journal.pone.008855724551119  [Google Scholar]  [CrossRef]  [PubMed]

[55]Wimberly H, Brown JR, Schalper K, Haack H, Silver MR, Nixon C, PD-L1 expression correlates with tumour-infiltrating lymphocytes and response to neoadjuvant chemotherapy in breast cancer Cancer Immunol Res 2015 3:326-32.10.1158/2326-6066.CIR-14-013325527356  [Google Scholar]  [CrossRef]  [PubMed]

[56]Ghebeh H, Tulbah A, Mohammed S, ElKum N, Amer SM, Al-Tweigeri T, Expression of B7-H1 in breast cancer patients is strongly associated with high proliferative Ki-67-expressing tumour cells Int J Cancer 2007 121(4):751-58.10.1002/ijc.2270317415709  [Google Scholar]  [CrossRef]  [PubMed]

[57]Jang BS, Kim IA, A radiosensitivity gene signature and PD-L1 status predict clinical outcome of patients with invasive breast carcinoma in The Cancer Genome Atlas (TCGA) dataset Radiother Oncol 2017 124(3):403-10.10.1016/j.radonc.2017.05.00928579282  [Google Scholar]  [CrossRef]  [PubMed]