Open Access

Status and prognostic value of immunological biomarkers of breast cancer

  • Authors:
    • Chisa Murazawa
    • Nozomi Hashimoto
    • Kana Kuraishi
    • Mutsumi Motoyama
    • Shin-Ichiro Hashimoto
    • Mayumi Ikeuchi
    • Shoko Norimura
    • Toru Matsunaga
    • Koji Teramoto
    • Reiji Haba
    • Noriko Abe
    • Toshiki Yajima
    • Keiichi Kontani
  • View Affiliations

  • Published online on: March 8, 2023     https://doi.org/10.3892/ol.2023.13750
  • Article Number: 164
  • Copyright: © Murazawa et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The immune response to cancer serves an important role in disease progression and patient prognosis. For triple‑negative breast cancer showing aggressive behavior, immunotherapy has a good efficacy because of the potent immunogenicity of this type of cancer. However, the dominant subtype, luminal human epidermal growth factor receptor‑2 (HER2)‑negative breast cancer, is less immunogenic. To determine whether luminal HER2‑negative cancer reacts to the anticancer immune response, the present study analyzed the status and prognostic value of the principal immunological biomarkers of breast cancer, including tumor‑infiltrating lymphocytes (TILs), CD8+ T lymphocytes, the major histocompatibility complex and programmed cell death ligand‑1 (PD‑L1). The biomarkers were compared between patients with luminal HER2‑negative breast cancer and those with immunogenic subtypes including triple‑negative and HER2‑overexpressed breast cancer. A total of 71 patients with primary breast cancer were classified into the immunogenic non‑luminal (n=23) and less immunogenic luminal HER2‑negative groups (n=48) based on immunogenicity. In the luminal HER2‑negative group, compared with patients with low TIL levels, those with high TIL levels were at an advanced stage of cancer (P=0.024) and showed worse relapse‑free survival (P=0.057); however, the remaining biomarkers exhibited no association with cancer progression or prognosis. In the non‑luminal group, patients with high TIL levels showed significantly better RFS than those with low TIL levels (P=0.014). Compared with non‑luminal patients negative for PD‑L1, those positive for PD‑L1 exhibited better overall survival (P=0.064). Notably, TIL status was found to exhibit contrasting prognostic predictions based on immunogenicity. In conclusion, TILs are a strong candidate for prognostic prediction in breast cancer, regardless of the subtype. PD‑L1 is a potential candidate for prognostic prediction in immunogenic breast cancers, but not in the luminal HER2‑negative subtype.

Introduction

Several biomarkers predict cancer progression, patient prognosis, and therapeutic efficacy in cancer. Some of the most useful biomarkers are involved in the growth or metastasis of cancers (14). In breast cancer, the biomarker human epidermal growth factor receptor-2 (HER2) can be used to predict both patient prognosis and anti-HER2 therapeutic efficacy (511). The use of trastuzumab, a HER2-targeting agent, for treating patients with HER2-overexpressed breast cancer that exhibits aggressive clinical behaviors and poor prognosis has significantly improved the prognoses of these patients; for these patients, treatment with trastuzumab has resulted in a prognosis as favorable as that of patients with luminal HER2-negative breast cancer in both postoperative and metastatic settings (1115). However, for triple-negative (TN) breast cancer that exhibits clinical aggressiveness and lacks the expression of target molecules and biomarkers available for treatment, no efficient therapeutics have been established. Recent studies have found that the immune response plays a crucial role in the disease progression and prognosis of patients with cancer. Therefore, studies have investigated the use of immune checkpoint inhibitors targeting the programmed cell death 1-programmed cell death-ligand-1 (PD-1-PD-L1) axis in cancer treatment; this has resulted in dramatic positive effects against immunogenic cancers (1627). Although breast cancer is among the less immunogenic cancers (2830), certain aggressive subtypes with typically poor prognoses, such as TN and HER2-overexpressed breast cancer, have been found to be potently immunogenic (3133).

Clinical trials of the therapeutic potential of immune checkpoint inhibitors, such as atezolizumab and pembrolizumab, in the treatment of advanced TN breast cancer have achieved an objective treatment response rate of 53.2-56.0%, with a significantly longer progression-free survival compared with that of placebo group patients (3438). PD-1 and PD-L1 are molecules responsible for immune checkpoint processes. PD-L1 is expressed in various cells, including macrophages, monocytes, T cells, B cells, and tumor cells and binds to PD-1 receptors on T and B cells. PD-L1 is overexpressed in tumor cells and binds to PD-1 on cytotoxic T lymphocytes (CTLs). This initiates the lysis and apoptosis of cancer cells by CTLs. However, prolonged exposure to cancer cells can lead to CTL exhaustion, which reduces their ability to kill tumor cells (39). Inhibiting the interaction between PD-1 and PD-L1 prevents CTLs from becoming less responsive and helps them restore their cytotoxic activity against cancer cells. For the body to recognize cancer cells and ensure that CTLs attack them, immune cells should infiltrate the tumor mesenchyme. Immune cells can include antigen-presenting cells such as macrophages, dendritic cells, and B lymphocytes. Moreover, CD4+ helper lymphocytes and CD8+ CTLs can infiltrate the tumor mesenchyme and recognize antigenic epitopes present on major histocompatibility complex (MHC) molecules. Therefore, tumor-infiltrating lymphocytes (TILs) and MHC molecules are essential for inducing an antitumor immune response. Several studies have reported the utility of the level of TILs and CD8+ T lymphocyte infiltrates, and expression of MHC and PD-1-PD-L1 axis for prognostic prediction in cancer (4049). Cancer cells can gradually acquire the ability to evade the immune surveillance system of antitumor immune cells, thereby leading to cancer progression (50). Among these evasive tactics against antitumor immunity is the deletion of MHC class I molecules on the surface of cancer cells. This prevents the interaction of CTLs with T cell receptors, which is necessary for the recognition of the cancer cells by CTLs. Thus, MHC class 1 molecules also have prognostic significance (4850).

As described above, antitumor immune responses can affect cancer progression and patient prognosis in immunogenic subtypes such as TN and HER2-overexpressed breast cancer; however, antitumor immunity is unlikely to affect luminal HER2-negative breast cancer, a dominant subtype, because of its lower immunogenicity. Indeed, compared with other cancer types, breast cancer including luminal HER2-negative breast cancer, which occurs in a majority of the population, exhibits fewer mismatch repair deficiencies and microsatellite instabilities; this is partly because breast cancer is a well-differentiated and slow-growing cancer (28,51). This results in a low production of non-self antigenic proteins during cancer progression. This low immunogenicity has been verified in a clinical trial evaluating an immune checkpoint inhibitor; in the trial, the treatment was inefficacious in patients with luminal HER2-negative breast cancer compared with the beneficial effects observed in those with TN breast cancer (30).

In this study, we retrospectively evaluated the status and prognostic value of the immunological breast cancer biomarkers, TILs, CD8+ T lymphocyte infiltrates, MHC molecules, and PD-L1. We compared these biomarkers between patients with less immunogenic luminal HER2-negative breast cancer and those with immunogenic non-luminal breast cancer including TN and non-luminal HER2-overexpressed breast cancers.

Materials and methods

Patients

Seventy-one female patients with primary breast cancer who had undergone surgery such as mastectomy or partial resection for primary lesions with either axillary dissection or sentinel node biopsy from January 2010 to December 2021 at Kagawa University Hospital were included in this study. The exclusion criteria were as follows: previous invasive breast cancer or non-breast cancer within 5 years before surgery for primary breast cancer; any previous chemotherapy or endocrine therapy for cancer; any previous anti-HER2 therapy or other previous anticancer biologic therapy or immunotherapy; and concurrent serious diseases interfering with adjuvant therapy for breast cancer. The median patient age was 59 (35–85) years. At the time of surgery, 27 of the patients were in clinical stage 1, 42 were in stage 2, and 2 were in stage 3 (Table I). The cohort comprised 48 patients with luminal HER2-negative, 21 with TN, and 2 with non-luminal HER2-overexpressed breast cancer. Tissue samples of the main breast tumor obtained by either surgical resection or preoperative biopsy were examined.

Table I.

Clinical features and prognoses of the primary breast cancer patient cohort in this study.

Table I.

Clinical features and prognoses of the primary breast cancer patient cohort in this study.

VariableAll (n=71)Non-luminal (n=23)Luminal HER2 (−) (n=48)P-value
Median age, years59 (31–85)58 (31–78)60 (32–85)0.681
Median tumor size, cm2 (0.5-8.5)2.1 (0.5-6.5)1.7 (0.7-8.0)0.269
N-positive, %43.6%60.9%31.3%0.018a
Stage
  1274230.014a
  24219230.006a
  32020.324
MHC-positive, %70.4%78.3%66.7%0.319
High TILs, %60.6%82.7%50.0%0.009a
Median no. CD8+ T, %66.0 (1.0-176.3)88.0 (17.3-176.3)55.7 (1.0-130.0)0.001a
PDL1-E1L3N (+), %19.7%39.1%10.4%0.005a
PDL1-SP263 (+), %54.9%60.9%47.9%0.174
RFS at 10 years60.3%52.1%66.7%0.059
OS at 10 years78.1%60.9%87.5%0.038a

a P<0.05. RFS, relapse-free survival; OS, overall survival; MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes Median age and tumor size, and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer and that of patients with each clinical stage, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test. The duration of RFS and OS was assessed using Kaplan-Meier survival analyses and log-rank tests.

Evaluation of TIL levels

TIL levels in patient tissue samples were evaluated. After the samples were fixed in formalin and embedded in paraffin, they were sectioned into 4-µm-thick slices and stained in hematoxylin-eosin solution, as previously described (52). All mononuclear cells, including lymphocytes and plasma cells, were selected for the evaluation of TIL levels. Granulocytes and other polymorphonuclear leukocytes were excluded. As recommended in previous studies, stromal TIL levels were determined according to the area of stromal tissues occupied by mononuclear inflammatory cells over the total intratumoral stromal area (=% stromal TILs). The denominator used to determine the % stromal TIL level is the area of stromal tissue and not the number of stromal cells (5254). Three representative fields of view were selected and the average of each TIL level was determined. We used a cutoff score of 10% as previously established (55). Therefore, a stromal TIL level ≥10% was designated as a high TIL level, and that <10% was designated as a low TIL level.

Immunohistochemistry

Serial sections (4-µm-thick) of formalin-fixed paraffin-embedded tissue specimens were stained via standard indirect immunoperoxidase procedures for PD-L1, CD8, and MHC class I molecules, according to the staining kit manufacturer's instructions. Briefly, each tissue section was deparaffinized in xylene and rehydrated in ethanol and distilled water. Antigen retrieval was performed via 10 min of microwave treatment in 10 mM sodium citrate buffer (pH 6.0) for PD-L1 or 10 mM Tris/1 mM ethylenediaminetetraacetic acid (pH 9.0) for MHC class I molecules. Endogenous peroxidase activity was blocked by treatment with 3% H2O2 for 10 min. After blocking in Tris-buffered saline with Tween-20 and 5% normal goat serum for 1 h at room temperature, the sections were incubated at 4°C overnight with antihuman PD-L1 monoclonal antibodies (clone: E1L3N, diluted 1:200, Cell Signaling Technology, Danvers, MA, USA; SP263, diluted 1:100, Ventana Medical Systems, Tucson, AZ, USA), which were produced by immunizing rabbits with peptides derived from the C-terminus of PD-L1 protein, anti-CD8 monoclonal antibody (clone: SP57, diluted 1:100, Ventana Medical Systems), or an anti-HLA class I monoclonal antibody (clone: EMR8-5, diluted 1:500, Hokudo Co., Ltd., Sapporo, Japan). The sections were then incubated with SignalStain boost IHC detection reagent (Cell Signaling Technology) for PD-L1 or Envision Dako ChemMate (Dako Ltd., Kyoto, Japan) for CD8 and MHC class I molecules. They were visualized using a SignalStain DAB (3,3′-diaminobenzidine) substrate kit (Cell Signaling Technology) for PD-L1 or Envision Dako ChemMate/horseradish peroxidase (HRP) DAB for CD8 and MHC class I molecules for 1 min. This was followed by counterstaining with hematoxylin. Isotype-matched control antibodies were used for immunohistochemistry. These were rabbit immunoglobulin G (IgG) monoclonal antibody (Cell Signaling Technology) for PD-L1, and mouse IgG monoclonal antibody (Dako) for CD8 and MHC class I molecules.

Evaluation of PD-L1, MHC class 1, and CD8 expression

Serial sections of stained tumor tissues were independently examined by two researchers, including a pathologist. To compare the cellular staining intensities of PD-L1, CD8, and MHC class I molecules, cells from the serial sections were evaluated microscopically (magnification: ×200). Three representative fields of view were selected and any expression of PD-L1 and MHC class I molecules was identified in 100 tumor cells per field. Cases in which the proportion of tumor cells positive for PD-L1 was ≥1% were considered PD-L1+ tumor cell-dominant. Cases in which the proportion of tumor cells positive for MHC class I molecules was ≥80% were considered MHC class I+ tumor cell-dominant, as previously reported (56). To evaluate CD8+ T lymphocyte levels, we counted the number of CD8+ T lymphocytes in three stroma fields of view and calculated the median number of CD8+ T lymphocytes per field.

Statistical analysis

All statistical analyses were performed using SPSS Statistics for Windows (IBM Corp., Armonk, NY, USA) software. For comparisons between two groups, we used the Mann-Whitney U test or the χ2 test. The effects of clinical and demographic variables, clinical responses, and prognostic parameters on the duration of survival and risk of progression were assessed using Kaplan-Meier survival analyses and log-rank tests. A 95% confidence interval for the median of each variable was calculated using the Brookmeyer and Crowley method (57). All analyses were two-sided and P<0.05 was considered to indicate a statistically significant difference.

Results

Association of clinicopathological patient variables and immunological biomarker status with cancer progression and prognosis

The patient cohort included 48 (67.6%) patients with luminal HER2-negative, 21 (29.6%) with TN, and two (2.8%) with non-luminal HER2-overexpressed breast cancer. At the time of the study, 30 patients experienced relapsed lesions. The relapse-free survival (RFS) and overall survival (OS) rates 10 years after the primary operation were 60.3 and 78.1%, respectively (Table I). There were 50 (70.4%) patients positive for MHC expression and 43 (60.6%) with high TIL levels (Tables I and II). Microscopic images of low and high TIL levels are shown in Fig. 1A and B, respectively. Microscopic images of breast cancer tumors positive and negative for MHC expression are shown in Fig. 1C and D, respectively. Reactivity to E1L3N was observed in 14 (19.7%) patients and reactivity to SP263 in 39 (54.9%) patients (Tables I and III). Microscopic images of tumors positive and negative for E1L3N and SP263 were shown in Fig. 2. Although the sensitivity of reaction of the two monoclonal antibodies observed differed, all patients responsive to E1L3N exhibited reactivity to SP263 because both the monoclonal antibodies recognized antigenic determinants near the C-terminus of PD-L1 protein. Furthermore, MHC expression was significantly associated with tumor size (P=0.017) and clinical stage (P=0.046) and TIL level was significantly associated with tumor size (P=0.021), nodal involvement (P=0.004), and clinical stage (P=0.006); however, neither MHC expression nor TIL level was associated with RFS or OS (Table II; Figs. 3 and 4). The number of CD8+ T lymphocyte infiltrates in the tumor stroma was significantly associated with MHC and PD-L1 expression and TIL levels (Table II). Microscopic images of low and high counts of CD8+ T lymphocyte infiltrates are shown in Fig. 5A and B, respectively. The proportion of patients positive for SP263 was significantly higher in patients positive for E1L3N than that in patients negative for E1L3N (E1L3N-negative and E1L3N-positive patients: 42.1 and 100%, respectively, P<0.001, Table III) and the proportion of patients positive for E1L3N was significantly higher in patients positive for SP263 than that in patients negative for SP263 (SP263-negative and SP263-positive patients: 0 and 36.8%, P<0.001). Regarding the status of these immunological biomarkers, the proportion of patients with high TIL levels, the proportion of patients reactive to E1L3N, and the number of CD8+ T lymphocyte infiltrates were significantly higher in the non-luminal group than in the luminal group (high TIL levels, 82.7% vs. 50.0%, P=0.009; E1L3N positivity, 39.1% vs. 10.4%, P=0.005; number of CD8+ T lymphocyte infiltrates, 88.0 vs. 55.7, P=0.001, Table I). However, the status of these biomarkers showed no prognostic value, except for an almost but not quite significant association between E1L3N reactivity and shorter RFS (RFS rate at 10 years: 66.1 and 42.9% for patients negative and positive for E1L3N, respectively; P=0.052; Figs. 3 and 4).

Table II.

Relationships between MHC and TILs status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of the patient cohort.

Table II.

Relationships between MHC and TILs status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of the patient cohort.

VariableMHC(−) (n=21)MHC(+) (n=50)P-valueLow TIL (n=28)High TIL (n=43)P-value
Median age, years61 (46–73)57 (31–85)0.28660 (37–77)58 (32–85)0.861
Median tumor size, cm1.7 (0.5–3.5)2.1 (0.7–6.6)0.017a1.5 (0.7–3.5)2.1 (0.5–8.0)0.021a
N-positive, %27.2%45.1%0.22921.4%55.6%0.005a
Median stage1 (12)2 (13)0.046 a1 (12)2 (13)0.006a
MHC-positive, %---50.0%83.7%0.004a
High TILs, %31.8%70.6%0.004 a---
Median no. CD8+ T39.7 (1.0–109.7)74.3 (27.0–176.3)0.001a33.7 (1.0–74.3)83.3 (31.0–176.3) <0.001a
PDL1-E1L3N (+), %0%27.5%0.007a3.6%30.2%0.008a
PDL1-SP263 (+), %31.8%60.8%0.016a32.1%65.1%0.018a

a P<0.05. MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes. Median age, stage and tumor size, and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test.

Table III.

Relationships between PD-L1 status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of the patient cohort.

Table III.

Relationships between PD-L1 status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of the patient cohort.

VariableE1L3N(−) (n=57)E1L3N(+) (n=14)P-valueSP263(−) (n=32)SP263(+) (n=39)P-value
Median age, years60 (31–75)57 (41–85)0.71462 (32–77)58 (31–85)0.328
Median tumor size, cm2.0 (0.5–8.0)2.0 (1.3–6.5)0.6241.9 (0.5–8.0)2.0 (0.7–6.5)0.766
N-positive, %39.0%57.1%0.21941.2%41.0%0.791
Median stage2 (13)2 (12)0.5532 (13)2 (12)0.705
MHC-positive, %63.2%100%0.007a57.6%81.6%0.016a
High TILs, %52.6%92.90.008a45.5%73.7%0.018a
Median no. CD8+ T58.3 (1.0–125.7)100.3 (54.0–176.3) <0.001a43.3 (12.0–109.7)79.0 (1.0–176.3) <0.001a
PDL1-E1L3N (+), %---0%36.8% <0.001a
PDL1-SP263 (+), %42.1%100% <0.001a---

a P<0.05. MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes. Median age, stage and tumor size and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test.

Associations of immunological biomarker status with cancer progression and prognosis in patients with immunogenic non-luminal cancer

The non-luminal group included 21 patients with TN and 2 with non-luminal HER2-overexpressed breast cancer. The proportion of patients positive for PD-L1 expression was significantly higher in patients positive for MHC expression than that in patients negative for MHC expression (P=0.048 for E1L3N and P=0.019 for SP263, Table IV). There was no difference in the proportion of patients with high TIL levels between MHC status (MHC-negative and MHC-positive patients: 60.0 and 88.9%, respectively, P=0.140).) Reciprocally, the proportion of patients with MHC expression was significantly higher in patients positive for E1L3N or SP263 than that in patients negative for PD-L1 expression (E1L3N-negative and E1L3N-positive patients: 64.3 and 100%, respectively, P=0.048; SP263-negative and SP263-positive patients: 50.0 and 93.3%, respectively, P=0.019, Table V). No association of high and low TIL levels with MHC and PD-L1 expression was observed (P=0.140 with MHC, P=0.084 for E1L3N, and P=0.069 for SP263). In the non-luminal group, compared with patients with low TIL levels, patients with high TIL levels showed significantly longer RFS (low levels: median RFS of 14 months; high levels: RFS rate of 63.2% at 10 years, P=0.014; Fig. 6); however, TIL levels were not associated with cancer progression (Table IV). Of the remaining markers in this group, SP263 reactivity was associated with prognosis, with reactive patients showing slightly better OS rates 10 years after their primary operation compared with nonreactive patients (37.5 and 73.3%, respectively, P=0.064; Fig. 7).

Table IV.

Relationships between MHC and TILs status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with non-luminal breast cancer.

Table IV.

Relationships between MHC and TILs status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with non-luminal breast cancer.

VariableMHC(−) (n=5)MHC(+) (n=18)P-valueLow TIL (n=4)High TIL (n=19)P-value
Median age, years66 (53–77)57 (31–78)0.14147 (3160)61 (40–78)0.109
Median tumor size, cm1.8 (0.5–3.5)2.2 (1.2–6.5)0.3593.3 (1.0–4.0)2.0 (0.5–6.5)0.545
N-positive, %60.0%61.1%0.96575.0%57.9%0.533
Median stage2 (12)2 (12)0.3101 (12)2 (12)0.750
MHC-positive, %---50.0%84.2%0.140
High TILs, %60.0%88.9%0.140---
Median no. CD8+ T31.0 (17.3–109.7)97.8 (32.7–176.3)0.05228.7 (17.3–74.3)99.7 (51.7–176.3)0.007a
PDL1-E1L3N (+), %0%50.0%0.048a0%47.3%0.084
PDL1-SP263 (+), %20.0%77.8%0.019a25.0%73.7%0.069

a P<0.05. MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes. Median age, stage and tumor size, and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test.

Table V.

Relationships between PD-L1 status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with non-luminal breast cancer.

Table V.

Relationships between PD-L1 status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with non-luminal breast cancer.

VariableE1L3N(−) (n=14)E1L3N(+) (n=9)P-valueSP263(−) (n=8)SP263(+) (n=15)P-value
Median age, years61 (31–77)53 (40–76)0.25064 (41–77)56 (31–76)0.125
Median tumor size, cm2.4 (0.5–3.5)2.0 (1.2–6.5)0.7852.6 (0.5–3.5)2.1 (1.2–6.5)0.716
N-positive, %57.1%66.7%0.65562.5%60.0%0.909
Median stage2 (12)2 (12)0.5202 (12)2 (12)0.150
MHC-positive, %64.3%100%0.048a50.0%93.3%0.019a
High TILs, %71.4%100%0.08462.5%93.3%0.069
Median no. CD8+ T75.3 (17.3–125.7)101.0 (71.7–176.3)0.012a42.2 (17.3–109.7)101.0 (71.7–176.3)0.004a
PDL1-E1L3N (+), %---0%60.0%0.006a
PDL1-SP263 (+), %42.9%100%0.006a---

a P<0.05. MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes. Median age, stage tumor size, and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test.

Associations of immunological biomarker status with progression and prognosis in patients with luminal HER2-negative cancer

In patients with luminal HER2-negative cancer, the proportion of patients positive for MHC expression and median number of CD8+ T lymphocyte infiltrates per patient were significantly higher in patients with high TIL levels than those in patients with low TIL levels (MHC-positive patients: low TIL and high TIL levels, 50.0 and 83.3%, respectively, P=0.016; median CD8+ T lymphocyte counts: low TIL and high TIL levels, 39.8 and 62.3, respectively, P<0.001, Table VI); moreover, a significantly higher number of patients with high TIL levels were in a more advanced stage of cancer compared with patients with low TIL levels (P=0.024). Neither MHC expression nor TIL levels showed any association with PD-L1 expression (MHC, P=0.099 for E1L3N and P=0.312 for SP263; TIL, P=0.161 for E1L3N and P=0.153 for SP263). Furthermore, patients with high TIL levels showed a marginal trend to significance of having a shorter RFS than those with low TIL levels (RFS rate at 10 years: 79.2 and 58.3% for low and high TIL levels, respectively; P=0.057, Fig. 8). Neither MHC nor PD-L1 expression was associated with progression or prognosis in this group (Figs 8 and 9; Tables VI and VII). Remarkably, the association between high TIL levels and shorter RFS in this subtype group was contrary to that observed in the immunogenic group, in which high TIL levels were associated with longer RFS.

Table VI.

Relationships between MHC and TILs status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with luminal HER2-negative breast cancer.

Table VI.

Relationships between MHC and TILs status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with luminal HER2-negative breast cancer.

VariableMHC(−) (n=16)MHC(+) (n=32)P-valueLow TIL (n=24)High TIL (n=24)P-value
Median age, years61 (46–73)58 (32–85)0.64161 (45–77)58 (32–85)0.411
Median tumor size, cm1.7 (0.7–3.1)1.9 (0.7–8.0)0.0631.5 (0.7–3.1)2.4 (0.8–8.0)0.009a
N-positive, %18.8%31.3%0.19112.5%50.0%0.006a
Median stage1 (12)2 (13)0.2701 (13)2 (13)0.024a
MHC-positive, %---50.0%83.3%0.016a
High TILs, %25.0%62.5%0.016a---
Median no. CD8+ T39.7 (1.0–75.3)62.3 (24.7–137.0)0.002a39.8 (1.0–66.7)62.3 (24.7–137.0) <0.001a
PDL1-E1L3N (+), %0%12.5%0.0994.2%16.7%0.161
PDL1-SP263 (+), %37.5%53.1%0.31337.5%58.3%0.153

a P<0.05. MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes. Median age, stage and tumor size, and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test.

Table VII.

Relationships between PD-L1 status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with luminal HER2-negative breast cancer.

Table VII.

Relationships between PD-L1 status of breast cancer and cancer progression and prognosis as determined by the clinicopathological feature of patients with luminal HER2-negative breast cancer.

VariableE1L3N(−) (n=43)E1L3N(+) (n=5)P-valueSP263(−) (n=25)SP263(+) (n=23)P-value
Median age, years59 (32–77)63 (53–85)0.35462 (32–77)59 (38–85)0.904
Median tumor size, cm1.7 (0.5–3.5)1.5 (1.3–4.0)0.9071.7 (0.7–8.0)1.5 (0.7–4.0)0.329
N-positive, %27.9%40.0%0.65936.0%26.1%0.464
Median stage2 (13)1 (12)0.5172 (13)1 (12)0.336
MHC-positive, %62.8%100%0.09860.0%73.9%0.312
%High TILs, %46.5%80.0%0.16140.0%60.8%0.153
Median no. CD8+ T53.3 (1.0–108.0)80.0 (54.0–130.0)0.09742.0 (12.0–120.3)66.0 (1.0–130.0)0.021a
PDL1-E1L3N (+), %---0%21.7%0.015a
PDL1-SP263 (+), %41.9%100%0.015a---

a P<0.05. MHC, major histocompatibility complex; TILs, tumor-infiltrating lymphocytes. Median age, stage and tumor size, and the number of CD8+ T lymphocytes were compared between two groups using Mann-Whitney-U test. Positive rates of MHC and PD-L1 expression, the proportion of patients with N-positive cancer, and high TIL rates were compared between two groups using χ2 test or Fisher's exact test.

Discussion

In patients with cancer, the antitumor immune response is crucial to the regulation of cancer progression and improvement of prognosis. However, cancer cells possess a wide range of mechanisms for evading host immune responses including the modification of cancer phenotypes, reduction or deletion of the expression of antigenic proteins and MHC molecules, and production of cytokines and factors that inhibit anticancer immune response activation (28,5865). Recently, immune checkpoint inhibitors that bind to PD-1- or PD-L1-inactivating CTLs have been found to be efficacious against reduced antitumor immunity, with the ability to restart the immune response to cancer when it slows or stops (1627). Several clinical trials have demonstrated drastically improved prognosis in patients with cancer when immune checkpoint inhibitors are added to chemotherapeutic agents (1622,2427). Breast cancer is among the less immunogenic cancers and tends to be minimally affected by antitumor immunity (28,29). In fact, TIL levels in this population are not sufficiently high to exhibit prognostic value (52,66). Nevertheless, some subtypes of breast cancer such as TN and HER2-overexpressed cancer both of which can grow rapidly and show aggressive behavior have been reported to be sensitive to antitumor immune responses and have demonstrated favorable responses to immune checkpoint inhibitors (3438). Immunological biomarkers, including TILs, CD8+ T lymphocyte infiltrates, MHC, and the PD-1-PD-L1 axis, have been found to be useful for predicting cancer progression and prognosis of patients with the immunogenic subtypes of breast cancer (4049). However, these markers have been found to predict different prognostic outcomes (35,4042,67) owing to differences in patient backgrounds and disease stage as well as the proportion of each subtype and combination of biomarkers studied. Furthermore, no report has analyzed associations of cancer progression and patient prognosis with the four principal immunological biomarkers simultaneously in each subtype of breast cancer. Therefore, although immunological biomarkers are expected to be useful in breast cancer, it remains unclear whether they have any prognostic value, particularly in luminal HER2-negative breast cancer.

We evaluated the status of immunological biomarkers, including TIL levels, CD8+ T lymphocyte infiltrate count, MHC expression, and PD-L1 expression, in the tumors of 71 patients with primary breast cancer to determine their utility as predictors of cancer progression and prognosis. To date, only B cells and macrophages in TILs have been found to predict survival rates in luminal HER2-negative breast cancer (52); to the best of our knowledge, the prognostic value of other biomarkers has not been previously evaluated in this population. This is the first report on the status and prognostic value of principal immunological biomarkers such as MHC, TILs, CD8+ T lymphocyte infiltrates, and PD-L1 in luminal HER2-negative and other breast cancer subtypes. Monoclonal antibodies against PD-1 or PD-L1 (SP142 and 22C3) currently available as a companion diagnostic agent for potential breast cancer treatment with immune checkpoint inhibitors have demonstrated different prognostic capacities (3741,67). These monoclonal antibodies exhibited positivity in <5% of patients with luminal HER2-negative breast cancer in our preliminary study (data not shown). Therefore, in the present study, we used alternative clones (E1L3N and SP263) available for use with non-small cell lung cancer (35,56,6870).

We observed that PD-L1 expression (reactive with both E1L3N and SP263) was generally associated with MHC expression in tumor cells and with stromal TIL levels (Table III). We further found that CD8+ T lymphocyte infiltrate counts were significantly associated with the status of all biomarkers in all breast cancer subtypes. These results suggest that according to breast cancer subtype, CD8+ CTLs in stromal TILs can recognize tumor antigens to varying degrees in an MHC-restricted manner and lyse cancer cells. Furthermore, the CTL response to breast cancer can be inactivated by PD-1-PD-L1 interaction.

The present study results suggested that MHC and TIL status were strongly associated with cancer progression and that PD-L1 status (in terms of its reactivity with E1L3N) exhibited possible prognostic value for all breast cancer subtypes. To determine whether the status of these biomarkers differed between cancer subtypes and whether they interacted with cancer progression or prognosis in each subtype, the patients were classified into two cancer subtype groups according to immunogenicity: patients with less immunogenic luminal HER2-negative cancer and those with immunogenic non-luminal breast cancers. In the immunogenic group, no association was observed between TIL levels and MHC expression; however, TIL levels and MHC expression were closely associated with PD-L1 expression (Tables IV and V). Of the patients who tested negative for MHC expression, those reactive to E1L3N and SP263 accounted for only 0 and 20% (1 case), respectively (Table IV). Similarly, the number of patients reactive to E1L3N and SP263 among those with low TIL levels was quite low (no case and 1 case, respectively). Indeed, there is a small population who is deficient in biomarker expression in immunogenic breast cancer subtypes. These patients are unlikely to be affected by antitumor immunity. Furthermore, TIL status was found to be a good predictor of prognosis in this group, with high TIL levels indicating significantly longer RFS. These results are consistent with those of previous studies in that high TIL levels were associated with good prognoses in TN and HER2-overexpressed breast cancer (52,66). Moreover, high SP263 reactivity was associated with longer OS (Fig. 7D). The close relationship of PD-L1 expression with good patient prognosis has previously been reported in several studies (4042,67), and our results are consistent with those findings. Therefore, TIL levels and PD-L1 expression can be useful prognostic biomarkers in immunogenic breast cancer. However, contrary to our expectations, none of the biomarkers was associated with cancer progression in this subtype. It is possible that antitumor immunity is merely one of the factors influencing cancer progression. Tumor characteristics such as growth ability, differentiation grade, and metastatic ability, are also likely to considerably contribute to cancer progression.

In the less immunogenic luminal HER2-negative breast cancer group, high TIL levels were strongly associated with cancer progression and associated with poor prognoses (Table VI and Fig. 8B). In the luminal group, among patients negative for MHC expression or those with low TIL levels, only few patients exhibited reactivity to both anti-PD-L1 monoclonal antibodies (Table VI). Therefore, it is difficult to arrive at a conclusion from the data of patients who tested negative for biomarker expression. The other biomarkers showed no association with either cancer progression or prognosis. Remarkably, the relationship between high TIL levels and poor prognosis in the luminal HER2-negative group was contrary to that observed in the immunogenic non-luminal group, in which high TIL levels were associated with good prognoses. This suggests that in the immunogenic non-luminal population, TILs in the tumor stroma contribute to the immunosuppression of cancer, thereby prolonging RFS. Conversely, the lower level of immunogenicity in luminal HER2-negative tumor cells reduces their receptivity to host immune responses, thus allowing more aggressive growth and progression. Based on the relationship observed between high TIL levels and poor patient prognosis as well as the significantly lower proportion of patients with PD-L1 expression or those with high TIL levels in the luminal group compared with those in the non-luminal group, we confirmed that luminal HER2-negative breast cancer is less immunogenic. Therefore, TIL status in different breast cancer subtypes appears to reflect the distinct microbiology of tumor cells of the given subtype, in terms of the marked difference in their susceptibility to host immune responses. In the literature, only two studies have reported the relationship between TIL levels and patient prognosis in luminal HER2-negative breast cancer. Denkert et al (52) reported significant correlations between high TIL levels and shorter OS. The result was consistent with the findings of the present study. However, the other study reported no significant relationship between them (66). In both studies, good correlations were observed between high TIL levels and favorable patient prognoses in TN and HER2-overexpressed breast cancer subtypes. To clarify the association between TIL levels and prognosis, further studies including larger cohorts of patients with luminal HER2-negative cancer are required; these studies should aim to perform a detailed analysis for determining the lymphocyte and antigen-presenting cell populations that infiltrate the tumor stroma and the specific cytokines (e.g., interferon-gamma or tumor growth factor-beta) responsible for immune activation.

One of the limitations of our study is the small sample size (n=71); we thus could not classify a sufficient number of patients into groups to perform more convincing comparative analyses. Furthermore, we did not analyze systemic immunological responses, such as leukocyte profiles in peripheral blood, immunoglobulin and complement levels, or cytokine production in the studied patients. By including analysis of systemic immunological responses in patients with breast cancer in a future study, we will be able to understand the role of antitumor immunity more comprehensively in breast cancer. In this study, we used two anti-PD-L1 monoclonal antibodies, which were produced by immunizing rabbits with synthetic peptides derived from residues near the C-terminus of PD-L1 protein. The sensitivity of SP263 in detecting PD-L1 expression was generally higher than that of E1L3N. Although the precise epitopes of both monoclonal antibodies has not been reported, these may be different but located nearby. As these antibodies recognize their antigenic determinants on the three-dimensional components of the target protein in immunological assays, the sensitivity of each monoclonal antibody is expected to differ. Regarding the prognostic value of the PD-L1 status in luminal HER2-negative breast cancer, Zhang et al (67) found no association between PD-L1 expression and OS; this finding was consistent with our results. However, significant associations were observed between PD-L1 expression and survival rates in TN and HER2-overexpressed breast cancer subtypes.

In conclusion, the immunological biomarkers MHC, TIL, and PD-L1 exhibited different patterns of expression depending on the breast cancer subtype of the patient. However, CD8+ T lymphocyte infiltrate counts were closely associated with TIL levels and MHC and PD-L1 expression regardless of the breast cancer subtype. Of these biomarkers, only TIL levels are expected to be associated with cancer progression and patient prognosis, regardless of the breast cancer subtype. Although the PD-L1 protein reactive to SP263 is a potential prognostic biomarker in immunogenic cancers, it is unrelated to either cancer progression or patient prognosis in luminal HER2-negative breast cancer.

Acknowledgements

The authors would like to thank Ms. Hiromi Kita and Ms. Miho Takigawa (Department of Thoracic, Breast and Endocrine Surgery, Kagawa University Faculty of Medicine, Kagawa, Japan) for their editorial assistance with an earlier version of this manuscript.

Funding

This study was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Science, Sports and Culture, Japan (grant nos. 10671249, 13671380, 14571262 and 15591340).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

CM, KeK and KT conceived and designed the present study. KaK, SN, SH, MM and TM contributed to data acquisition and analysis. KeK, CM, TY and RH were major contributors in writing the manuscript. TY RH and NH were involved in data interpretation and discussion. NA and MI performed the statistical analysis. NH, NA and MI confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The research protocol for this study complied with the guidelines of the Ethics Committee at Kagawa University Hospital and was approved by the ethical review board of Kagawa University (approval no. HEISEI23-085); it conformed to the provisions in the Declaration of Helsinki in 1995. Written informed consent to participate was obtained from all study participants.

Patient consent for publication

When patients were given written information about the present study, written patient consent for publication was also obtained.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CD

cluster of differentiation

CTL

cytotoxic T lymphocyte

DAB

3,3′-diaminobenzidine

HER2

human epidermal growth factor receptor-2

MHC

major histocompatibility complex

OS

overall survival

PBS

phosphate-buffered saline

PD-1

programmed cell death 1

PD-L1

programmed cell death-ligand-1

RFS

relapse-free survival

TIL

tumor-infiltrating lymphocyte

TN

triple-negative

References

1 

Molina R, Jo J, Filella X, Zanon G, Pahisa J, Mu noz M, Farrus B, Latre ML, Escriche C, Estape J and Ballesta AM: c-erbB-2 oncoprotein, CEA, and CA 15.3 in patients with breast cancer: Prognostic value. Breast Cancer Res Treat. 51:109–119. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Berry DA, Cirrincione C, Henderson IC, Citron ML, Budman DR, Goldstein LJ, Martino S, Perez EA, Muss HB, Norton L, et al: Estrogen-receptor status and outcomes of modern chemotherapy for patients with node-positive breast cancer. JAMA. 295:1658–1667. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Bast RC Jr, Ravdin P, Hayes DF, Bates S, Fritsche H Jr, Jessup JM, Kemeny N, Locker GY, Mennel RG, Somerfield MR, et al: 2000 Update of recommendations for the use of tumor markers in breast and colorectal cancer: Clinical practice guidelines of the American society of clinical oncology. J Clin Oncol. 19:1865–1878. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Penault-Llorca F, André F, Sagan C, Lacroix-Triki M, Denoux Y, Verriele V, Jacquemier J, Baranzelli MC, Bibeau F, Antoine M, et al: Ki67 expression and docetaxel efficacy in patients with estrogen receptor-positive breast cancer. J Clin Oncol. 27:2809–2815. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Yarden Y: The EGFR family and its ligands in human cancer. Signalling mechanisms and therapeutic opportunities. Eur J Cancer. 37 (Suppl 4):S3–S8. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A and McGuire WL: Human breast cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science. 235:177–182. 1987. View Article : Google Scholar : PubMed/NCBI

7 

Sjögren S, Inganäs M, Lindgren A, Holmberg L and Bergh J: Prognostic and predictive value of c-erbB-2 overexpression in primary breast cancer, alone and in combination with other prognostic markers. J Clin Oncol. 16:462–469. 1998. View Article : Google Scholar : PubMed/NCBI

8 

Gabos Z, Sinha R, Hanson J, Chauhan N, Hugh J, Mackey JR and Abdulkarim B: Prognostic significance of human epidermal growth factor receptor positivity for the development of brain metastasis after newly diagnosed breast cancer. J Clin Oncol. 24:5658–5663. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Vogel CL, Cobleigh MA, Tripathy D, Gutheil JC, Harris LN, Fehrenbacher L, Slamon DJ, Murphy M, Novotny WF, Burchmore M, et al: Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER2-overexpressing metastatic breast cancer. J Clin Oncol. 20:719–726. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Roche PC and Ingle JN: Increased HER2 with U.S. food and drug administration-approved antibody. J Clin Oncol. 17:4341999. View Article : Google Scholar : PubMed/NCBI

11 

Slamon D, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Dawood S, Broglio K, Buzdar AU, Hortobagyi GN and Giordano SH: Prognosis of women with metastatic breast cancer by HER2 status and trastuzumab treatment: An institutional-based review. J Clin Oncol. 28:92–98. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Sundquist M, Brudin L and Tejler G: Improved survival in metastatic breast cancer 1985–2016. Breast. 31:46–50. 2017. View Article : Google Scholar : PubMed/NCBI

14 

Smith I, Procter M, Gelber RD, Guillaume S, Feyereislova A, Dowsett M, Goldhirsch A, Untch M, Mariani G, Baselga J, et al: 2-Year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: A randomised controlled trial. Lancet. 369:29–36. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr, Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, et al: Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 353:1673–1684. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, Patt D, Chen TT, Berman DM and Wolchok JD: Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 33:1889–1894. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al: Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 363:711–723. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 369:134–144. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al: Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 366:2443–2454. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al: PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma. N Engl J Med. 372:311–319. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O'Donnell PH, Balmanoukian A, Loriot Y, et al: Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet. 387:1909–1920. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Fehrenbacher L, Spira A, Ballinger M, Kowanetz M, Vansteenkiste J, Mazieres J, Park K, Smith D, Artal-Cortes A, Lewanski C, et al: Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet. 387:1837–1846. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, et al: Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 515:563–567. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Loi S, Adams S, Schmid P, Cortes J, Cescon DW, Winer EP, Toppmeyer DL, Rugo HS, De Laurentiis M, Nanda R, et al: Relationship between tumor infiltrating lymphocyte (TIL) levels and response to pembrolizumab (pembro) in metastatic triple-negative breast cancer (mTNBC): Results from KEYNOTE-086. Ann Oncol. 28 (Suppl 5):v6082017. View Article : Google Scholar

25 

AlHarbi M, Ali Mobark N, AlMubarak L, Aljelaify R, AlSaeed M, Almutairi A, Alqubaishi F, Hussain ME, Balbaid AAO, Said Marie A, et al: Durable response to nivolumab in a pediatric patient with refractory glioblastoma and constitutional biallelic mismatch repair deficiency. Oncologist. 23:1401–1406. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Overman MJ, McDermott R, Leach JL, Lonardi S, Lenz HJ, Morse MA, Desai J, Hill A, Axelson M, Moss RA, et al: Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): An open-label, multicentre, phase 2 study. Lancet Oncol. 18:1182–1191. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Overman MJ, Lonardi S, Wong KYM, Lenz HJ, Gelsomino F, Aglietta M, Morse MA, Van Cutsem E, McDermott R, Hill A, et al: Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer. J Clin Oncol. 36:773–779. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Fremda C, Hlevnjakb M, Zapatkab M, Zoernig I, Halamaa N, Fejzibegovica N, Thewesb V, Lichterb P, Schirmacherc P, Kloorc M, et al: Mismatch repair deficiency drives durable complete remission by targeting programmed death receptor 1 in a metastatic luminal breast cancer patient. Breast Care (Basel). 14:53–59. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Bates JP, Derakhshandeh R, Jones L and Webb TJ: Mechanisms of immune evasion in breast cancer. BMC Cancer. 18:5562018. View Article : Google Scholar : PubMed/NCBI

30 

Rugo HS, Delord JP, Im SA, Ott PA, Piha-Paul SA, Bedard PL, Sachdev J, Tourneau CL, van Brummelen EMJ, Varga A, et al: Safety and antitumor activity of pembrolizumab in patients with estrogen receptor-positive/human epidermal growth factor receptor 2-negative advanced breast cancer. Clin Cancer Res. 24:2804–2811. 2018. View Article : Google Scholar : PubMed/NCBI

31 

Zacharakis N, Huq LM, Seitter SJ, Kim SP, Gartner JJ, Sindiri S, Hill VK, Li YF, Paria BC, Ray S, et al: Breast cancers are immunogenic: Immunologic analyses and a phase II pilot clinical trial using mutation-reactive autologous lymphocytes. J Clin Oncol. 40:1741–1754. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Hoda RS, Brogi E, Dos Anjos CH, Grabenstetter A, Ventura K, Patil S, Selenica P, Weigelt B, Reis-Filho JS, Traina T, et al: Clinical and pathologic features associated with PD-L1 (SP142) expression in stromal tumor-infiltrating immune cells of triple-negative breast carcinoma. Mod Pathol. 33:2221–2232. 2020. View Article : Google Scholar : PubMed/NCBI

33 

Emens LA, Cruz C, Eder JP, Braiteh F, Chung C, Tolaney SM, Kuter I, Nanda R, Cassier PA, Delord JP, et al: Long-term clinical outcomes and biomarker analyses of atezolizumab therapy for patients with metastatic triple-negative breast cancer: A phase 1 study. JAMA Oncol. 5:74–82. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Nanda R, Chow LQ, Dees EC, Berger R, Gupta S, Geva R, Pusztai L, Pathiraja K, Aktan G, Cheng JD, et al: Pembrolizumab in patients with advanced triple-negative breast cancer: Phase Ib KEYNOTE-012 study. J Clin Oncol. 34:2460–2467. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Emens LA, Kok M and Ojalvo LS: Targeting the programmed cell death-1 pathway in breast and ovarian cancer. Curr Opin Obstet Gynecol. 28:142–147. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Hegg R, Im SA, Shaw Wright G, et al: Atezolizumab and Nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 379:2108–2121. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Cortes J, Cescon DW, Rugo HS, Nowecki Z, Im SA, Yusof MM, Gallardo C, Lipatov O, Barrios CH, Holgado E, et al: Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): A randomised, placebo-controlled, double-blind, phase 3 clinical trial. Lancet. 396:1817–1828. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Mittendorf EA, Philips AV, Meric-Bernstam F, Qiao N, Wu Y, Harrington S, Su X, Wang Y, Gonzalez-Angulo AM, Akcakanat A, et al: PD-L1 expression in triple-negative breast cancer. Cancer Immunol Res. 2:361–370. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Schalper KA, Velcheti V, Carvajal D, Wimberly H, Brown J, Pusztai L and Rimm DL: In situ tumor PD-L1 mRNA expression is associated with increased TILs and better outcome in breast carcinomas. Clin Cancer Res. 20:2773–2782. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Baptista MZ, Sarian LO, Derchain SF, Pinto GA and Vassallo J: Prognostic significance of PD-L1 and PD-L2 in breast cancer. Hum Pathol. 47:78–84. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Sabatier R, Finetti P, Mamessier E, Adelaide J, Chaffanet M, Ali HR, Viens P, Caldas C, Birnbaum D and Bertucci F: Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget. 6:5449–5464. 2015. View Article : Google Scholar : PubMed/NCBI

42 

Bae SB, Cho HD, Oh MH, Lee JH, Jang SH, Hong SA, Cho J, Kim SY, Han SW, Lee JE, et al: Expression of programmed death receptor ligand 1 with high tumor-infiltrating lymphocytes is associated with better prognosis in breast cancer. J Breast Cancer. 19:242–251. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Mahmoud SMA, Paish EC, Powe DG, Macmillan RD, Grainge MJ, Lee AHS, Ellis IO and Green AR: Tumor-infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 29:1949–1955. 2011. View Article : Google Scholar : PubMed/NCBI

44 

Muenst S, Schaerli AR, Gao F, Däster S, Trella E, Droeser RA, Muraro MG, Zajac P, Zanetti R, Gillanders WE, et al: Expression of programmed death ligand 1 (PD-L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat. 146:15–24. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Li Z, Dong P, Ren M, Song Y, Qian X, Yang Y, Li S, Zhang X and Liu F: PD-L1 expression is associated with tumor FOXP3(+) regulatory T-cell infiltration of breast cancer and poor prognosis of patient. J Cancer. 7:784–793. 2016. View Article : Google Scholar : PubMed/NCBI

46 

Qin T, Zeng Y, Qin G, Xu F, Lu JB, Fang WF, Xue C, Zhan JH, Zhang XK, Zheng QF, et al: High PD-L1 expression was associated with poor prognosis in 870 Chinese patients with breast cancer. Oncotarget. 6:33972–33981. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Madjd Z, Spendlove I, Pinder SE, Ellis IO and Durrant LG: Total loss of MHC class I is an independent indicator of good prognosis in breast cancer. Int J Cancer. 117:248–255. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Gudmundsdóttir I, Gunnlaugur Jónasson J, Sigurdsson H, Olafsdóttir K, Tryggvadóttir L and Ogmundsdóttir HM: Altered expression of HLA class I antigens in breast cancer: Association with prognosis. Int J Cancer. 89:500–505. 2000. View Article : Google Scholar : PubMed/NCBI

49 

Zitvogel L, Tesniere A and Kroemer G: Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat Rev Immunol. 6:715–727. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Savas P, Salgado R, Denkert C, Sotinou C, Darcy PK, Smyth MJ and Loi S: Clinical relevance of host immunity in breast cancer: From TILs to the clinic. Nat Rev Clin Oncol. 13:228–241. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Davies H, Morganella S, Purdie CA, Jang SJ, Borgen E, Russnes H, Glodzik D, Zou X, Viari A, Richardson AL, et al: Whole-genome sequencing reveals breast cancers with mismatch repair deficiency. Cancer Res. 77:4755–4762. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Denkert C, von Mincwitz G, Darb-Esfahni S, Lederer B, Heppner BI, Weber KE, Budczies J, Huober J, Klauschen F, Furlanetto J, et al: Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: A pooled analysis of 3771 patients treated with neoadjuvant therapy. Lancet Oncol. 19:40–50. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Denkert C, Wienert S, Poterie A, Loibl S, Budczies J, Badve S, Bago-Horvath Z, Bane A, Bedri S, Brock J, et al: Standardized evaluation of tumor-infiltrating lymphocytes in breast cancer: Results of the ring studies of the international immuno-oncology biomarker working group. Mod Pathol. 29:1155–1164. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Salgado R, Denkert C, Demaria S, Sirtaine N, Klauschen F, Pruneri G, Wienert S, Van den Eynden G, Baehner FL, Penault-Llorca F, et al: The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: Recommendations by an international TILs working group 2014. Ann Oncol. 26:259–271. 2015. View Article : Google Scholar : PubMed/NCBI

55 

Loi S, Michiels S, Adams S, Loibl S, Budczies J, Denkert C and Salgado R: The journey of tumor-infiltrating lymphocytes as a biomarker in breast cancer: Clinical utility in an era of checkpoint inhibition. Ann Oncol. 32:1236–1244. 2021. View Article : Google Scholar : PubMed/NCBI

56 

Igarashi T, Teramoto K, Ishida M, Hanaoka J and Daigo Y: Scoring of PD-L1 expression intensity on pulmonary adenocarcinomas and the correlations with clinicopathological factors. ESMO Open. 1:e0000832016. View Article : Google Scholar : PubMed/NCBI

57 

Brookmeyer R and Crowley J: A k-sample median test for censored data. J Am Stat Assoc. 77:433–440. 1982. View Article : Google Scholar

58 

Wen ZF, Liu H, Gao R, Zhou M, Ma J, Zhang Y, Zhao J, Chen Y, Zhang T, Huang F, et al: Tumor cell-released autophagosomes (TRAPs) promote immunosuppression through induction of M2-like macrophages with increased expression of PD-L1. J Immunother Cancer. 6:1512018. View Article : Google Scholar : PubMed/NCBI

59 

Vitale I, Manic G, Coussens LM, Kroemer G and Galluzzi L: Macrophages and metabolism in the tumor microenvironment. Cell Metab. 30:36–50. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Quail DF and Joyce JA: Microenvironmental regulation of tumor progression and metastasis. Nat Med. 19:1423–1437. 2013. View Article : Google Scholar : PubMed/NCBI

61 

Drake CG, Jaffee E and Pardoll DM: Mechanisms of immune evasion by tumors. Adv Immunol. 90:51–81. 2006. View Article : Google Scholar : PubMed/NCBI

62 

Mamessier E, Sylvain A, Thibult ML, Houvenaeghel G, Jacquemier J, Castellano R, Gonçalves A, André P, Romagné F, Thibault G, et al: Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity. J Clin Invest. 121:3609–3622. 2011. View Article : Google Scholar : PubMed/NCBI

63 

Kim R, Emi M and Tanabe K: Cancer immunoediting from immune surveillance to immune escape. Immunology. 121:1–14. 2007. View Article : Google Scholar : PubMed/NCBI

64 

Fang Y, Wang L, Wan C, Sun Y, Van der Jeught K, Zhou Z, Dong T, So KM, Yu T, Li Y, et al: MAL2 drives immune evasion in breast cancer by suppressing tumor antigen presentation. Clin Invest. 131:e1408372021. View Article : Google Scholar

65 

Spranger S and Gajewski TF: Impact of oncogenic pathways on evasion of antitumour immune responses. Nat Rev Cancer. 18:139–147. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Loi S, Sirtaine N, Piette F, Salgado R, Viale G, Van Eenoo F, Rouas G, Francis P, Crown JPA, Hitre E, et al: Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol. 31:860–867. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Zhang N, Sun H, Zhao S, Wang Y, Pu H, Wang Y and Zhang Q: Expression of PD-L1 and prognosis in breast cancer: A meta-analysis. Oncotarget. 8:31347–31354. 2017. View Article : Google Scholar : PubMed/NCBI

68 

Igarashi T, Teramoto K, Ishida M, Hanaoka J and Daigo Y: The mechanism of de novo expression of programmed cell death-ligand 1 in squamous cell carcinoma of the lung. Oncol Rep. 38:2189–2196. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Teramoto K, Igarashi T, Kataoka Y, Ishida M, Hanaoka J, Sumimoto H and Daigo Y: Biphasic prognostic significance of PD-L1 expression status in patients with early- and locally advanced-stage non-small cell lung cancer. Cancer Immunol Immunother. 70:1063–1074. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Smith J, Robida MD, Acosta K, Vennapusa B, Mistry A, Martin G, Yates A and Hnatyszyn HJ: Quantitative and qualitative characterization of two PD-L1 clones: SP263 and E1L3N. Diag Pathol. 11:442016. View Article : Google Scholar

Related Articles

Journal Cover

April-2023
Volume 25 Issue 4

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Murazawa C, Hashimoto N, Kuraishi K, Motoyama M, Hashimoto S, Ikeuchi M, Norimura S, Matsunaga T, Teramoto K, Haba R, Haba R, et al: Status and prognostic value of immunological biomarkers of breast cancer. Oncol Lett 25: 164, 2023
APA
Murazawa, C., Hashimoto, N., Kuraishi, K., Motoyama, M., Hashimoto, S., Ikeuchi, M. ... Kontani, K. (2023). Status and prognostic value of immunological biomarkers of breast cancer. Oncology Letters, 25, 164. https://doi.org/10.3892/ol.2023.13750
MLA
Murazawa, C., Hashimoto, N., Kuraishi, K., Motoyama, M., Hashimoto, S., Ikeuchi, M., Norimura, S., Matsunaga, T., Teramoto, K., Haba, R., Abe, N., Yajima, T., Kontani, K."Status and prognostic value of immunological biomarkers of breast cancer". Oncology Letters 25.4 (2023): 164.
Chicago
Murazawa, C., Hashimoto, N., Kuraishi, K., Motoyama, M., Hashimoto, S., Ikeuchi, M., Norimura, S., Matsunaga, T., Teramoto, K., Haba, R., Abe, N., Yajima, T., Kontani, K."Status and prognostic value of immunological biomarkers of breast cancer". Oncology Letters 25, no. 4 (2023): 164. https://doi.org/10.3892/ol.2023.13750