Observational Study Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Exp Med. Sep 20, 2025; 15(3): 108984
Published online Sep 20, 2025. doi: 10.5493/wjem.v15.i3.108984
Concordance of programmed death-ligand 1 expression assessments determined via two immunohistochemical tests and the polymerase chain reaction method
Marina A Senchukova, Dmitry G Tagabilev, Scientific and Clinical Center No. 3, Petrovsky National Research Centre of Surgery, Moscow 108840, Troitsk, Russia
Natalia V Saidler, Department of Pathology, Orenburg Regional Cancer Clinic, Orenburg 460021, Orenburgskaya Oblast’, Russia
Evgeniya Yu Zubareva, Department of Oncology, Orenburg State Medical University, Orenburg 460021, Orenburgskaya Oblast’, Russia
Alexander B Prokofiev, Molecular Genetics Laboratory, Orenburg State Medical University, Orenburg 460000, Orenburgskaya Oblast’, Russia
ORCID number: Marina A Senchukova (0000-0001-8371-740X); Natalia V Saidler (0009-0007-3641-1580); Evgeniya Yu Zubareva (0000-0001-7025-0206); Alexander B Prokofiev (0009-0008-5236-8065); Dmitry G Tagabilev (0000-0003-2823-635X).
Author contributions: Senchukova MA formulated the idea and aims of the study, wrote the first version of the manuscript and performed the analysis of the results and statistical processing of the results; Saidler NV made a significant contribution to the development of the study methodology, participated in the discussion of the obtained results, and revised and approved the final version; Zubareva EYu collected and analyzed the data and made a significant contribution to the concept and design of the study, participated in the preparation of tables and figures; Prokofiev AB made a significant contribution to the development of the methodology of the molecular genetic study and participated in the discussion of the obtained results; Tagabilev DG made a significant contribution to the selection and analysis of data and their discussion, as well as to the interpretation of the obtained results; All the authors wrote and approved the final version of the manuscript.
Supported by Russian Science Foundation, No. 23-25-00183.
Institutional review board statement: This study was reviewed and approved by Ethics Committee of Orenburg State Medical University (Russia, Orenburg).
Informed consent statement: This article presents data from a clinical study involving 148 patients with breast cancer. The patient has signed the informed consent form.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
STROBE statement: The authors have read the STROBE Statement—checklist of items, and the manuscript was prepared and revised according to the STROBE Statement—checklist of items.
Data sharing statement: Data from patients included in the study in Statistica10 table or Excel table formats can be provided upon request to the corresponding author at masenchukova@yandex.com.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Marina A Senchukova, MD, PhD, Professor, Scientific and Clinical Center No. 3, Petrovsky National Research Centre of Surgery, Oktyabrsky Prospekt 3, Moscow 108840, Troitsk, Russia. masenchukova@yandex.com
Received: April 27, 2025
Revised: May 21, 2025
Accepted: July 29, 2025
Published online: September 20, 2025
Processing time: 107 Days and 16.1 Hours

Abstract
BACKGROUND

We previously demonstrated that the antibody against programmed cell death protein 1 ligand 1 (PDCD1 LG1) is a promising new marker of programmed death-ligand 1 (PD-L1) expression that correlates with both breast cancer (BC) clinicopathological characteristics and tumor sensitivity to chemotherapy. However, the concordance of PDCD1 LG1 expression scoring with immunohistochemical (IHC) tests approved for clinical use and with the polymerase chain reaction (PCR) method has not been previously studied.

AIM

To evaluate the concordance of methods for assessing PD-L1 expression, IHC tests with anti-PD-L1 (PDCD1 LG1) and anti-PD-L1 (SP142) antibodies and PCR.

METHODS

This prospective single-center observational cohort study included 148 patients with BC. PD-L1 expression in immune cells was assessed by the IHC method with anti-PD-L1 (PDCD1 LG1) and anti-PD-L1 (SP142) antibodies and by PCR. The concordance of PD-L1 scores between tests was assessed with positive percentage agreement (PPA) and negative percentage agreement (NPA). The strength of the agreement between the methods was calculated via the Cohen kappa index. P < 0.05 was considered statistically significant.

RESULTS

Regardless of the method used to assess marker expression, PD-L1 expression was significantly more often detected in patients with negative estrogen receptor status, human epidermal growth factor receptor-2-positive (HER2+) status, luminal B HER+ BC, nonluminal HER+ BC and triple-negative BC. PPA and NPA were 38.3% and 70.4%, respectively, for PD-L1 (PDCD1 LG1) and PD-L1 (SP142); 26.3% and 63.3%, respectively, for PD-L1 (PDCD1 LG1) and PD-L1 (PCR); and 36.5% and 74.4%, respectively, for PD-L1 (SP142) and PD-L1 (PCR). Cohen's kappa index for PD-L1 (PDCD1 LG1) and PD-L1 (SP142) was 0.385 (95%CI: 0.304–0.466), that for PD-L1 (PDCD1 LG1) and PD-L1 (PCR) was 0.207 (95%CI: 0.127–0.287), and that for PD-L1 (SP142) and PD-L1 (PCR) was 0.389 (95%CI: 0.309–0.469).

CONCLUSION

Thus, all three markers of PD-L1 expression are associated with the characteristics of aggressive BC, demonstrating moderate concordance between the tests.

Key Words: Breast cancer; Cohen kappa index; Negative percentage agreement; Positive percentage agreement; Programmed death-ligand 1; Programmed cell death protein 1 ligand 1

Core Tip: We determined the expression of programmed cell death ligand 1 in immune cells using immunohistochemistry with antibodies against programmed cell death protein 1 ligand 1 (PDCD1 LG1) and SP142 and polymerase chain reaction (PCR) and then examined the agreement between the results of three assays in patients with breast cancer (BC). All three tests were positively correlated with estrogen receptor-negative status, human epidermal growth factor receptor-2-positive (HER2+) status, and luminal B HER+, nonluminal HER+, and triple-negative BCs. Cohen's kappa index was 0.385 for PDCD1 LG1 and SP142 and 0.389 for SP142 and programmed death-ligand 1 (PCR), which is considered moderate agreement between the markers.



INTRODUCTION

Breast cancer (BC) remains a major medical, social and economic problem and is the leading cause of cancer mortality among the female population[1]. Analysis shows that in the coming decades, the burden of BC worldwide will only increase[2]. A feature of BC is the diversity of clinical, morphological and molecular genetic forms that require different approaches for diagnosis and treatment. Considering that BC is a systemic disease, the effectiveness of drug therapy largely determines the long-term results of treatment for this pathology. In this context, the search for new promising prognostic and predictive markers of BC has not lost its relevance.

One of the promising markers associated with the prognosis and sensitivity of BC to chemotherapy and immunotherapy is programmed death-ligand 1 (PD-L1)[3]. PD-L1 is a transmembrane protein expressed by tumor cells (TCs) and immune cells (ICs) and is involved in tumor evasion of immune surveillance[4,5]. The binding of PD-L1 to programmed cell death protein 1 (PD-1), which is located on the surface of activated T lymphocytes, natural killers, B lymphocytes, macrophages, dendritic cells and monocytes, leads to a decrease in the proliferation of PD-1-positive cells and the suppression of cytokine secretion, resulting in the inhibition of T-cell activation and eventually the induction of TC immune escape[4,5].

Currently, several markers, including anti-PD-L1 (SP142), anti-PD-L1 (SP263), and anti-PD-L1 (22C3) antibodies, have been approved for assessing PD-L1 expression in clinical practice. However, clinical trial data indicate low consistency in the results of PD-L1 expression assessment when different markers are used and inconsistent assessments of their prognostic and predictive significance[3,6,7]. In addition, in clinical practice, immune checkpoint (ICP) inhibitors that block the PD-L1/PD-1 pathway have unfortunately failed to meet expectations[8]. This is because PD-L1 expression in TCs and ICs is regulated by various mechanisms, including epigenetic regulation, the activation of various oncogenic pathways and transcription factors, and posttranscriptional regulation[9]. In addition, PD-L1 is present not only on the membranes of TCs but also in the cytoplasm, exosomes and nuclei of TCs, and the functions of PD-L1 are much broader than those of tumor evasion from the immune response[10-12]. On the basis of these data, it can be assumed that different types of antibodies can react with different PD-L1 “epitopes”, which are localized in different cellular components. The study of the intracellular distribution of the marker and its functions is still at the very beginning of the "path" and conceals many interesting "discoveries".

We previously demonstrated that the antibody against programmed cell death protein 1 Ligand 1 (PDCD1 LG1) is a promising new marker of PD-L1 expression that correlates with both BC clinicopathological characteristics and tumor sensitivity to chemotherapy[13-15]. However, the concordance of PD-L1 expression estimates between anti-PD-L1 (PDCD1 LG1) and markers approved for clinical use has not been studied. Therefore, the objectives of this study are (1) To determine the frequency of PD-L1 expression in ICs by immunohistochemical (IHC) using anti-PD-L1 (PDCD1 LG1) and anti-PD-L1 (SP142) antibodies and in tumor tissue by polymerase chain reaction (PCR); (2) To evaluate the frequency of PD-L1 (PDCD1 LG1), PD-L1 (SP142) and PD-L1 (PCR) expression in accordance with BC clinical and pathological characteristics; and (3) To establish the concordance of the results of PD-L1 expression assessment between these markers.

MATERIALS AND METHODS
Patient characteristics

This prospective single-center observational cohort study included 148 patients who underwent radical surgery for BC between January 15, 2023, and June 30, 2023, at the Orenburg Regional Clinical Oncology Dispensary (Orenburg, Russia). The age of the patients was 62.8 ± 26.0 years (median: 62.0 years). The study was performed in accordance with the 1964 Helsinki Declaration and internationally recognized guidelines. The study protocol was approved by Ethics Committee of Orenburg State Medical University. All patients signed an informed consent form to participate in this study. The inclusion and exclusion criteria for patients included in the study are presented in Table 1.

Table 1 Inclusion and exclusion criteria for patients in the study.
Inclusion criteria
Exclusion criteria
Stage breast cancer Tis-2N0-1M0Chemotherapy, targeted, hormone therapy or radiation therapy before surgery
Radical surgical treatment (R0) at the first stageTaking corticosteroids or nonsteroidal anti-inflammatory drugs
Axillary lymph node dissection at levels 1-2Refusal to participate in the study
Voluntary informed consent to participate in a clinical trial

The BC stage was determined according to the 8th TNM classification of malignant tumors. Data on patient age at surgery, T and N stages, tumor grade (G), estrogen receptor (ER) status, progesterone receptor (PR) status, human epidermal growth factor receptor 2 (HER2) status, the Ki67 index, the molecular biological subtype of BC, the presence of lymphovascular invasion (LVI), perineural invasion (PNI) and the intraductal component (IDC) were obtained from outpatient records and pathologists' reports. The distribution of patients included in the study according to clinicopathological characteristics and the molecular biological subtypes of BC is presented in Figure 1.

Figure 1
Figure 1 Distribution of patients with breast cancer. A: According to T and N stages and tumor grade; B: According to their molecular and biological characteristics. Abbreviations: ER: Estrogen receptor; HER2: Human epidermal growth factor receptor-2; PR: Progesterone receptor; TNBC: Triple negative breast cancer.

According to the presented data, the studied group predominantly included patients with T1c-T2 stages of BC (89.8%), N0-N1 stages (86.5%), tumor malignancy grades G2 and G3 (89.2%), ER-positive (ER+) status (89.2%), PR-positive (PR+) status (72.3%), HER2-negative (HER2-) tumor status (85.5%) and luminal A and luminal B HER2-BC (82.2%). LVI and PNI were detected in 56.8% and 77.7% of the tumor samples, respectively, and the IDC was noted in 41 (27.7%) BC samples. In 20 BC patients, the disease stage changed after surgery, as metastases were detected in 4 or more axillary lymph nodes.

Immunohistochemistry

IHC staining for PD-L1 was performed on 4-μm-thick sections from formalin-fixed paraffin-embedded tissue blocks according to the manufacturer’s instructions. The blocks were previously subjected to deparaffinization, dehydration with xylene, and rehydration in a graded series of alcohol solutions. Staining was performed on fully automated staining systems: (1) A BOND-MAX staining system (Leica Biosystems Melboume Pty Ltd., Mount Waverley, Australia) for antibodies against PDCD1 LG1 (1:100 dilution, Cloud-Clone Corp., Shanghai, China); and (2) A BenchMark XT IHC/ISH (Ventana Medical Systems) for antibodies against SP142 (ready to use dilution, Ventana Medical System, Oro Valley, AZ, United States). The visualization system included diaminobenzidine with hematoxylin. For the negative control sections, the primary antibodies were replaced with phosphate-buffered saline, and the samples were processed in the same way. Benign tonsil tissue was used as a positive (marker expression in lymphocytes and macrophages in the germinative centers and reticular epithelium of the crypts) and negative (no marker expression in the interfollicular areas and squamous epithelium) control.

PDCD1 LG1+ IC and SP142+ IC scoring

The expression of tumor-infiltrating ICs (lymphocytes, macrophages, dendritic cells, and granulocytes) was determined in 5 fields of view at 200 × magnification and calculated as the proportion of the tumor area (nonnecrotic and nonsclerotic areas) occupied by PDCD1 LG1-positive (PDCD1 LG1+) and SP142-positive (SP142+) ICs of any intensity. We used two assessment intervals for PDCD1 LG1+ ICs (< 10% and ≥ 10%) and two assessment intervals for SP142+ ICs (< 1% and ≥ 1%). Examples of the quantification of PDCD1 LG1+ IC and SP142+ IC scores are shown in Figure 2.

Figure 2
Figure 2 Assessment of tumor-infiltrating programmed cell death protein 1 ligand 1-positive and SP142+ immune cells. A: Programmed cell death protein 1 ligand 1-positive (PDCD1 LG1+) immune cell (IC) score < 10%; B: PDCD1 LG1+ IC score ≥ 10%; C: SP142 IC score < 1%; D: SP142+ IC score ≥ 1%. Staining with anti-PDCD1 LG1 antibodies (A and B), 200 ×; staining with anti-programmed death-ligand 1 antibodies (clone SP142)(C and D), 200 ×.

All samples were examined by two researchers (Senchukova MA and Saidler NV) who were blinded to the clinical and pathological data of the patients. Histological preparations were examined via light microscopy (Levenhuk D740T digital microscope connected to a 5.1 MP camera, Levenhuk, Russia).

Determination of PD-L1 expression by PCR

Tumor tissue for PCR analysis was also collected within 30–40 minutes after removal of the surgical sample, placed in Eppendorf tubes (2 mL), and stored at -40 °C until analysis. Total cellular RNA was isolated via RNA-Extran reagent (Synthol, Russia) according to the manufacturer's instructions. Next, cDNA was synthesized via an OT-1 reverse transcription kit (Synthol, Russia) containing a buffer with oligonucleotides, the MMLV-RT enzyme, and the Random-6 and Oligo-15 primers. The reverse transcription conditions were as follows: (1) 37 °C for 30 minutes; and (2) 92 °C for 5 minutes. Real-time PCR was performed on a DTLite device (DNA-Technology, Russia) with the obtained cDNA and a 2.5 × reaction mixture for RT-PCR in the presence of EVA Green dye. The RT-PCR conditions were as follows: (1) 95 °C for 5 minutes; (2) 95 °C for 15 seconds; and (3) 62 °C for 45 seconds for 55 cycles. To detect a specific product (gene expression), the following primer sequence was used: (1) Forward, 5’-GGCATTTGCTGAACGCAT-3’; and (2) Reverse, 5’- CAATTAGTGCAGCCAGGT-3’.

Statistical analysis

Statistical processing of the results was performed with Statistica 12.0 software. Correlations between different indicators were estimated via nonparametric Spearman's rank correlation. χ² tests were performed to analyze differences in distribution between categorized data. To plot the receiver operating characteristic (ROC) curve and measure the area under the curve (AUC), software, which is available free, was used. The best threshold (cutoff) values were determined by the largest Youden index (J = sensitivity + specificity - 1). The effectiveness of the predictive model was assessed by the AUC. The concordance of PD-L1 scores between tests was assessed with positive percentage agreement (PPA) and negative percentage agreement (NPA). The strength of the agreement between the methods was calculated with the Cohen kappa index. P < 0.05 was considered statistically significant.

RESULTS
Specificities of PD-L1 (PDCD1 LG1) and PD-L1 (SP142) expression in benign tonsil tissue

When tonsil tissue was stained with anti-PD-L1 (PDCD1 LG1) antibodies, diffuse cytoplasmic expression of the marker was observed in most ICs. The expression of the marker in the squamous epithelium was absent. However, in the germinative centers and reticular epithelium of the crypts, cells with pronounced hyperexpression of the marker were observed. The number and localization of these cells corresponded to the number and localization of cells expressing PD-L1 (SP142) in tonsil tissue. It can be assumed that cells with PD-L1 (PDCD1 LG1) hyperexpression and cells expressing PD-L1 (SP142) have similar characteristics, but their histopathology requires clarification. Figure 3 shows the results of staining tonsil tissue with anti-PD-L1 (PDCD1 LG1) and anti-PD-L1 (SP142) antibodies.

Figure 3
Figure 3 Programmed death-ligand 1 expression in benign tonsil tissue. A: Staining with anti-programmed cell death protein 1 Ligand 1 antibody, 200 ×; B: Staining with anti-programmed death-ligand 1 (clone SP142) antibody, 200 ×.
ROC analysis results

ROC analysis was performed to determine the optimal cutoff point for the PD-L1 (PDCD1 LG1) score corresponding to a negative PD-L1 (SP142) status (< 1%). We used four assessment intervals for PD-L1 (PDCD1 LG1) scores in ICs: (1) < 1%; (2) ≥ 1% and < 10%; (3) ≥ 10% and < 30%; and (4) ≥ 30%. According to the obtained data, a PD-L1 (PDCD1 LG1) IC score < 10% (cutoff) corresponded to a negative status of PD-L1 expression in ICs assessed with an anti-PD-L1 (SP142) antibody. The AUC was 0.743 (95%CI: 0.648–0.843, P < 0.0001). The sensitivity and specificity of the method were 63.9% and 79.3%, respectively. Figure 4 shows the ROC curve discriminating PD-L1 (PDCD1 LG1) scores corresponding to positive and negative PD-L1 (SP142) statuses (< 1% and ≥ 1%).

Figure 4
Figure 4 Receiver operating characteristic curve discriminating programmed cell death protein 1 Ligand 1 scores corresponding to positive and negative programmed death-ligand 1 (clone SP142) status (< 1% and ≥ 1%). AUC: Area under the curve.
Frequency of PD-L1 expression, depending on the method of marker determination and the clinical and pathological characteristics of BC

Data on the frequency of PD-L1 expression, depending on the method of marker determination and the clinical and pathological characteristics of BC patients, are presented in Table 2.

Table 2 Frequency of programmed death-ligand 1 expression depending on marker determination methods and the clinical and pathological characteristics of breast cancer patients.
BC characteristicsProgrammed cell death protein 1 Ligand 1 score
SP142 score
Polymerase chain reaction test
P value or χ² test
< 10%
≥ 10%
< 1%
≥ 1%
Negative
Positive
n
%
n
%
n
%
n
%
n
%
n
%
T stage
Тin situ250.0250.04100004100000.0211, 0.262, 0.0053
Т1в436.4763.6654.6545.4436.4763.6
Т1с5966.33033.76876.42123.67685.41314.6
Т23681.8818.23477.31022.72965.91534.1
N stage
N05871.62328.46074.12125.96175.32024.70.701, 0.432, 0.733
N13166.01634.03676.61123.43676.61123.4
N2-31260.0840.01680.0420.01680.0420.0
Tumor grade
G11062.5637.51487.5212.51275.0425.00.251, 0.000152, 0.823
G26473.62326.47485.11314.96878.21921.8
G32760.01840.02453.32146.73373.31226.7
Lymphovascular invasion
Absent6071.42428.66678.61821.46476.22023.80.341, 0.352, 0.963
Present4164.12335.94671.91828.14976.61523.4
Perineural invasion
Absent7968.73631.38473.03127.08473.03127.00.821, 0.162, 0.083
Present2266.71133.32884.9515.12987.9412.1
Intraductal component
Absent7368.23431.87872.92927.18579.42220.60.991, 0.202, 0.153
Present2868.31331.73482.9717.12868.31331.7
Estrogen receptor status
Negative531.31168.700161001062.5637.50.00081, 0.00002, 0.173
Positive9672.73627.311284.92015.110378.02922.0
Progesterone receptor status
Negative2561.01639.01639.02561.02765.81434.20.241, 0.00002, 0.0633
Positive7671.03129.09689.71110.38680.42119.6
HER2 status
Negative9977.32922.711287.51612.510380.52519.50.00001, 0.00002, 0.0033
Positive210.01890.000201001050.01050.0
BC subtypes
Luminal A4275.01425.05089.3610.75089.3610.70.00001, 0.00002, 0.00053
Luminal B HER2-5481.81218.26293.946.15075.81624.2
Luminal B HER2+00101000010100330.0770.0
Nonluminal HER2+220.0880.00010100770.0330.0
Triple negative BC350.0350.0006100350.0350.0

According to the data presented, regardless of the method of marker determination used, PD-L1 expression in ICs was significantly more often detected in patients with negative ER status, HER2+ status, luminal B HER+ BC, nonluminal HER+ BC and triple negative BC (TNBC). Interestingly, marker expression was also detected more often in the T1b stage of invasive BC than in the T1c and T2 stages. In ductal carcinoma in situ, PD-L1 (PDCD1 LG1) expression was positive in two of the four BC samples, and PD-L1 (SP142) and PD-L1 (PCR) expression was negative in all samples. However, the small number of patients in the Tin situ (4 patients) and T1b (11 patients) groups does not allow us to consider these conclusions sufficiently reliable. The frequency of PD-L1 (SP142) expression was greater in patients with a tumor grade 3 and negative PR status, but the frequency of PD-L1 (PDCD1 LG1) and PD-L1 (PCR) expression did not depend on these BC characteristics. The Ki67 indices for negative and positive PD-L1 statuses were 23.6% ± 13.0% and 51.7% ± 29.0%, respectively, for PD-L1 (SP142) (P < 0.00001); 25.1% ± 14.8% and 41.9% ± 29.1χ%, respectively, for PD-L1 (PDCD1 LG1) (P = 0.002); and 25.1% ± 15.2% and 47.7% ± 29.9%, respectively, for PD-L1 (PCR) (P = 0.00005). No differences were found in the frequency of PD-L1 expression of all three markers depending on N stage, the presence of LVI and PNI, or the IDC.

Concordance between the results of PD-L1 expression assessment using immunohistochemistry and PCR

The expression of PD-L1 (PDCD1 LG1), PD-L1 (SP142) and PD-L1 (PCR) was detected in 47 (31.8%), 35 (23.6%) and 36 (24.3%) BC samples, respectively. Figure 5 demonstrate the frequency of expression of the markers and their combinations.

Figure 5
Figure 5 Frequencies of programmed death-ligand 1 expression in immune cells according to marker determination methods. A: Frequencies of programmed death-ligand 1 (PD-L1) expression for all three markers and their combinations; B: Frequencies of programmed cell death protein 1 Ligand 1 (PDCD1 LG1) and PD-L1 (clone SP142) expression and their combination; C: Frequencies of PDCD1 LG1 and PD-L1 [polymerase chain reaction (PCR)] expression and their combination; D: Frequencies of PD-L1 (clone SP142) and PD-L1 (PCR) expression and their combination. PCR: Polymerase chain reaction; PDCD1 LG1: Programmed cell death protein 1 Ligand 1; PD-L1: Programmed death-ligand 1.

The Spearman rank order correlation between PD-L1 (PDCD1 LG1) and PD-L1 (SP142) was 0.391 (P < 0.0001), that between PD-L1 (PDCD1 LG1) and PD-L1 (PCR) was 0.201 (P = 0.014), and that between PD-L1 (SP142) and PD-L1 (PCR) was 0.389 (P < 0.0001). PPA and NPA for PD-L1 (PDCD1 LG1) and PD-L1 (SP142), PD-L1 (PDCD1 LG1) and PD-L1 (PCR) and PD-L1 (SP142) and PD-L1 (PCR) were 18.1% and 56.3%, 38.3% and 70.4%, 26.3% and 63.3%, and 36.5% and 74.4%, respectively. Cohen's kappa index for PD-L1 (PDCD1 LG1) and PD-L1 (SP142) was 0.385 (CI: 0.304–0.466); for PD-L1 (PDCD1 LG1) and PD-L1 (PCR), it was 0.207 (95%CI: 0.127–0.287); and for PD-L1 (SP142) and PD-L1 (PCR), it was 0.389 (95%CI: 0.309–0.469).

DISCUSSION

The effectiveness of BC treatment depends on the accuracy of the disease prognosis and the effectiveness of the therapy. Ineffective treatment involves not only the absence of a result but also the negative consequences for the health of patients. BC is characterized by a variety of clinical, morphological and molecular genetic forms that differ not only in the prognosis of the disease but also in sensitivity to drugs and radiotherapy. In this context, the search for new prognostic and predictive markers of BC has not lost its relevance.

The immune system plays a decisive role in the occurrence and progression of malignant neoplasms. In addition, the severity and cellular composition of the immune infiltrate of the tumor stroma significantly affect the effectiveness of antitumor radiation and drug therapy and, consequently, the prognosis of the disease[16-18]. However, the cellular composition of tumor-associated ICs is very diverse[18,19]. Some ICs are involved in the activation of tumor progression factors, whereas others, on the contrary, have antitumor activity[20-22]. A special place in the IC population is occupied by cells expressing PD-L1. PD-L1 expression on TCs is associated with immune escape mechanisms, but the role of PD-L1 expression in ICs is not clear. For example, a high density of PD-L1-positive ICs in the tumor stroma is associated with more aggressive BC characteristics, including negative ER and PR statuses, HER+ BC, TNBC and a high Ki67 index[23,24]. However, high infiltration of the tumor stroma with ICs, including PD-L1+ ICs[22,25,26], and a high density of PD-L1+ intratumoral macrophages[27] are associated with a better prognosis of the disease and response to chemotherapy and chemoimmunotherapy in patients with TNBC.

The discovery of ICP inhibitors has given hope for significant breakthroughs in the treatment of the most aggressive malignancies, including TNBC. Unfortunately, the results of the use of ICP inhibitors in BC patients have not been convincing enough, and the advisability of their use in this disease continues to be debated[8,28]. The lack of effect on treatment with ICP inhibitors may be due to insufficient understanding of the role of PD-L1 in tumor progression and the lack of reliable markers that allow stratification of patients into groups for whom therapy with ICP inhibitors is indicated and those who will not benefit from this treatment. Accordingly, the search for new reliable markers of PD-L1 expression has not lost its relevance.

Previously, we examined the expression of PD-L1 (PDCD1 LG1), a new marker of PD-L1 expression, and showed that its expression in TCs and ICs is associated with BC clinical and pathological characteristics and tumor sensitivity to chemotherapy[13,14]. Unlike antibodies approved for clinical use, including SP142, 22C3, SP263, 22C3, 28-8 and other antibodies, which are characterized by membrane expression of the marker in TCs, PD-L1 (PDCD1 LG1) is expressed in the cytoplasm of TCs. We explain the differences in the expression of PD-L1 markers by the fact that different antibodies can visualize different PD-L1 epitopes localized in different cellular structures. This assumption is supported by data from recent studies indicating that PD-L1 can be localized not only on cell membranes but also in the cytoplasm, exosomes and even cell nuclei and that the functions of PD-L1 Localized in different parts of the cell can differ significantly[10,11].

Despite the potential prognostic and predictive value of this new marker, the comparability of PD-L1 (PDCD1 LG1) expression estimates and markers approved for clinical use has not been practically studied. Therefore, we compared the results of PD-L1 (PDCD1 LG1), PD-L1 (SP142) and PD-L1 (PCR) expression assessments. Given that antibodies against PD-L1 (PDCD1 LG1) stain the cytoplasm of TCs and that antibodies against PD-L1 (SP142) stain the membrane of TCs, comparisons of the expression of markers in TCs would be incorrect. For this reason, we did not compare the results of PD-L1 expression assessment in TCs and focused on ICs.

We first compared the results of PD-L1 (PDCD1 LG1) and PD-L1 (SP142) expression in benign tonsil tissue, which was used as a positive control. We found that the PDCD1 LG1 antibody diffusely stains the cytoplasm of most ICs. However, in the background of diffuse staining, in the germinal centers and crypt reticular epithelium, we found that cells overexpressing PD-L1 (PDCD1 LG1) were similar to cells expressing PD-L1 (SP142) in the tonsils. We hypothesize that cells overexpressing PD-L1 (PDCD1 LG1) and cells expressing PD-L1 (SP142) in the tonsils may have similar characteristics and perform the same functions. However, this hypothesis needs to be verified with double IHC staining to exclude or confirm the coexpression of markers.

We subsequently performed ROC analysis and determined that PD-L1 (PDCD1 LG1) scores in ICs < 10% and ≥ 10% corresponded to PD-L1 (SP142) negative (< 1%) and positive (≥ 1%) statuses in ICs. We compared the frequency of PD-L1 (PDCD1 LG1), PD-L1 (SP142) and PD-L1 (PCR) expression with the clinicopathological characteristics of BC and found that the PD-L1-positive status for all three markers was significantly more often observed in the negative ER status, HER2+ status, and in Luminal B HER+ BC, nonluminal HER+ BC and TNBC. In addition, the Ki67 index was significantly greater in patients with positive PD-L1 status for all three markers. These results are consistent with the results of most researchers who have used other antibodies to assess PD-L1 expression. Thus, in the study of Ni et al[23], a positive status of PD-L1 (SP142) expression was associated with negative ER and PR statuses and HER2+ BC and positively correlated with the Ki67 index. PD-L1 (SP142) expression was also associated with better disease-free survival and BC-specific survival[23]. In another study, the Ki67 index was positively correlated with PD-L1 expression, but this dependence was noted only for clones 28-8 and E1 L3N but not for SP142. In multivariate analysis, among the three markers (SP142, 28-28 and E1 L3N), only negative PD-L1 status in the ICs for clones 28-8 was associated with decreased overall survival[29]. Cha et al[3] reported that negative PD-L1 (22C3) status was significantly associated with decreased recurrence-free survival and distant metastasis-free survival in patients with BC. However, PD-L1 (SP142) expression and PD-L1 (SP263) expression were not associated with disease prognosis.

In the study by Yazaki et al[30], the PD-L1 status in ICs was examined using SP142, SP263, and 22C3 antibodies. A positive PD-L1 status in early BC was significantly more often observed in patients with a histological grade 3 and HER2+ BC. However, this dependence was observed only with a high density of tumor-infiltrating lymphocytes (TILs). With a low TIL density, no differences were observed between the patient groups. Notably, in this study, a positive PD-L1 status in ICs was associated with decreased survival in patients with early BC[30].

Some researchers noted an association between positive PD-L1 (SP142) status in ICs and the absence of metastases in the lymph nodes but did not find an association with tumor grade[6]. In our study, only the PD-L1 (SP142)-positive status was correlated with high-grade tumors. The correlations of the studied markers with the risk of regional metastasis have not been established. However, our study predominantly involved patients with early-stage BC, which, of course, must be taken into account when the results are interpreted.

Interestingly, in some studies, the prognostic significance of PD-L1 expression in BC was assessed via a combined score[31,32]. We believe that this approach is not optimal because PD-L1 expression in TCs is more often associated with decreased patient survival[33-35], whereas PD-L1 expression in ICs is associated with better survival of BC patients[23,36,37].

An important stage of our study was the assessment of the consistency of the results of determining PD-L1 expression in ICs with antibodies against PD-L1 (PDCD1 LG1) and PD-L1 (SP142) and in tumor tissue by PCR. Although all three markers correlated with each other, the consistency of the PD-L1 expression estimates between the markers was quite low. The highest consistency in the assessment of markers was noted between PD-L1 (PDCD1 LG1) and PD-L1 (SP142) and between PD-L1 (SP142) and PD-L1 (PCR). The NPA and PPA were 70.4% and 38.3% for PD-L1 (PDCD1 LG1) and PD-L1 (SP142) and 74.4% and 36.5% for PD-L1 (SP142) and PD-L1 (PCR), respectively. Cohen's kappa index was 0.385 (95%CI: 0.304–0.466) for PD-L1 (PDCD1 LG1) and PD-L1 (SP142) and 0.389 (95%CI: 0.309–0.469) for PD-L1 (SP142) and PD-L1 (PCR), which is considered moderate agreement between these markers.

Our results are consistent with other studies showing low agreement in PD-L1 expression assessments using different antibody clones[38,39]. Sun et al[29] examined PD-L1 expression in patients with TNBC using three antibodies (E1 L3N, 28-8, and SP142). The authors used four cutoff values for TCs (1%, 5%, 10%, and 50%) and three cutoff values for marker expression in ICs [negative; low-positive, stained ICs ≤ 30/high power field (HPF); and high-positive, stained ICs > 30/HPF]. The kappa values were highest for clones 28-8 and E1 L3N: For TCs, the kappa values were 0.660 and 0.997 for the 10% cutoff and 50% cutoff, respectively; for ICs, the kappa value was 0.607. For clones 28-8 and SP142 and for SP142 and E1 L3N, the kappa values were significantly lower, especially for ICs. For antibodies 28-8 and SP142, the kappa value was 0.309, and for antibodies SP142 and E1 L3N, the kappa value was 0.305[29].

A study of the concordance of PD-L1 expression assessments in ICs between the SP142, 22C3 and SP263 assays revealed that the concordance between the SP142 and SP263 assays was better than that between the SP142 and 22C assays. The kappa value for the SP142 and SP263 assays was 0.831, and for the SP142 and 22C3 assays, it was 0.114[40]. Other studies have reported low concordance of PD-L1 expression estimates between SP142, 22C3 and SP263[6,7,24]; between SP263 and SP142[41]; and between SP142 and 22C3[42].

CONCLUSION

Thus, the study of PD-L1 expression in ICs by IHC with antibodies against PD-L1 (PDCD1 LG1) and PD-L1 (SP142) and in tumor tissue by PCR revealed that, regardless of the method used for marker determination, PD-L1 expression is significantly more often detected in patients with an ER-negative status, HER2+ status, and in Luminal B HER+ BC, nonluminal HER+ BC and TNBC. The highest agreement in the assessment of PD-L1 expression was noted between PD-L1 (PDCD1 LG1) and PD-L1 (SP142) and between PD-L1 (SP142) and PD-L1 (PCR), and taking into account Cohen's kappa index, the agreement between these markers was assessed as moderate. The differences in the frequency of PD-L1 expression between markers may be because different antibodies and methods can visualize and detect different PD-L1 epitopes localized in different cell structures. The features of the intracellular distribution of PD-L1 and its role in tumor progression are of undoubted interest for future research. This study has limitations related to the small sample size, single-center nature of the study, predominance of patients with early BC and luminal BC subtypes, and insufficient number of patients with TNBC and HER2+ BC. We believe that further study of the features of PD-L1 (PDCD1 LG1) expression in TCs and ICs and the relationships of this marker with BC clinicopathological characteristics and disease prognosis will contribute to a better understanding of the role of PD-L1 in tumor progression. Studies performed in different scientific and clinical centers and on larger cohorts of patients will help improve the accuracy and reliability of the method and justify the possibility of its use in clinical practice.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Medicine, research and experimental

Country of origin: Russia

Peer-review report’s classification

Scientific Quality: Grade A

Novelty: Grade A

Creativity or Innovation: Grade A

Scientific Significance: Grade A

P-Reviewer: Kudo C, MD, Japan S-Editor: Luo ML L-Editor: A P-Editor: Xu ZH

References
1.  Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, Jemal A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74:229-263.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5690]  [Cited by in RCA: 8001]  [Article Influence: 8001.0]  [Reference Citation Analysis (2)]
2.  Zhang S, Jin Z, Bao L, Shu P. The global burden of breast cancer in women from 1990 to 2030: assessment and projection based on the global burden of disease study 2019. Front Oncol. 2024;14:1364397.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
3.  Cha YJ, Kim D, Bae SJ, Ahn SG, Jeong J, Lee HS, Jeon S, Yoo TK, Park WC, Yoon CI. PD-L1 expression evaluated by 22C3 antibody is a better prognostic marker than SP142/SP263 antibodies in breast cancer patients after resection. Sci Rep. 2021;11:19555.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
4.  Liu Z, Yu X, Xu L, Li Y, Zeng C. Current insight into the regulation of PD-L1 in cancer. Exp Hematol Oncol. 2022;11:44.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 57]  [Reference Citation Analysis (0)]
5.  Syamsu SA, Faruk M, Smaradania N, Sampepajung E, Pranoto AS, Irsandy F, Tammasse IFU. PD-1/PD-L1 pathway: Current research in breast cancer. Breast Dis. 2024;43:79-92.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 7]  [Reference Citation Analysis (0)]
6.  Pang JB, Castles B, Byrne DJ, Button P, Hendry S, Lakhani SR, Sivasubramaniam V, Cooper WA, Armes J, Millar EKA, Raymond W, Roberts-Thomson S, Kumar B, Burr M, Selinger C, Harvey K, Chan C, Beith J, Clouston D, O'Toole SA, Fox SB; kConFab. SP142 PD-L1 Scoring Shows High Interobserver and Intraobserver Agreement in Triple-negative Breast Carcinoma But Overall Low Percentage Agreement With Other PD-L1 Clones SP263 and 22C3. Am J Surg Pathol. 2021;45:1108-1117.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 25]  [Cited by in RCA: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
7.  Rugo HS, Loi S, Adams S, Schmid P, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Winer EP, Kockx MM, Peeters D, Chui SY, Lin JC, Nguyen-Duc A, Viale G, Molinero L, Emens LA. PD-L1 Immunohistochemistry Assay Comparison in Atezolizumab Plus nab-Paclitaxel-Treated Advanced Triple-Negative Breast Cancer. J Natl Cancer Inst. 2021;113:1733-1743.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 66]  [Cited by in RCA: 105]  [Article Influence: 26.3]  [Reference Citation Analysis (0)]
8.  Kalantari Khandani N, Ghahremanloo A, Hashemy SI. Role of tumor microenvironment in the regulation of PD-L1: A novel role in resistance to cancer immunotherapy. J Cell Physiol. 2020;235:6496-6506.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 13]  [Cited by in RCA: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
9.  Yi M, Niu M, Xu L, Luo S, Wu K. Regulation of PD-L1 expression in the tumor microenvironment. J Hematol Oncol. 2021;14:10.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 103]  [Cited by in RCA: 417]  [Article Influence: 104.3]  [Reference Citation Analysis (0)]
10.  Hanks BA. The "Inside" Story on Tumor-Expressed PD-L1. Cancer Res. 2022;82:2069-2071.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
11.  Wang Y, Zhou Y, Yang L, Lei L, He B, Cao J, Gao H. Challenges Coexist with Opportunities: Spatial Heterogeneity Expression of PD-L1 in Cancer Therapy. Adv Sci (Weinh). 2024;11:e2303175.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 7]  [Cited by in RCA: 25]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
12.  Xiong W, Gao Y, Wei W, Zhang J. Extracellular and nuclear PD-L1 in modulating cancer immunotherapy. Trends Cancer. 2021;7:837-846.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 15]  [Cited by in RCA: 56]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
13.  Zubareva E, Senchukova M, Karmakova T. Predictive significance of HIF-1α, Snail, and PD-L1 expression in breast cancer. Clin Exp Med. 2023;23:2369-2383.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
14.  Zubareva EY, Senchukova MA, Saidler NV. Cytoplasmic and nuclear programmed death ligand 1 expression in peritumoral stromal cells in breast cancer: Prognostic and predictive value. World J Exp Med. 2025;15:102761.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (2)]
15.  Zubareva E, Senchukova M, Shubin D, Prokofiev A. Assessment of PD-L 1 expression in a tumor improves the accuracy of predicting the risk of regional breast cancer metastases. Trends Immunother. 2023;7:2414.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 1]  [Cited by in RCA: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
16.  Zhang Y, Chen H, Mo H, Zhao N, Sun X, Liu B, Gao R, Xu B, Zhang Z, Liu Z, Ma F. Distinct cellular mechanisms underlie chemotherapies and PD-L1 blockade combinations in triple-negative breast cancer. Cancer Cell. 2025;43:446-463.e7.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 4]  [Cited by in RCA: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
17.  Dieci MV, Miglietta F, Guarneri V. Immune Infiltrates in Breast Cancer: Recent Updates and Clinical Implications. Cells. 2021;10:223.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 75]  [Cited by in RCA: 148]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
18.  van den Ende NS, Nguyen AH, Jager A, Kok M, Debets R, van Deurzen CHM. Triple-Negative Breast Cancer and Predictive Markers of Response to Neoadjuvant Chemotherapy: A Systematic Review. Int J Mol Sci. 2023;24:2969.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 89]  [Cited by in RCA: 82]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
19.  Wang Y, Zong B, Yu Y, Wang Y, Tang Z, Chen R, Huang M, Liu S. Ki67 Index Changes and Tumor-Infiltrating Lymphocyte Levels Impact the Prognosis of Triple-Negative Breast Cancer Patients With Residual Disease After Neoadjuvant Chemotherapy. Front Oncol. 2021;11:668610.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
20.  Hammerl D, Massink MPG, Smid M, van Deurzen CHM, Meijers-Heijboer HEJ, Waisfisz Q, Debets R, Martens JWM. Clonality, Antigen Recognition, and Suppression of CD8(+) T Cells Differentially Affect Prognosis of Breast Cancer Subtypes. Clin Cancer Res. 2020;26:505-517.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 18]  [Cited by in RCA: 36]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
21.  Loi S, Drubay D, Adams S, Pruneri G, Francis PA, Lacroix-Triki M, Joensuu H, Dieci MV, Badve S, Demaria S, Gray R, Munzone E, Lemonnier J, Sotiriou C, Piccart MJ, Kellokumpu-Lehtinen PL, Vingiani A, Gray K, Andre F, Denkert C, Salgado R, Michiels S. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J Clin Oncol. 2019;37:559-569.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 547]  [Cited by in RCA: 586]  [Article Influence: 97.7]  [Reference Citation Analysis (0)]
22.  Arole V, Nitta H, Wei L, Shen T, Parwani AV, Li Z. M2 tumor-associated macrophages play important role in predicting response to neoadjuvant chemotherapy in triple-negative breast carcinoma. Breast Cancer Res Treat. 2021;188:37-42.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 16]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
23.  Ni Y, Tsang JY, Shao Y, Poon IK, Tam F, Shea KH, Tse GM. Combining Analysis of Tumor-infiltrating Lymphocytes (TIL) and PD-L1 Refined the Prognostication of Breast Cancer Subtypes. Oncologist. 2022;27:e313-e327.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 3]  [Cited by in RCA: 23]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
24.  Shin E, Kim HM, Koo JS. Expression of PD-L1 in breast invasive lobular carcinoma. PLoS One. 2024;19:e0309170.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
25.  Emens LA, Molinero L, Loi S, Rugo HS, Schneeweiss A, Diéras V, Iwata H, Barrios CH, Nechaeva M, Nguyen-Duc A, Chui SY, Husain A, Winer EP, Adams S, Schmid P. Atezolizumab and nab-Paclitaxel in Advanced Triple-Negative Breast Cancer: Biomarker Evaluation of the IMpassion130 Study. J Natl Cancer Inst. 2021;113:1005-1016.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 75]  [Cited by in RCA: 218]  [Article Influence: 54.5]  [Reference Citation Analysis (0)]
26.  Yam C, Yen EY, Chang JT, Bassett RL, Alatrash G, Garber H, Huo L, Yang F, Philips AV, Ding QQ, Lim B, Ueno NT, Kannan K, Sun X, Sun B, Parra Cuentas ER, Symmans WF, White JB, Ravenberg E, Seth S, Guerriero JL, Rauch GM, Damodaran S, Litton JK, Wargo JA, Hortobagyi GN, Futreal A, Wistuba II, Sun R, Moulder SL, Mittendorf EA. Immune Phenotype and Response to Neoadjuvant Therapy in Triple-Negative Breast Cancer. Clin Cancer Res. 2021;27:5365-5375.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 48]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
27.  Wang L, Guo W, Guo Z, Yu J, Tan J, Simons DL, Hu K, Liu X, Zhou Q, Zheng Y, Colt EA, Yim J, Waisman J, Lee PP. PD-L1-expressing tumor-associated macrophages are immunostimulatory and associate with good clinical outcome in human breast cancer. Cell Rep Med. 2024;5:101420.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 36]  [Cited by in RCA: 31]  [Article Influence: 31.0]  [Reference Citation Analysis (0)]
28.  Kossai M, Radosevic-Robin N, Penault-Llorca F. Refining patient selection for breast cancer immunotherapy: beyond PD-L1. ESMO Open. 2021;6:100257.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 21]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
29.  Sun WY, Lee YK, Koo JS. Expression of PD-L1 in triple-negative breast cancer based on different immunohistochemical antibodies. J Transl Med. 2016;14:173.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 78]  [Cited by in RCA: 95]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
30.  Yazaki S, Shimoi T, Yoshida M, Sumiyoshi-Okuma H, Arakaki M, Saito A, Kita S, Yamamoto K, Kojima Y, Nishikawa T, Tanioka M, Sudo K, Noguchi E, Murata T, Shiino S, Takayama S, Suto A, Ohe Y, Fujiwara Y, Yonemori K. Integrative prognostic analysis of tumor-infiltrating lymphocytes, CD8, CD20, programmed cell death-ligand 1, and tertiary lymphoid structures in patients with early-stage triple-negative breast cancer who did not receive adjuvant chemotherapy. Breast Cancer Res Treat. 2023;197:287-297.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 11]  [Reference Citation Analysis (0)]
31.  Huynh CG, Huynh NX, Truong HBT, Thai TT, Doan TPT. PD-L1 and TILs expression and their association with clinicopathological characteristics in Vietnamese women with primary invasive breast cancer. Medicine (Baltimore). 2023;102:e34222.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
32.  Zhang L, Wang XI, Ding J, Sun Q, Zhang S. The predictive and prognostic value of Foxp3+/CD25+ regulatory T cells and PD-L1 expression in triple negative breast cancer. Ann Diagn Pathol. 2019;40:143-151.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 21]  [Cited by in RCA: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
33.  Huang W, Ran R, Shao B, Li H. Prognostic and clinicopathological value of PD-L1 expression in primary breast cancer: a meta-analysis. Breast Cancer Res Treat. 2019;178:17-33.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 50]  [Cited by in RCA: 72]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
34.  Tsang JY, Au WL, Lo KY, Ni YB, Hlaing T, Hu J, Chan SK, Chan KF, Cheung SY, Tse GM. PD-L1 expression and tumor infiltrating PD-1+ lymphocytes associated with outcome in HER2+ breast cancer patients. Breast Cancer Res Treat. 2017;162:19-30.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 68]  [Cited by in RCA: 77]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
35.  Davey MG, Ryan ÉJ, Davey MS, Lowery AJ, Miller N, Kerin MJ. Clinicopathological and prognostic significance of programmed cell death ligand 1 expression in patients diagnosed with breast cancer: meta-analysis. Br J Surg. 2021;108:622-631.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 17]  [Cited by in RCA: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
36.  Savaş P, Serin G, Gürsoy P, Zekioğlu O, Özdemir N. Prognosis-related novel immunostaining pattern for programmed cell death ligand 1 and prognostic value of tumour-infiltrating lymphocytes in triple-negative breast cancer. Pol J Pathol. 2023;74:65-74.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
37.  Gajaria PK, Gupta MR, Patil A, Desai SB, Shet TM. Programmed cell death ligand - 1 expression in triple negative breast carcinoma and its prognostic significance in Indian population. Indian J Pathol Microbiol. 2021;64:664-670.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
38.  Angelico G, Broggi G, Tinnirello G, Puzzo L, Vecchio GM, Salvatorelli L, Memeo L, Santoro A, Farina J, Mulé A, Magro G, Caltabiano R. Tumor Infiltrating Lymphocytes (TILS) and PD-L1 Expression in Breast Cancer: A Review of Current Evidence and Prognostic Implications from Pathologist's Perspective. Cancers (Basel). 2023;15:4479.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 20]  [Reference Citation Analysis (0)]
39.  Stovgaard ES, Dyhl-Polk A, Roslind A, Balslev E, Nielsen D. PD-L1 expression in breast cancer: expression in subtypes and prognostic significance: a systematic review. Breast Cancer Res Treat. 2019;174:571-584.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 55]  [Cited by in RCA: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
40.  Lee SE, Park HY, Lim SD, Han HS, Yoo YB, Kim WS. Concordance of Programmed Death-Ligand 1 Expression between SP142 and 22C3/SP263 Assays in Triple-Negative Breast Cancer. J Breast Cancer. 2020;23:303-313.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 21]  [Cited by in RCA: 28]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
41.  Reisenbichler ES, Han G, Bellizzi A, Bossuyt V, Brock J, Cole K, Fadare O, Hameed O, Hanley K, Harrison BT, Kuba MG, Ly A, Miller D, Podoll M, Roden AC, Singh K, Sanders MA, Wei S, Wen H, Pelekanou V, Yaghoobi V, Ahmed F, Pusztai L, Rimm DL. Prospective multi-institutional evaluation of pathologist assessment of PD-L1 assays for patient selection in triple negative breast cancer. Mod Pathol. 2020;33:1746-1752.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 73]  [Cited by in RCA: 103]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
42.  Sigurjonsdottir G, De Marchi T, Ehinger A, Hartman J, Bosch A, Staaf J, Killander F, Niméus E. Comparison of SP142 and 22C3 PD-L1 assays in a population-based cohort of triple-negative breast cancer patients in the context of their clinically established scoring algorithms. Breast Cancer Res. 2023;25:123.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]