This article presents a comprehensive review of the current evidence related to PD‐L1 testing in breast cancer and its use as a biomarker in prediction of response to immunotherapies.
Keywords: Programmed cell death ligand 1, Immunotherapy, Immune checkpoint inhibitors, Breast cancer, Biomarkers, Patient selection
Abstract
In the light of recent advances in the immunotherapy field for breast cancer (BC) treatment, especially in the triple‐negative subtype, the identification of reliable biomarkers capable of improving patient selection is paramount, because only a portion of patients seem to derive benefit from this appealing treatment strategy. In this context, the role of programmed cell death ligand 1 (PD‐L1) as a potential prognostic and/or predictive biomarker has been intensively explored, with controversial results. The aim of the present review is to collect available evidence on the biological relevance and clinical utility of PD‐L1 expression in BC, with particular emphasis on technical aspects, prognostic implications, and predictive value of this promising biomarker.
Implications for Practice.
In the light of the promising results coming from trials of immune checkpoint inhibitors for breast cancer treatment, the potential predictive and/or prognostic role of programmed cell death ligand 1 (PD‐L1) in breast cancer has gained increasing interest. This review provides clinicians with an overview of the available clinical evidence regarding PD‐L1 as a biomarker in breast cancer, focusing on both data with a possible direct impact on clinic and methodological pitfalls that need to be addressed in order to optimize PD‐L1 implementation as a clinically useful tool for breast cancer management.
摘要
鉴于人们在乳腺癌 (BC)(特别是三阴性亚型)治疗的免疫治疗领域取得的最新进展,确定能够改善患者选择的可靠生物标志物至关重要,因为仅有一部分患者似乎可以从这种具有吸引力的治疗策略中获益。在这种背景下,程序性细胞死亡配体 1 (PD‐L1) 作为一种潜在的预后和/或预测生物标志物的作用已被深入探索,并取得了具有争议性的结果。本报告旨在收集关于BC中的 PD‐L1 表达的生物相关性和临床实用性的有效证据,特别强调了技术因素、预后意义以及这种前景可期的生物标志物的预测价值。
实践意义:鉴于针对乳腺癌治疗的免疫检查点抑制剂的试验结果前景可期,程序性细胞死亡配体 1 (PD‐L1) 在乳腺癌中的潜在预测和/或预后作用已得到人们越来越多的重视。本综述为临床医生提供了关于 PD‐L1 作为乳腺癌生物标志物的现有临床证据的概述,重点关注可能对临床产生直接影响的数据和需要解决的方法学缺陷,从而对在临床上对乳腺癌管理十分有益的工具 PD‐L1 的应用进行优化。
Introduction
In the constantly evolving era of immunotherapy, the blockade of the programmed cell death 1 (PD1)/programmed cell death ligand 1 (PD‐L1) immune checkpoint pathway represents one of the most promising strategies to revert immune evasion in the cancer immunoediting process. PD1 is a cell surface membrane protein, member of the B7 family of immune checkpoints, which is activated by its ligands PD‐L1 and PD‐L2. Activated lymphocytes induce the expression of PD‐L1 on the surface of T cells, Natural Killer cells (NK), macrophages, and, most importantly, tumor cells, through different mechanisms, among which the secretion of interferon gamma (IFN‐gamma) is the most important. Once engaged, the PD1/PD‐L1 pathway leads to the mitigation of T‐cell‐mediated immune response through the inhibition of T‐cell activation and the promotion of the regulatory function of T lymphocytes. In solid tumors, this process may be exploited by the tumor microenvironment to silence or at least attenuate the antitumor immune response [1], [2], [3].
The use of immune checkpoint inhibitors led to striking results in several solid tumors such as melanoma, bladder cancer, and non‐small cell lung cancer (NSCLC). Although breast cancer (BC) is not traditionally considered immunogenic, a growing body of evidence suggests that certain BC subtypes, namely, triple‐negative (TN) and human epidermal growth receptor 2 (HER2)‐positive (HER2+), may exhibit a strong infiltration by immune cells with prognostic and even predictive implications [4], [5]. This evidence also fostered the evaluation of immune checkpoint inhibitors in BC, with promising results, especially in the TN subtype. Importantly, a statistically significant progression‐free survival (PFS) benefit with the combination of atezolizumab plus nab‐paclitaxel compared with placebo plus nab‐paclitaxel in patients with TN metastatic BC (MBC) has recently been reported [6]. In this context, as a result of the growing interest in the identification of reliable prognostic and/or predictive biomarkers for patients treated with immunotherapy, the scientific interest focused on PD‐L1 expression. However, the clinical relevance of PD‐L1 as a biomarker in BC remains to be clearly defined.
We therefore conducted a review on the role of PD‐L1 expression in BC with the aim of collecting available evidence coming from clinical studies.
Materials and Methods
Relevant studies were searched in the PubMed database with the following keywords: “breast cancer,” “breast tumor,” “breast neoplasm,” “programmed cell death ligand 1,” “programmed cell death 1,” “PD‐L1,” “PD‐1,” “B7‐H1,” “CD274,” “CD279,” and “immune checkpoint.”
In addition, reference lists of retrieved articles were manually reviewed.
In order to include the most recent data, we also searched for relevant studies presented in the form of abstracts in major international medical oncology conferences (American Society of Clinical Oncology 2016, 2017, 2018; San Antonio Breast Cancer Symposium 2016, 2017, 2018; European Society for Medical Oncology 2016, 2017, 2018).
The language was restricted to English.
PD‐L1 Expression in BC
PD‐L1 expression by immunohistochemistry (IHC) or gene expression has been observed in approximately 20%–40% of all BCs across different studies, and it has been shown to be significantly higher in invasive disease compared with normal breast tissue [7], [8], [9], [10], [11], [12] and premalignant lesions as in situ carcinoma [13].
In addition, it has been reported that PD‐L1 is differentially expressed across different BC subtypes. In particular, available evidence consistently reports greater expression of PD‐L1 in the TN subtype (up to 60% of PD‐L1 expression) compared with non‐TNBC [14], [15], [16], [17], [18], [19], [20], [21]. These data appear to be coherent with the observation that PD‐L1 tumor expression is positively associated with stromal tumor‐infiltrating lymphocytes (TILs) in this BC subtype [22], [23], [24], [25], [26], [27], [28], [29], which is known to be more frequently infiltrated by stromal TILs than non‐TNBC [5], as summarized in Table 1, thus possibly suggesting that these two immune biomarkers tend to run parallel.
Table 1. Studies reporting a correlation between PD‐L1 and TILs.
Abbreviations: BC, breast cancer; DapB, dihydrodipicolinate reductase; H&E, hematoxylin and eosin; HER2, human epidermal growth receptor 2; IBC, inflammatory breast cancer; IC, immune cells; IHC, immunohistochemistry; iTILs, intratumoral TILs; LPBC, lymphocyte‐predominant breast cancer; NA, not available; PD‐L1, programmed cell death ligand 1; QIF, quantitative fluorescence; sTILs, stromal TILs; TAM, tumor‐associated macrophage; TC, tumor cells; TILs, tumor infiltrating lymphocytes; TN, triple‐negative; T/NB ratio, tumor/normal breast ratio.
Data on PD‐L1 expression in HER2‐positive BC are more controversial. In fact, whereas in some studies HER2 positivity has been correlated with higher expression of PD‐L1 (up to 50%) compared with HER2‐negative BC [10], [14], [18], [20], others failed to report any difference [9], [15], [16], [17], [19], [30].
Results from two meta‐analyses including partially overlapping studies confirmed the greater PD‐L1 expression in TNBC [31], [32] compared with non‐TN subtypes but were not consistent in reporting the association between HER2 status and PD‐L1 expression.
However, when considering molecular intrinsic BC subtypes by gene expression profiling rather than IHC, both basal‐like and HER2‐enriched subgroups were found to be enriched in PD‐L1 expression with respect to luminal BC [10], [25], [33], [34], [35], thus possibly highlighting that a more subtle classification in BC subtypes may help better capture the relevance of immune microenvironment for TN and HER2‐positive BC.
Association with Clinicopathological Characteristics
Several studies evaluated PD‐L1 expression according to baseline clinicopathological features of patients with BC, consistently reporting a correlation between higher PD‐L1 expression and unfavorable classic prognostic factors, particularly poorer histological grade [10], [11], [14], [18], [30], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45], higher proliferative index [10], [38], [39], [46], [47], more advanced N stage [14], [30], [44], [48], larger tumor size [10], [11], [14], [30], [44], [48], and younger age at BC diagnosis [14], [39], [40], [48].
A possible explanation for such observations may be attributable to the immune escape phenomenon. Indeed, a high expression of PD‐L1 may reflect the activation of the immune checkpoint PD1/PD‐L1 pathway leading to the mitigation of the host's antitumor immune response, thus ultimately resulting in increased tumor aggressiveness [31]. Although intriguing, further evidence is needed in order to confirm this hypothesis. In addition, it should be considered that aggressive clinicopathological characteristics are typical features of the TN subtype, thus possibly posing a bias in the interpretation of these data.
Association with Prognosis
Several authors explored the possible prognostic role of PD‐L1 expression in early BC, reporting conflicting results, as shown in Table 2.
Table 2. Prognostic role of PD‐L1 in untreated BC.
Abbreviations: BC, breast cancer; BCSS, breast cancer‐specific survival; DapB, dihydrodipicolinate reductase; DFS, disease‐free survival; DMFS, distant metastasis‐free survival; EBC, early breast cancer; FOVs, fields of view; HER2, human epidermal growth receptor 2; HPF, high‐power field; HR, hormone receptor; IBC, inflammatory breast cancer; IC, immune cells; IHC, immunohistochemistry; MFS, metastasis‐free survival; OS, overall survival; OSS, overall specific survival; PD‐L1, programmed cell death ligand 1; QIF, quantitative fluorescence; qPCR, quantitative polymerase chain reaction; qRT‐PCR, quantitative reverse transcription polymerase chain reaction; RFS, recurrence‐free survival; ROC, receiver operating characteristic; SISH, silver in situ hybridization; TC, tumor cells; TN, triple‐negative.
PD‐L1 expression evaluated by IHC on untreated primary BC has been associated with both better and poorer clinical outcome. In particular, several authors reported poorer disease‐free survival (DFS)/recurrence‐free survival (RFS) and/or overall survival (OS) in cases of higher PD‐L1 expression on primary BC [14], [19], [30], [49], [50], [51], [52], [53], especially in the TN subtype [19], [50], [53]. These results seems to be consistent with the previously mentioned correlation between PD‐L1 expression and unfavorable clinicopathological BC features, thus possibly indicating that PD‐L1 expression may be part of the immune evasion process taking place in the context of tumor microenvironment [14].
Counterintuitively, a prognostic role in the opposite direction has also been suggested.
In particular, PD‐L1 expression has been positively and independently associated with DFS/RFS and/or OS in several unselected primary BC cohorts [16], [21], [22], [24], [25], [40], [44], [46], [48], [54], [55], [56], [57], [58]. In addition, when considering specific BC subtypes, it has been suggested that PD‐L1 protein expression may retain a positive prognostic role in TNBC [21], [22], [24], [46], [55], [56] and in HER2+ BC, in both trastuzumab‐treated and untreated patients [44], [57].
Such inconsistency may reflect the fact that in the above‐mentioned studies, PD‐L1 expression was assessed at the protein level by IHC. However, it should be considered that the assessment of PD‐L1 by IHC on BC tissue lacks standardization, thus possibly impairing the reproducibility of results across different studies. Interestingly, when considering the prognostic role of PD‐L1 assessed at mRNA expression level, available evidence is consistent in suggesting an association between PD‐L1 and better outcome in terms of both metastasis‐free survival/DFS and OS, especially in TN/basal‐like BC [10], [12], [59]. Actually, although the positive prognostic value of PD‐L1 may appear paradoxical, it has been reported that mRNA expression of immunosuppressive checkpoint molecules such as PD‐L1 strongly correlates with other immune markers with proimmune activity [27]. In addition, a positive correlation between higher PD‐L1 mRNA expression and an immune signature of genes associated with a strong cytotoxic activity has been reported [10]. In this context, PD‐L1 expression may reflect a negative feedback mechanism following the activation of cytotoxic antitumor immune response, rather than an isolated immunosuppressive process. Indeed, this hypothesis may also be biologically plausible if considering that the expression of immune checkpoint molecules is also triggered by activated T cells through an interferon‐gamma‐mediated feedback mechanism [60].
Although the positive prognostic value of PD‐L1 may appear paradoxical, it has been reported that mRNA expression of immunosuppressive checkpoint molecules such as PD‐L1 strongly correlates with other immune markers with proimmune activity. In addition, a positive correlation between higher PD‐L1 mRNA expression and an immune signature of genes associated with a strong cytotoxic activity has been reported.
Association with Treatment Response
Immunotherapy.
In the last decades, immune checkpoint inhibitors have emerged as a promising treatment strategy for metastatic BC, especially in the TN subtype, where the immune microenvironment is thought to play a major role in tumorigenesis and tumor progression. Indeed, the encouraging results from several early‐phase trials testing immune checkpoint inhibitors in heavily pretreated MBC fostered the evaluation of such agents in phase II and III trials both as single agent and in combination with conventional treatment strategies, including targeted therapies. However, only a portion of patients seem to derive benefit from these agents. For this reason, a proper and reliable selection of patients is strongly needed in order to discriminate potential responders from nonresponders.
In this context, PD‐L1 expression has been proposed as potentially capable of predicting the benefit from anti‐PD1/PD‐L1 agents in the context of several prospective trials, as summarized in Table 3.
Table 3. Studies of immune checkpoint inhibitors: association with PD‐L1 status.
Patients included in the translational analysis.
Abbreviations: BC, breast cancer; CPS, combined positive score; DCR, disease control rate; HER2, human epidermal growth receptor 2; IC, immune cells; IHC, immunohistochemistry; ITT, intention‐to‐treat population; MBC, metastatic breast cancer; NA, not available; ORR, overall response rate; OS, overall survival; PD‐L1, programmed cell death ligand 1; PFS, progression‐free survival; TC, tumor cells; TNMBC, triple‐negative metastatic breast cancer.
In detail, the KEYNOTE‐012 phase Ib trial tested pembrolizumab monotherapy in advanced TNBC. The enrollment of only PD‐L1‐positive patients did not allow for a conclusion on the predictive role of PD‐L1 expression to be drawn; however, the authors reported a trend toward greater clinical benefit from pembrolizumab in cases of higher PD‐L1 expression evaluated with a prototype scoring assay (IHC assay using clone 22C3) [61].
Pembrolizumab monotherapy has also been tested in the context of the KEYNOTE‐086 phase II trial, which included two cohorts of patients: cohort A enrolled previously treated patients with TNMBC irrespective of PD‐L1 status, whereas cohort B included previously untreated PD‐L1‐positive (IHC assay using clone 22C3) TNMBC. In cohort A, overall response rate (ORR) appeared to be modest and independent from PD‐L1 status; however, a trend toward a greater clinical benefit from pembrolizumab in terms of both disease control rate (DCR) and duration of response was observed in PD‐L1‐positive versus PD‐L1‐negative patients [62]. In cohort B, higher ORR (21.4%) and longer duration of responses (median 10.4 months) were observed as compared with cohort A, thus strengthening the hypothesis that patients with BC harboring PD‐L1 positivity may show good responses to pembrolizumab monotherapy, especially in the earliest lines of treatment for metastatic disease [63].
The combination of pembrolizumab plus eribulin has been evaluated in the context of the KEYNOTE‐150 phase IB/II trial, which included TNMBC regardless of PD‐L1 status. No association between treatment response and PD‐L1 status (IHC assay using clone 22C3) was reported [64].
The association of PD‐L1 protein expression with treatment response has also been evaluated in HER2+ BC. In the phase Ib/II PANACEA trial, the combination of pembrolizumab plus trastuzumab was explored in HER2+ MBC in both PD‐L1‐positive and ‐negative patients (IHC assay using clone 22C3). The authors reported higher ORR (15.2% vs. 0%) and longer 1‐year OS (65% vs. 12%) for PD‐L1‐positive as compared with PD‐L1‐negative patients [28].
Atezolizumab monotherapy was tested in an expansion cohort of a phase Ia trial of both PD‐L1‐positive and PD‐L1‐negative TNMBC. The authors reported that PD‐L1 positivity was associated with higher response rates [65].
In the phase Ib trial of atezolizumab in combination with nab‐paclitaxel for TNMBC (regardless of PD‐L1 status), the biomarker analysis revealed that PD‐L1 expression on either tumor or immune cells (by IHC, clone SP142;) correlated with ORR [66], [67].
The subsequent phase III trial—Impassion130—randomized patients with TNMBC to receive atezolizumab + nab‐paclitaxel versus placebo + nab‐paclitaxel. Stratification factors included PD‐L1 status assessed on TILs by IHC (clone SP142; intention‐to‐treat [ITT] population = 902, PD‐L1‐positive patients = 369). The results showed only a slight PFS improvement associated with atezolizumab (median 7.2 vs. 5.5 months; hazard ratio [HR] 0.80; 95% confidence interval [CI] 0.69–0.92; p = .002) and no effect on OS (median 21.3 vs. 17.6 months; HR 0.84; 95% CI 0.69–1.02; p = not significant) in the ITT population. However, when considering only PD‐L1‐positive patients, a significant PFS benefit (median 7.5 vs. 5.0 months; HR 0.62; 95% CI 0.49–0.78; p < .001) and a trend in improved OS (25 vs. 15.5 months; HR 0.62; 95% CI 0.45–0.86; no formal testing performed) were observed in the atezolizumab arm compared with the placebo arm, thus demonstrating for the first time in a randomized clinical trial the possible predictive value of PD‐L1 in TNMBC [6].
Single anti‐PD‐L1 agent avelumab has been evaluated in a cohort of MBC in the context of the phase Ib Javelin trial. In the biomarker analysis, different compartments for PD‐L1 evaluation (tumor cells vs. tumor‐associated immune cells) and different PD‐L1 positivity thresholds (for tumor cells: ≥1% vs. ≥5% with any staining intensity and ≥ 25% with moderate‐to‐high staining; for tumor‐associated immune cells: ≥10% at any staining) were evaluated, reporting a trend toward higher ORR in the overall population and TN subgroup when PD‐L1 positivity was determined on tumor‐associated immune cells (≥10%) rather than on tumor cells [68].
Neoadjuvant Chemotherapy.
In the last decades, neoadjuvant chemotherapy (NACT) has been increasingly used in the management of locally advanced BC, especially in the TN and HER2+ subtype, where the achievement of a pathological complete response (pCR) after NACT represents a strong positive prognostic factor [69]. For this reason, the identification of reliable biomarkers capable of identifying the subset of patients more likely to obtain a pCR after NACT is of great interest in BC translational research.
In this context, the possible association between baseline PD‐L1 expression and efficacy of conventional neoadjuvant treatments has been recently evaluated. Studies addressing this issue have reported partially conflicting results, as shown in Table 4.
Table 4. Studies reporting an association between pretreatment PD‐L1 and response to neoadjuvant therapy.
Patients included in the PD‐L1 analysis.
Abbreviations: BC, breast cancer; CT, chemotherapy; ET, endocrine therapy; FOVs, fields of view; HER2, human epidermal growth receptor 2; HR, hormone receptor; IC, immune cells; IHC, immunohistochemistry; MP, Miller‐Payne; NA, not available; pCR, pathologic complete response, PD‐L1, programmed cell death ligand 1; RCB, residual cancer burden; RCT, randomized clinical trial; RT‐PCR, real‐time polymerase chain reaction; TC, tumor cells; TNBC, triple‐negative breast cancer.
In detail, PD‐L1 mRNA upregulation has been associated with increased pCR rates in two cohorts of patients with BC treated with anthracycline‐based chemotherapy (CT) [10] in a large retrospective study and anthracycline‐taxane ± carboplatin [27] in the context of the GeparSixto randomized trial. The association between PD‐L1 and pCR was only confirmed for basal‐like/TN and HER2‐enriched/HER2‐positive subsets.
A positive relationship between PD‐L1 protein expression and pCR has been reported as well. In particular, two retrospective studies reported that higher levels of PD‐L1 expression were independently associated with increased pCR rates after anthracycline‐based CT in hormone receptor‐positive/HER2‐negative BC [17] and in TNBC [26], respectively.
In addition, the translational analysis of the phase II HER2+ hormone receptor‐negative WSG‐ADAPT trial revealed that baseline PD‐L1 expression on infiltrating immune cells was positively associated with pCR in the Trastuzumab emtansine (T‐DM1) arm [70]. A similar association between baseline PD‐L1 protein expression and pCR has been reported in the HER2‐negative subtype in the context of two prospective trials testing neoadjuvant anthracycline‐based CT ± bevacizumab, where PD‐L1 was reported as positively associated with better response to neoadjuvant therapy [71], [72].
On the other hand, PD‐L1 protein expression has also been related to pCR in the opposite direction in the context of a retrospective study reporting that patients with TNBC with higher basal PD‐L1 protein expression experienced lower rates of pCR after anthracycline‐taxane NACT [19].
Although these contradictory results indicate that further study of the possible role of PD‐L1 in affecting either response or resistance to conventional neoadjuvant treatments in the context of adequately powered clinical studies is needed, it must be noted that the most robust body of evidence supports the notion that baseline PD‐L1 may be positively associated with pCR. The potential capability of baseline PD‐L1 to predict pCR after NACT may gain further relevance when considering that CT could be strategically used with the aim of enhancing antitumor immune response, turning a cold tumor into a hot tumor, and ultimately boosting the efficacy of immunotherapy.
The strategy of combining immunotherapy and chemotherapy in the neoadjuvant setting is the subject of several clinical trials, some of which are ongoing ([73], [74], [75], [76], NCT02620280). Moreover, post‐neoadjuvant immunotherapy for patients with triple‐negative breast cancer who did not achieve a pCR after NACT is being tested in randomized trials (NCT02954874, NCT02926196). As discussed further in this review, because PD‐L1 is a dynamic marker, its expression can be further modulated by NACT, and ongoing adjuvant immunotherapy trials will possibly clarify whether post‐treatment PD‐L1 is able to predict immunotherapy efficacy.
PD‐L1 Testing: Technical and Biological Heterogeneity
The implementation of PD‐L1 as a reliable biomarker for the selection or exclusion of patients with BC for immunotherapy has thus far been complicated by several issues mainly attributable to technical and biological heterogeneity, as summarized in Figure 1.
Figure 1.
Programmed cell death ligand 1 (PD‐L1) testing in breast cancer (BC): technical and biological heterogeneity. (A): Analytical level: PD‐L1 can be assessed at both protein and mRNA level. (B): Tumor microenvironment compartment: PD‐L1 expression can be detected on both tumor and stromal cells, such as tumor‐infiltrating lymphocytes, macrophages, and fibroblast‐like cells. (C): Temporal and spatial heterogeneity: PD‐L1 expression has been evaluated on both primary BC and matched metastatic lesions (lymph node metastases and/or distant metastases). (D): PD‐L1 has been mainly assessed on tumor tissue; however, it can also be detected on circulating tumor‐related material.
Abbreviations: AB, antibodies; IHC, immunohistochemistry; RT‐PCR, reverse transcriptase polymerase chain reaction.
Analytic Levels.
Available studies on the potential predictive/prognostic role of PD‐L1 in BC mainly focused on its expression at the protein level by IHC, frequently reporting conflicting and inconclusive results. Such inconsistency may reflect the current lack of standardization of PD‐L1 testing techniques, particularly regarding the reproducibility and specificity of available PD‐L1 antibodies and diversity of cutoff for positivity.
The comparison between different commercially available PD‐L1 antibodies revealed a general good concordance in BC, especially between Ventana SP263, Dako 22c3, and rbMCAL10, and between Dako 28‐8 and E1L3N [46], [77]. However, comparative analyses of different PD‐L1 antibodies on NSCLC tissue samples consistently reported low sensitivity of SP142 antibody, because it was associated with significantly lower rates of PD‐L1 detection on both tumor and immune cells [78], [79].
In addition, the adoption of different scoring systems and thresholds for PD‐L1 positivity may contribute to further reducing the reproducibility of PD‐L1 assessment. Indeed, in BC, discordant results were reported when different positivity cutoffs were applied and when tumor cells and tumor‐infiltrating immune cells were differentially considered [46], [77].
In this context, the adoption of digital pathology and software‐assisted methods may increase accuracy, reduce human error, and ultimately improve reproducibility of PD‐L1 assessment and interpretation [80]. Recently, PD‐L1 expression measured by IHC and assessed by digital pathology platforms has been positively associated with outcome in two cohorts of patients with TN early BC treated with surgery and standard CT [29], [81].
In addition, alternative analytic methods for PD‐L1 assessment have been also suggested.
In particular, the evaluation of PD‐L1 at the mRNA level emerged as a method potentially capable of overcoming the major limitations of PD‐L1 assessment by IHC, because it relies on an antibody‐independent method. Indeed, the more consistent data on the possible positive prognostic role of PD‐L1 have been derived from studies evaluating PD‐L1 mRNA rather than protein expression. However, a possible limitation of mRNA evaluation may be that it does not discriminate between PD‐L1 expression on tumor cells and nontumor cells, such as TILs [10].
Site of PD‐L1 Expression.
Cellular Compartment.
Tumoral expression of PD‐L1 encompasses both membrane and cytoplasm. It is unclear whether a differential evaluation of the two compartments may affect PD‐L1 biological and clinical value in BC. However, preliminary data coming from a TNBC patient cohort suggest that cytoplasmic PD‐L1 expression may be more biologically relevant compared with the membranous expression [21]. Of course, these data need to be confirmed in other clinical series.
Tumor Microenvironment Compartment.
An additional source of variability in the assessment of PD‐L1 protein expression is that PD‐L1 may be evaluated in both tumor and stromal compartments.
In contrast to NSCLC, where regulatory approval for anti‐PD1/PD‐L1 therapy (pembrolizumab) is based on PD‐L1 positivity assessed on tumor cells [82], in BC, PD‐L1 seems to be predominantly expressed by stromal compartment [20], [25], [29], [38], [83], [84]. However, so far, no consistent data exist on the possible biological and clinical implications of a differential expression of PD‐L1 by either tumor cells or tumor‐infiltrating immune cells. Of note, it has been suggested that a significant proportion of PD‐L1‐negative tumors assessed on tumor cells may actually be classified as PD‐L1 positive if assessed on stromal immune cells, thus enriching the subset of patients that might be candidates for immunotherapy [20], [25], [38]. However, Tawfik and colleagues also suggested that the adoption of a stricter cutoff for PD‐L1 positivity (10% instead of 1%) may help increase the agreement between stromal and tumor compartments [38].
In contrast to NSCLC, where regulatory approval for anti‐PD1/PD‐L1 therapy (pembrolizumab) is based on PD‐L1 positivity assessed on tumor cells, in BC, PD‐L1 seems to be predominantly expressed by stromal compartment. However, so far, no consistent data exist on the possible biological and clinical implications of a differential expression of PD‐L1 by either tumor cells or tumor‐infiltrating immune cells.
Interestingly, results from the phase Ib Javelin trial with avelumab suggested that the predictive value of PD‐L1 may be larger when PD‐L1 is evaluated on TILs rather than tumor cells [68]. Indeed, the first phase III trial suggesting the predictive role of PD‐L1 in TNMBC actually defined as PD‐L1‐positive tumors those expressing PD‐L1 only on immune cells [6], [84].
It should also be noted that PD‐L1 stromal expression encompasses not only TILs but also macrophages and fibroblast‐like cells, thus further increasing the complexity of the biological role of PD‐L1 in possibly affecting response to immunotherapy [85].
Temporal and Spatial Heterogeneity.
In the last few years, several authors investigated the dynamic nature of PD‐L1 by assessing its protein expression in primary and matched metastatic tumor samples.
In more detail, higher concordance rates between primary and secondary lesions have been reported when PD‐L1 was assessed on tumor cells rather than on TILs [20], [38]. In addition, several authors reported that discordant cases in terms of PD‐L1 status between primary and secondary lesions (encompassing both distant and local lymph node metastases) tended to exhibit a gain in PD‐L1 expression rather than a loss [20], [36], [38], [42]. Finally, it has been reported that patients with TNBC with PD‐L1 gain from primary to paired local lymph node metastasis experienced worse DFS compared with patients with PD‐L1 negativity in both primary tumor and lymph node metastasis [42].
Although these findings seem to suggest that PD‐L1 may increase from primary to secondary lesions, available evidence on its spatial and temporal heterogeneity remains limited as well as potentially biased by the fact that the majority of available data are derived from studies assessing PD‐L1 on lymph node metastasis, where the reliability of immune‐related biomarker detection is currently unclear.
The immune landscape of metastatic lesions may be even more complex when considering that a trend in the opposite direction with regard to TILs has been reported. In particular, results from two large retrospective cohorts of patients with MBC showed that TILs tended to decrease from primary to metastatic lesions in the TN subtype [36], [83], especially in patients receiving CT (for the advanced disease) prior to metastasis biopsy [83].
It remains therefore unclear whether PD‐L1 expression assessed on secondary rather than primary lesions may provide additional and clinically relevant information. Indeed, Dieci et al. reported that whereas TILs assessed on metastatic lesions from patients with TNMBC were positively associated with outcome, stromal PD‐L1 expression did not retain any prognostic value (with either 5% or 1% cutoff for positivity) [83].
The dynamic nature of PD‐L1 has been further explored in the neoadjuvant setting, where modifications of PD‐L1 expression from baseline to post‐NACT BC samples were reported. In particular, it has recently been shown that PD‐L1 significantly increased on residual disease after NACT compared with baseline in a large retrospective cohort of patients with TN early BC [29]. The induction of PD‐L1 expression by chemotherapy is consistent with observations in other cancer types [85], [86], [87], [88], [89] and with the notion that chemotherapy is able to induce an adaptive immune response through various mechanisms, including immunogenic cell death and the activation of the damage response c‐GAS/STING [90]. Indeed, it has been shown that CT may boost the immunogenicity of the tumor by increasing tumor immune infiltrate from baseline to post‐NACT samples, with a high rate of conversion from low‐TIL to high‐TILs tumor [91]. Interestingly, in the advanced setting, results from the adaptive phase II randomized Tonic trial, testing the anti‐PD1 agent nivolumab after an induction treatment in TNBC revealed that induction chemotherapy (with doxorubicin or cisplatin) resulted in T‐cell and T‐cell clonality increase from baseline to on‐nivolumab biopsies of responders patients [92].
To conclude, although available data highlight the highly dynamic nature of PD‐L1, robust evidence on its spatial and temporal heterogeneity is missing, and it is not currently possible to draw a conclusion on the ideal timing for PD‐L1 testing.
Conclusion
Targeting the PD1‐PD‐L1 pathway is emerging as a promising treatment strategy for patients with BC, especially in the TN subtype. However, whereas some patients experience good response to immune checkpoint inhibitors, a subset of patients seem to derive little or no benefit.
Indeed, as already suggested by Adams et al. [93], who recently reviewed the current status of immunotherapy in BC, a closer understanding of tumor, microenvironment, and host factors that affect response to immunotherapy may help identify reliable biomarkers and thus ultimately optimize patient selection for immunotherapeutic strategies. In this context, PD‐L1 evaluation represents a good candidate. However, many technical and biological issues need to be addressed. In particular, PD‐L1 testing on BC tumor tissue currently lacks standardization in terms of diversity in assays (IHC, gene expression), antibodies for IHC testing, scoring systems and thresholds for PD‐L1 positivity, compartments of the tumor microenvironment included in the analysis (tumor cells, immune cells, or both), and nature of tumor samples (primary, metastatic, or post‐NACT), along with the lack of proper widespread resources in terms of PD‐L1 antibody platforms for PD‐L1 testing. In addition, no data are currently available on the effect of preanalytical variables (e.g., fixation time, type of fixative, storage, etc.) on the reproducibility of PD‐L1 testing in BC. Moreover, selection of the optimal PD‐L1 test and score to be incorporated in clinical trials is paramount in order to accurately understand the role of immunotherapy in selected patients. In this regard, practical risk‐assessment recommendations have recently been suggested for effective integration of biomarkers in clinical trials [94].
Indeed, further efforts are needed to implement PD‐L1 testing as a tool for properly selecting patients for immunotherapy. In this context, it is not acceptable that the same patient should be labeled as PD‐L1 positive or negative depending on which PD‐L1 assay or scoring system is used.
Furthermore, current scientific interest is pointed to the identification of alternative or complementary methods to improve patient selection for immunotherapy.
In this regard, TILs may provide additional information. In particular, translational analyses of three clinical trials of anti‐PD1/PD‐L1 agents revealed that patients with pretreatment higher TIL levels tended to derive a greater benefit from immune checkpoint inhibitors [28], [62], [63], [65], [95]. In detail, data from the KEYNOTE‐086 trial that predominantly relied on newly collected metastatic samples revealed a significant correlation between PD‐L1 and TILs. The latter were found to be positively associated with greater responses to pembrolizumab, especially in the first‐line setting (cohort A: ORR 6% vs. 2% for TIL ≥ vs. < median, respectively; median TILs 10% vs. 5% in responders vs. nonresponders, respectively; cohort B: ORR 39% vs. 9% for TIL ≥ vs. < median, respectively; median TILs 50% vs. 15% in responders vs. nonresponders, respectively) [62], [63]. In an expansion cohort of a phase Ia trial with atezolizumab in TNMBC, the presence of >10% TILs was associated with a trend toward higher ORR and longer OS [65]. In the PD‐L1‐positive cohort of the PANACEA Ib/II trial, higher baseline stromal TILs were significantly associated with better ORR (stromal TILs ≥5% vs. <5%: 39% vs. 5%) and DCR (stromal TILs ≥5% vs. <5%: 47% vs. 5%) [28].
However, preliminary translational analysis of the randomized phase III Impassion130 trial revealed that the evaluation of TILs did not provide additional predictive information beyond that provided by PD‐L1 status [6], [84]. Nevertheless, it should be noticed that the cutoff for TIL positivity (low vs. intermediate/high) was set at 10%. In addition, as already mentioned, results from both retrospective and prospective studies showed that PD‐L1 and TILs tend to be significantly associated with each other [22], [23], [24], [25], [26], [27], [28], [29]. For these reasons, the evaluation of TILs as a predictive biomarker for immunotherapy deserves further investigation in the light of the recent publication of a consensus for standardized TIL assessment in metastatic lesions [96]. In addition, the quantification of TILs does not require any additional tissue availability or processing because it may be performed on diagnostic hematoxylin and eosin‐stained slides, thus representing a more accessible and less expensive tool as compared with PD‐L1 evaluation by IHC.
Recently, the evaluation of PD‐L1 by liquid biopsy has emerged as a promising strategy potentially capable of better capturing the dynamic nature of this biomarker compared with its assessment on tumor tissue. Indeed, it has been stated that patients with BC frequently harbor PD‐L1‐positive circulating epithelial tumor cells [97], [98], peripheral blood mononuclear cells [99], or circulating tumor RNA. Interestingly, it has also been reported that serum PD‐L1 is associated with tumor burden [97], [99] and outcome [100]. These preliminary data suggest that liquid biopsy may represent a noninvasive and feasible strategy for dynamic assessment and serial monitoring of PD‐L1 of patients with BC, thus potentially providing a real‐time picture of PD‐L1 status.
The Cancer Genome Atlas data from more than 8,000 tumor samples (across 31 cancer types) revealed that PD1 mRNA may be a potential good predictor for anti‐PD1/PD‐L1 monotherapy activity [101].
Finally, although no data on patients with BC are currently available, gut microbiome and mutation burden have recently emerged as promising predictors of the benefit from immune checkpoint blockade in other solid malignancies, such as melanoma and NSCLC [4], [102], [103], [104], [105], [106]. These data fostered the conduction of several early‐stage clinical studies—which are currently ongoing—on the possible association between response to immunotherapy and these pioneering biomarkers in patients with BC (mutational burden: NCT01375842; gut microbiome: NCT02079662, NCT03358511).
Author Contributions
Conception/design: Federica Miglietta, Gaia Griguolo, Valentina Guarneri, Maria Vittoria Dieci
Collection and/or assembly of data: Federica Miglietta, Gaia Griguolo, Valentina Guarneri, Maria Vittoria Dieci
Manuscript writing: Federica Miglietta, Gaia Griguolo, Valentina Guarneri, Maria Vittoria Dieci
Final approval of manuscript: Federica Miglietta, Gaia Griguolo, Valentina Guarneri, Maria Vittoria Dieci
Disclosures
Maria Vittoria Dieci: Eli Lilly and Company, Genomic Health (C/A), Eli Lilly and Company, Celgene (SAB). The other authors indicated no financial relationships.
(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board
References
- 1.Schutz F, Stefanovic S, Mayer L et al. PD‐1/PD‐L1 pathway in breast cancer. Oncol Res Treat 2017;40:294–297. [DOI] [PubMed] [Google Scholar]
- 2.Bardhan K, Anagnostou T, Boussiotis VA. The PD1:PD‐L1/2 pathway from discovery to clinical implementation. Front Immunol 2016;7:550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Zou W, Wolchok JD, Chen L. PD‐L1 (B7‐H1) and PD‐1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 2016;8:328rv4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Savas P, Salgado R, Denkert C et al. Clinical relevance of host immunity in breast cancer: From TILs to the clinic. Nat Rev Clin Oncol 2016;13:228–241. [DOI] [PubMed] [Google Scholar]
- 5.Stanton SE, Adams S, Disis ML. Variation in the incidence and magnitude of tumor‐infiltrating lymphocytes in breast cancer subtypes: A systematic review. JAMA Oncol 2016;2:1354–1360. [DOI] [PubMed] [Google Scholar]
- 6.Schmid P, Adams S, Rugo HS et al. Atezolizumab and nab‐paclitaxel in advanced triple‐negative breast cancer. N Engl J Med 2018;379:2108–2121. [DOI] [PubMed] [Google Scholar]
- 7.Engel JB, Honig A, Kapp M et al. Mechanisms of tumor immune escape in triple‐negative breast cancers (TNBC) with and without mutated BRCA 1. Arch Gynecol Obstet 2014;289:141–147. [DOI] [PubMed] [Google Scholar]
- 8.Guan H, Wan Y, Lan J et al. PD‐L1 is a critical mediator of regulatory B cells and T cells in invasive breast cancer. Sci Rep 2016;6:35651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bertucci F, Finetti P, Colpaert C et al. PDL1 expression in inflammatory breast cancer is frequent and predicts for the pathological response to chemotherapy. Oncotarget 2015;6:13506–13519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sabatier R, Finetti P, Mamessier E et al. Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 2015;6:5449–5464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ghebeh H, Mohammed S, Al‐Omair A et al. The B7‐H1 (PD‐L1) T lymphocyte‐inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: Correlation with important high‐risk prognostic factors. Neoplasia 2006;8:190–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Uhercik M, Sanders AJ, Owen S et al. Clinical significance of PD1 and PDL1 in human breast cancer. Anticancer Res 2017;37:4249–4254. [DOI] [PubMed] [Google Scholar]
- 13.Thompson E, Taube JM, Elwood H et al. The immune microenvironment of breast ductal carcinoma in situ. Mod Pathol 2016;29:249–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Muenst S, Schaerli AR, Gao F et al. Expression of programmed death ligand 1 (PD‐L1) is associated with poor prognosis in human breast cancer. Breast Cancer Res Treat 2014;146:15–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Gatalica Z, Snyder C, Maney T et al. Programmed cell death 1 (PD‐1) and its ligand (PD‐L1) in common cancers and their correlation with molecular cancer type. Cancer Epidemiol Biomarkers Prev 2014;23:2965–2970. [DOI] [PubMed] [Google Scholar]
- 16.Arias‐Pulido H, Cimino‐Mathews A, Chaher N et al. The combined presence of CD20 + B cells and PD‐L1 + tumor‐infiltrating lymphocytes in inflammatory breast cancer is prognostic of improved patient outcome. Breast Cancer Res Treat 2018;171:273–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wimberly H, Brown JR, Schalper K et al. PD‐L1 expression correlates with tumor‐infiltrating lymphocytes and response to neoadjuvant chemotherapy in breast cancer. Cancer Immunol Res 2015;3:326–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zawlik I, Gablo N, Szymanska B et al. Immune checkpoints in aggressive breast cancer subtypes. Neoplasma 2016;63:768–773. [DOI] [PubMed] [Google Scholar]
- 19.Asano Y, Kashiwagi S, Goto W et al. Prediction of treatment responses to neoadjuvant chemotherapy in triple‐negative breast cancer by analysis of immune checkpoint protein expression. J Transl Med 2018;16:87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Dill EA, Gru AA, Atkins KA et al. PD‐L1 expression and intratumoral heterogeneity across breast cancer subtypes and stages: An assessment of 245 primary and 40 metastatic tumors. Am J Surg Pathol 2017;41:334–342. [DOI] [PubMed] [Google Scholar]
- 21.Beckers RK, Selinger CI, Vilain R et al. Programmed death ligand 1 expression in triple‐negative breast cancer is associated with tumour‐infiltrating lymphocytes and improved outcome. Histopathology 2016;69:25–34. [DOI] [PubMed] [Google Scholar]
- 22.Mardones M, Grosse D, Levin M et al. PD‐L1 expression in triple‐negative breast cancer (TNBC) is associated with improved outcomes. Presented at: San Antonio Breast Cancer Symposium; 2016; San Antonio, TX; P2‐04‐20.
- 23.Choi Y, Moon S, Jung S et al. Changes of tumor‐infiltrating lymphocytes and programmed death‐ligand1 positivity after neoadjuvant chemotherapy in patients with locally advanced breast cancer. Presented at: San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX; P2‐04‐24.
- 24.AiErken N, Shi HJ, Zhou Y et al. High PD‐L1 expression is closely associated with tumor‐infiltrating lymphocytes and leads to good clinical outcomes in Chinese triple negative breast cancer patients. Int J Biol Sci 2017;13:1172–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ali HR, Glont SE, Blows FM et al. PD‐L1 protein expression in breast cancer is rare, enriched in basal‐like tumours and associated with infiltrating lymphocytes. Ann Oncol 2015;26:1488–1493. [DOI] [PubMed] [Google Scholar]
- 26.Cerbelli B, Pernazza A, Botticelli A et al. PD‐L1 expression in TNBC: A predictive biomarker of response to neoadjuvant chemotherapy? Biomed Res Int 2017;2017:1750925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Denkert C, von Minckwitz G, Brase JC et al. Tumor‐infiltrating lymphocytes and response to neoadjuvant chemotherapy with or without carboplatin in human epidermal growth factor receptor 2–positive and triple‐negative primary breast cancers. J Clin Oncol 2015;33:983–991. [DOI] [PubMed] [Google Scholar]
- 28.Loi S, Giobbie‐Hurder A, Gombos A et al. Phase Ib/II study evaluating safety and efficacy of pembrolizumab and trastuzumab in patients with trastuzumab‐resistant HER2‐positive advanced breast cancer: results from the PANACEA study (IBCSG 45‐13/BIG 4‐13/KEYNOTE‐014). Presented at: 2017 San Antonio Breast Cancer Symposium; December 5–9, 2017; San Antonio, TX; Abstract GS2‐06.
- 29.Dieci M, Orvieto E, Tsetkova V et al. PD‐L1 expression and prognosis in triple negative breast cancer: An analysis of 265 patients treated with standard therapy for stage I‐III disease. Presented at: San Antonio Breast Cancer Symposium; December 4–8, 2018; San Antonio, TX.
- 30.Qin T, Zeng YD, Qin G et al. High PD‐L1 expression was associated with poor prognosis in 870 Chinese patients with breast cancer. Oncotarget 2015;6:33972–33981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kim HM, Lee J, Koo JS. Clinicopathological and prognostic significance of programmed death ligand‐1 expression in breast cancer: A meta‐analysis. BMC Cancer 2017;17:690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Zhang M, Sun H, Zhao S et al. Expression of PD‐L1 and prognosis in breast cancer: A meta‐analysis. Oncotarget 2017;8:31347–31354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bertucci F, Finetti P, Birnbaum D et al. The PD1/PDL1 axis, a promising therapeutic target in aggressive breast cancers. Oncoimmunology 2015;5:e1085148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Tolaney S, Barry W, Guo H et al. Immune profile of small HER2+ tumors in the APT trial. PD3‐01. Presented at: San Antonio Breast Cancer Symposium; 2017; San Antonio, TX.
- 35.Mittendorf EA, Philips AV, Meric‐Bernstam F et al. PD‐L1 expression in triple‐negative breast cancer. Cancer Immunol Res 2014;2:361–370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Cimino‐Mathews A, Thompson E, Taube JM et al. PD‐L1 (B7‐H1) expression and the immune tumor microenvironment in primary and metastatic breast carcinomas. Hum Pathol 2016;47:52–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Ghebeh H, Barhoush E, Tulbah A et al. FOXP3+ Tregs and B7‐H1+/PD‐1+ T lymphocytes co‐infiltrate the tumor tissues of high‐risk breast cancer patients: Implication for immunotherapy. BMC Cancer 2008;8:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Tawfik O, Kimler BF, Karnik T et al. Clinicopathological correlation of PD‐L1 expression in primary and metastatic breast cancer and infiltrating immune cells. Hum Pathol 2018;80:170–178. [DOI] [PubMed] [Google Scholar]
- 39.Buisseret L, Garaud S, de Wind A et al. Tumor‐infiltrating lymphocyte composition, organization and PD‐1/ PD‐L1 expression are linked in breast cancer. Oncoimmunology 2016;6:e1257452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang ZQ, Milne K, Derocher H et al. PD‐L1 and intratumoral immune response in breast cancer. Oncotarget 2017;8:51641–51651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kitano A, Ono M, Yoshida M et al. Tumour‐infiltrating lymphocytes are correlated with higher expression levels of PD‐1 and PD‐L1 in early breast cancer. ESMO Open 2017;2:e000150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Li M, Li A, Zhou S et al. Heterogeneity of PD‐L1 expression in primary tumors and paired lymph node metastases of triple negative breast cancer. BMC Cancer 2018;18:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lou J, Zhou Y, Huang J et al. Relationship between PD‐L1 expression and clinical characteristics in patients with breast invasive ductal carcinoma. Open Med (Wars) 2017;12:288–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Li F, Ren Y, Wang Z. Programmed death 1 ligand 1 expression in breast cancer and its association with patients' clinical parameters. J Cancer Res Ther 2018;14:150–154. [DOI] [PubMed] [Google Scholar]
- 45.Hou Y, Nitta H, Wei L et al. Evaluation of immune reaction and PD‐L1 expression using multiplex immunohistochemistry in HER2‐positive breast cancer: The association with response to anti‐HER2 neoadjuvant therapy. Clin Breast Cancer 2018;18:e237–e244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Ghebeh H, Tulbah A, Mohammed S et al. Expression of B7‐H1 in breast cancer patients is strongly associated with high proliferative Ki‐67‐expressing tumor cells. Int J Cancer 2007;121:751–758. [DOI] [PubMed] [Google Scholar]
- 48.Baptista MZ, Sarian LO, Derchain SF et al. Prognostic significance of PD‐L1 and PD‐L2 in breast cancer. Hum Pathol 2016;47:78–84. [DOI] [PubMed] [Google Scholar]
- 49.Tsang JY, Au WL, Lo KY et al. 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. [DOI] [PubMed] [Google Scholar]
- 50.Mori H, Kubo M, Yamaguchi R et al. The combination of PD‐L1 expression and decreased tumor‐infiltrating lymphocytes is associated with a poor prognosis in triple‐negative breast cancer. Oncotarget 2017;8:15584–15592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Okabe M, Toh U, Iwakuma N et al. Predictive factors of the tumor immunological microenvironment for long‐term follow‐up in early stage breast cancer. Cancer Sci 2017;108:81–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Acs B, Madaras L, Tokes AM et al. PD‐1, PD‐L1 and CTLA‐4 in pregnancy‐related ‐ and in early‐onset breast cancer: A comparative study. Breast 2017;35:69–77. [DOI] [PubMed] [Google Scholar]
- 53.Adams TA, Vail PJ, Ruiz A et al. Composite analysis of immunological and metabolic markers defines novel subtypes of triple negative breast cancer. Mod Pathol 2018;31:288–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bae SB, Cho HD, Oh MH 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 2016;19:242–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Li X, Wetherilt CS, Krishnamurti U et al. Stromal PD‐L1 expression is associated with better disease‐free survival in triple‐negative breast cancer. Am J Clin Pathol 2016;146:496–502. [DOI] [PubMed] [Google Scholar]
- 56.Botti G, Scognamiglio G, Marra L et al. Programmed death ligand 1 (PD‐L1) expression in primary angiosarcoma. J Cancer 2017;8:3166–3172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kim A, Lee SJ, Kim YK et al. Programmed death‐ligand 1 (PD‐L1) expression in tumour cell and tumour infiltrating lymphocytes of HER2‐positive breast cancer and its prognostic value. Sci Rep 2017;7:11671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Barrett MT, Lenkiewicz E, Malasi S et al. The association of genomic lesions and PD‐1/PD‐L1 expression in resected triple‐negative breast cancers. Breast Cancer Res 2018;20:71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schalper KA. PD‐L1 expression and tumor‐infiltrating lymphocytes: Revisiting the antitumor immune response potential in breast cancer. Oncoimmunology 2014;3:e29288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Taube JM, Klein A, Brahmer JR et al. Association of PD‐1, PD‐1 ligands, and other features of the tumor immune microenvironment with response to anti‐PD‐1 therapy. Clin Cancer Res 2014;20:5064–5074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Nanda R, Chow LQ, Dees EC et al. Pembrolizumab in patients with advanced triple‐negative breast cancer: Phase Ib KEYNOTE‐012 study. J Clin Oncol 2016;34:2460–2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Adams S, Schmid P, Rugo HS et al. Pembrolizumab monotherapy for previously treated metastatic triple‐negative breast cancer: Cohort A of the phase II KEYNOTE‐086 study. Ann Oncol 2019;30:397–404. [DOI] [PubMed] [Google Scholar]
- 63.Adams S, Loi S, Toppmeyer D et al. Pembrolizumab monotherapy for previously untreated, PD‐L1‐positive, metastatic triple‐negative breast cancer: Cohort B of the phase II KEYNOTE‐086 study. Ann Oncol 2019;30:405–411. [DOI] [PubMed] [Google Scholar]
- 64.Tolaney S, Kalinsky K, Kaklamani V et al. Phase 1b/2 study to evaluate eribulin mesylate in combination with pembrolizumab in patients with metastatic triple‐negative breast cancer. Presented at: San Antonio Breast Cancer Symposium; December 5–9, 2017; San Antonio, TX.
- 65.Schmid P, Cruz C, Braiteh F et al. Atezolizumab in metastatic TNBC (mTNBC): Long‐term clinical outcomes and biomarker analyses. Presented at: AACR Annual Meeting; April 1–5, 2017; Washington, DC; Abstract 2986.
- 66.Adams S, Robinson J, Hamilton D et al. Phase Ib trial of atezolizumab in combination with nab‐paclitaxel in patients with metastatic triple‐negative breast cancer (mTNBC). J Clin Oncol 2016;34(suppl 15):1009a. [Google Scholar]
- 67.Pohlmann P, Diamond J, Hamilton E et al. Atezolizumab (atezo) + nabpaclitaxel (nab‐pac) in metastatic triple‐negative breast cancer (mTNBC): 2‐year update from a ph Ib trial. Cancer Res 2018;78(suppl 13):CT028a. [Google Scholar]
- 68.Dirix LY, Takacs I, Jerusalem G et al. Avelumab, an anti‐PD‐L1 antibody, in patients with locally advanced or metastatic breast cancer: A phase 1b JAVELIN Solid Tumor study. Breast Cancer Res Treat 2018;167:671–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cortazar P, Zhang L, Untch M et al. Pathological complete response and long‐term clinical benefit in breast cancer: The CTNeoBC pooled analysis. Lancet 2014;384:164–172. [DOI] [PubMed] [Google Scholar]
- 70.Harberk N, Matthias A, Kates R et al. The role of immune and apoptosis markers for prediction of pCR in the WSG‐ADAPT HER2+/HR+ phase II trial evaluating 12‐weeks of neoadjuvant TDM1 ± endocrine therapy (ET) versus T + ET in HER2‐positive hormone‐receptor‐positive early breast cancer (EBC). Presented at: San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX; P1‐09‐05.
- 71.Waks A, Stover D, Barry W et al. The immune microenvironment in hormone receptor‐positive breast cancer patients and relationship to treatment outcome following preoperative chemotherapy plus bevacizumab. Presented at: San Antonio Breast Cancer Symposium; December 5–9, 2017; San Antonio, TX; PD6‐09.
- 72.Pelekanou V, Barlow WE, Nahleh ZA et al. Tumor‐infiltrating lymphocytes and PD‐L1 expression in pre‐ and posttreatment breast cancers in the SWOG S0800 phase II neoadjuvant chemotherapy trial. Mol Cancer Ther 2018;17:1324–1331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Nanda R, Liu M, Yau C et al. Pembrolizumab plus standard neoadjuvant therapy for high‐risk breast cancer: Results from I‐SPY‐2. Presented at: San Antonio Breast Cancer Symposium; December 5–9, 2017; San Antonio, TX.
- 74.Loibl S, Untch MM, Burchardi N et al. Randomized phase II neoadjuvant study (GeparNuevo) to investigate the addition of durvalumab to a taxane‐anthracycline containing chemotherapy in triple‐negative breast cancer. J Clin Oncol 2018;36(suppl 15):104a. [Google Scholar]
- 75.Schmid P, Park Y, Munoz‐Couselo E et al. Pembrolizumab + chemotherapy as neoadjuvant treatment for triple‐negative breast cancer: Preliminary results from KEYNOTE‐173. J Clin Oncol 2017;35(suppl 15):556a. [DOI] [PubMed] [Google Scholar]
- 76.Kok M, Horlings HM, Van de Vijver K et al. Adaptive phase II randomized non‐comparative trial of nivolumab after induction treatment in triple‐negative breast cancer: TONIC‐trial. Ann Oncol 2017;28(suppl 5):V605v649. [Google Scholar]
- 77.Karnik T, Kimler BF, Fan F et al. PD‐L1 in breast cancer: comparative analysis of 3 different antibodies. Hum Pathol 2018;72:28–34. [DOI] [PubMed] [Google Scholar]
- 78.Tsao MS, Kerr KM, Kockx M et al. PD‐L1 immunohistochemistry comparability study in real‐life clinical samples: Results of blueprint phase 2 project. J Thorac Oncol 2018;13:1302–1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rimm DL, Han G, Taube JM et al. A prospective, multi‐institutional, pathologist‐based assessment of 4 immunohistochemistry assays for PD‐L1 expression in non‐small cell lung cancer. JAMA Oncol 2017;3:1051–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Robertson S, Azizpour H, Smith K et al. Digital image analysis in breast pathology‐from image processing techniques to artificial intelligence. Transl Res 2018;194:19–35. [DOI] [PubMed] [Google Scholar]
- 81.Humphries MP, Hynes S, Bingham V et al. Automated tumour recognition and digital pathology scoring unravels new role for PD‐L1 in predicting good outcome in ER‐/HER2+ breast cancer. J Oncol 2018;2018:2937012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.KEYTRUDA (pembrolizumab) FDA label. Available at https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/125514s035lbl.pdf. Accessed June 2018.
- 83.Dieci MV, Tsvetkova V, Orvieto E et al. Immune characterization of breast cancer metastases: Prognostic implications. Breast Cancer Res 2018;20:62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Emens L, Loi S, Rugo H et al. IMpassion130: Efficacy in immune biomarker subgroup from the global, randomized, double‐blind, placebo‐controlled, phase III study of atezolizumab + nab‐paclitaxel in patients wuth treatment‐naive, locally advanced or metastatic triple‐negative breast cancer. Presented at: San Antonio Breast Cancer Symposium; December 5, 2018; San Antonio, TX; Oral Session 1.
- 85.Leduc C, Adam J, Louvet E et al. TPF induction chemotherapy increases PD‐L1 expression in tumour cells and immune cells in head and neck squamous cell carcinoma. ESMO Open 2018;3:e000257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Dosset M, Vargas TR, Lagrange A et al. PD‐1/PD‐L1 pathway: An adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology 2018;7:e1433981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Ng HY, Li J, Tao L et al. Chemotherapeutic treatments increase PD‐L1 expression in esophageal squamous cell carcinoma through EGFR/ERK activation. Transl Oncol 2018;11:1323–1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.McDaniel AS, Alva A, Zhan T et al. Expression of PDL1 (B7‐H1) before and after neoadjuvant chemotherapy in urothelial carcinoma. Eur Urol Focus 2016;1:265–268. [DOI] [PubMed] [Google Scholar]
- 89.Ling E, Yang C, Lin C et al. Priming PD‐L1 expression by chemotherapeutic agents in non‐small cell lung cancers. J Clin Oncol 2017;35(suppl 15)e20087e. [Google Scholar]
- 90.Parkes EE, Walker SM, Taggart LE et al. Activation of STING‐dependent innate immune signaling by S‐phase‐specific DNA damage in breast cancer. J Natl Cancer Inst 2016;109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Dieci MV, Criscitiello C, Goubar A et al. Prognostic value of tumor‐infiltrating lymphocytes on residual disease after primarychemotherapy for triple‐negative breast cancer: A retrospective multicenter study. Ann Oncol 2014;25:611–618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kok M, Voorwerk L, Horlings H et al. Adaptive phase II randomized trial of nivolumab after induction treatment in TNBC ‐ Tonic trial. Presented at: ASCO; 2018. [Google Scholar]
- 93.Adams S, Gatti‐Mays ME, Kalinsky K et al. Current landscape of immunotherapy in breast cancer: A review. JAMA Oncol 2019. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Hall JA, Salgado R, Lively T et al. A risk‐management approach for effective integration of biomarkers in clinical trials: Perspectives of an NCI, NCRI, and EORTC working group. Lancet Oncol 2014;15:e184–e193. [DOI] [PubMed] [Google Scholar]
- 95.Loi S, Adams S, Schmid P et al. Relationship between tumor infiltrating lymphocyte levels and response to pembrolizumab in metastatic triple‐negative breast cancer: Results from Keynote‐086 trial. Presented at: European Society of Medical Oncology (ESMO) 2017 Congress; September 8–12, 2017; Madrid, Spain; Abstract LBA13.
- 96.Hendry S, Salgado R, Gevaert T et al. Assessing tumor‐infiltrating lymphocytes in solid tumors: a practical review for pathologists and proposal for a standardized method from the International Immunooncology Biomarkers Working Group: Part 1: Assessing the host immune response, TILs in invasive breast carcinoma and ductal carcinoma in situ, metastatic tumor deposits and areas for further research. Adv Anat Pathol 2017;24:235–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Schott DS, Pizon M, Pachmann U et al. Sensitive detection of PD‐L1 expression on circulating epithelial tumor cells (CETCs) could be a potential biomarker to select patients for treatment with PD‐1/PD‐L1 inhibitors in early and metastatic solid tumors. Oncotarget 2017;8:72755–72772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Mazel M, Jacot W, Pantel K et al. Frequent expression of PD‐L1 on circulating breast cancer cells. Mol Oncol 2015;9:1773–1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Kawaguchi K, Suzuki E, Yamaguchi A et al. Altered expression of major immune regulatory molecules in peripheral blood immune cells associated with breast cancer. Breast Cancer 2017;24:111–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Moku B, Shepherd L, Ali S et al. High serum PD‐L1 predicts for increased overall survival to lapatinib vs trastuzumab in the phase 3 CCTG MA.31 trial. Presented at: 2018 San Antonio Breast Cancer Symposium; December 5, 2018; San Antonio, TX.
- 101.Pare L, Pascual T, Segui E et al. Association between PD1 mRNA and response to anti‐PD1 monotherapy across multiple cancer types. Ann Oncol 2018;29:2121–2128. [DOI] [PubMed] [Google Scholar]
- 102.Yi M, Yu S, Qin S et al. Gut microbiome modulates efficacy of immune checkpoint inhibitors. J Hematol Oncol 2018;11:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hugo W, Zaretsky JM, Sun L et al. Genomic and transcriptomic features of response to anti‐PD‐1 therapy in metastatic melanoma. Cell 2016;165:35–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Rizvi NA, Hellmann MD, Snyder A et al. Cancer immunology. Mutational landscape determines sensitivity to PD‐1 blockade in non‐small cell lung cancer. Science 2015;348:124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Snyder A, Makarov V, Merghoub T et al. Genetic basis for clinical response to CTLA‐4 blockade in melanoma. N Engl J Med 2014;371:2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Van Allen EM, Miao D, Schilling B et al. Genomic correlates of response to CTLA‐4 blockade in metastatic melanoma. Science 2015;350:207–211. [DOI] [PMC free article] [PubMed] [Google Scholar]