Abstract

Background

In 2020, pembrolizumab was approved as a therapy for triple-negative breast cancer (TNBC) with the companion diagnostic DAKO 22C3 programmed death ligand-1 (PD-L1) immunohistochemistry assay. The study aimed to determine the landscape of PD-L1 expression as detected by the DAKO 22C3 PD-L1 assay in breast cancer subtypes and compare the clinicopathologic and genomic characteristics of PD-L1 positive and negative TNBC.

Methods

PD-L1 expression using the DAKO 22C3 antibody was scored using a combined positive score (CPS) and positive status was defined as CPS ≥10. Comprehensive genomic profiling was performed using the FoundationOne CDx assay.

Results

Of the 396 BC patients stained with DAKO 22C3, the majority were HR+/HER2− and TNBC (42% and 36%, respectively). Median PD-L1 expression and frequency of CPS ≥10 was highest in TNBC cases (median: 7.5, 50% CPS ≥10) and lowest in the HR+/HER2− group (median: 1.0, 15.5% CPS ≥10) (P < .0001). A comparison of PD-L1 positive and PD-L1 negative TNBC demonstrated no significant differences in clinicopathologic or genomic characteristics. TNBC tissue samples from the breast did have an observed enrichment for PD-L1 positivity compared to TNBC tissue samples from a metastatic site (57% vs. 44%), but this was not statistically significant (P = .1766). In the HR+/HER2− group, genomic alterations in TP53, CREBBP, and CCNE1 were more prevalent and genomic loss of heterozygosity was higher in the PD-L1(+) group compared to the PD-L1(−) group.

Conclusions

The subtypes of breast cancer have distinct patterns of PD-L1 expression, supporting that further research of immunotherapies may include specific evaluation of optimum cutoffs for non-TNBC patients. In TNBC, PD-L1 positivity is not associated with other clinicopathologic or genomic features and should be integrated into future studies of immunotherapy efficacy.

Implications for Practice

This article presents a retrospective analysis of the patterns of PD-L1 expression using the DAKO 22C3 assay across all subtypes of breast carcinoma and describes the correlation of PD-L1 with other genomic features. The prevalence of PD-L1 expression varied by subtype. Within TNBC, PD-L1 positivity did not correlate with other clinicopathologic or genomic characteristics suggesting the independent nature of PD-L1 as a biomarker in TNBC. These conclusions support that further research on immunotherapies in non-TNBC patients may include specific evaluation of optimum PD-L1 scoring cutoffs.

Introduction

Despite advances in early detection and treatment, breast cancer remains the second leading cause of cancer-related death for females in the USA.1,2 In routine clinical practice, breast cancers are classified based on the expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor (HER2). In 15% of breast cancers, there is an absence of expression of these 3 receptors, so-called triple-negative breast cancer (TNBC).3,4 TNBC is characterized by aggressive pathologic features and high rates of distant recurrence. TNBC have the lowest 5-year survival rate among the receptor expression-defined subtypes.1,3,5 In addition, TNBC is more prevalent in African American women and contributes to the disparate outcomes for this population.6,7

Chemotherapy remains the standard of care first-line treatment for metastatic TNBC. Various anthracyclines, taxanes, and alkylating agents are commonly used in this setting.1 One of the challenges of treating BRCA wild-type TNBC is that it lacks targeted therapy options, so there is a strong dependence on cytotoxic agents. A new therapeutic option became available for patients in 2019 with the approval of the immune checkpoint inhibitor (ICPI) atezolizumab in combination with nab-paclitaxel for locally advanced or metastatic TNBC that is PD-L1 positive based on improved progression-free survival (PFS) observed in the IMpassion130 trial using the VENTANA SP142 PD-L1 assay.8-10 However, the indication was subsequently withdrawn in the USA in 2021 after additional studies did not show improvement in survival.11,12 During this time, another immunotherapy option became available for PD-L1 positive TNBC patients; pembrolizumab in combination with chemotherapy was approved in November, 2020, which was supported by the results of the KEYNOTE-355 study.13 Pembrolizumab and atezolizumab are ICPI targeting the programmed death receptor-1 (PD-1) and programmed death ligand-1 (PD-L1), respectively. PD-L1 is expressed by tumor-infiltrating leukocytes (TILs) in the tumor microenvironment and by tumor cells themselves as an adaptation to evade anti-tumor immune responses.14,15 Multiple studies showed that approximately half of TNBC express PD-L1, predominantly in TILs rather than tumor cells.1,15-18

In the KEYNOTE-355 trial (NCT02819518), 847 patients with untreated locally recurrent, inoperable, or metastatic TNBC received either pembrolizumab plus chemotherapy, or placebo plus chemotherapy with the physician’s choice of nab-paclitaxel, paclitaxel, or gemcitabine plus carboplatin for the chemotherapy regimen.19 Patients were assessed for PD-L1 expression by combined positive score (CPS) using the DAKO PD-L1 IHC 22C3 pharmDx assay.20 Among patients with a CPS of ≥10, median PFS was 9.7 months for patients receiving pembrolizumab versus 5.6 months for patients receiving placebo (hazard ratio [HR] 0.65, 95% CI, 0.49-0.86, P = .0012). In addition, PFS was evaluated at a CPS cutoff of ≥1 (7.6 vs. 5.6 months, HR 0.82, 95% CI, 0.74-0.90, P = .0014) and in the intent-to-treat population (7.5 vs. 5.6 months, HR 0.82, 95% CI, 0.69-0.97), but the threshold for significance based on the pre-specified statistical criteria was not met in either of these populations. These study results led to the FDA accelerated approval in November, 2020 of pembrolizumab plus chemotherapy for TNBC patients with a PD-L1 CPS ≥10. The benefit was confirmed in the survival analysis which reported overall survival (OS) of 23 months for pembrolizumab plus chemotherapy versus 16.3 months for placebo plus chemotherapy (HR 0.73, 95% CI, 0.55-0.95, P = .0093).21

Biomarkers predicting benefits from ICPI other than PD-L1 expression have also emerged in recent years. Both microsatellite instability-high (MSI-H) and tumor mutational burden ≥10 mutations per megabase (TMB-High) have also been approved as pan-solid tumor companion diagnostics for pembrolizumab.22,23 In addition, CD274 gene (that encodes PD-L1), copy number alterations, mutations, and rearrangements are emerging as candidate biomarkers for benefit from ICPI.24-27 This study aimed to determine the landscape of PD-L1 protein expression in breast cancer using the DAKO 22C3 assay and to compare PD-L1 positive and PD-L1 negative cancers by comprehensive genomic profiling (CGP) to determine if these populations differ in clinical or genomic characteristics.

Methods

Patient Selection

All patients meeting the following criteria were included in this study: (1) CGP performed using the FoundationOne®CDx assay between November, 2018 and October, 2021; (2) a diagnosis of breast carcinoma as reported by the submitting physician and accompanying pathology report; and (3) PD-L1 IHC was evaluated by DAKO 22C3 assay and scored with CPS. Thus, this US national cohort consisted of patients from multiple institutions who submitted breast carcinoma specimens for CGP to Foundation Medicine during the specified time frame. ER, PR, and HER2 expression status were extracted from the documentation submitted as a part of the routine course of clinical testing. In cases where HER2 expression was not reported, detection of ERBB2 amplification by CGP was used to classify the patient’s receptor status. Patients that were positive for ER and/or PR expression and negative for HER2 expression were classified as HR+/HER2−, patients that were positive for HER2 expression regardless of ER/PR status were classified as HER2+, and patients that were negative for all three biomarkers were classified as triple-negative breast carcinoma (TNBC).

PD-L1 IHC

PD-L1 IHC was performed in a Clinical Laboratory Improvement Amendments (CLIA)-certified, College of American Pathologists (CAP)-accredited laboratory (Foundation Medicine, Morrisville, NC, USA). PD-L1 IHC was tested using the DAKO PD-L1 IHC 22C3 pharmDx assay which uses the mouse monoclonal 22C3 anti-PD-L1 clone. The assay was performed according to the package insert with appropriate controls.20 Scoring was performed by American Board of Pathology board-certified pathologists specifically trained in PD-L1 22C3 CDx scoring for TNBC. Borderline cases underwent review by a 2nd pathologist to come to a consensus for scoring. Scores are reported as a CPS which is the number of PD-L1 staining cells (including tumor cells, lymphocytes, and macrophages) divided by the total number of viable tumor cells and then multiplied by 100. A sample was considered PD-L1(+) when CPS was ≥10.

Comprehensive Genomic Profiling

CGP was performed in a CLIA-certified, CAP-accredited laboratory (Foundation Medicine, Cambridge, MA, USA and Morrisville, NC, USA). Sequencing was performed using adaptor-ligation and hybrid capture from ≥50 ng DNA extracted from formalin-fixed paraffin-embedded samples as previously described.28 Exons from 324 genes and select introns from 36 genes were interrogated for all classes of genomic alterations (GA) (short variants, copy number changes, and rearrangements) by the assay.29CD274 amplification was defined as a CN of ploidy +4 for the purposes of this study. Tumor mutational burden was assessed on 0.80 megabases (Mb) of sequenced DNA and calculated based on the number of somatic base substitution or insertion/deletion alterations per Mb after excluding known somatic and deleterious mutations as previously described.30 MSI was determined on 95 loci as previously described.31

As research uses only (RUO), genomic loss of heterozygosity (gLOH) and genetic ancestry were calculated. gLOH was calculated by quantifying the loss of heterozygosity at over 3 500 SNPs but excluding whole chromosome arm losses (>90% arm loss), a method that was described and validated in the ARIEL2 trial for ovarian cancer.32,33 Scores are reported as a percentage, and specimens were required to have ≥30% tumor nuclei to meet quality control for inclusion in the analysis. Predominant genetic ancestry was assessed using a SNP-based approach.34 Using the data from the 1000 Genomes set, a trained and validated classifier was developed based on over 40 000 germline SNPs that could be identified in both the 1000 Genomes data and the profiling assay used here. Individuals could be classified into one of five possible predominant ancestry groups: African (AFR), European (EUR), Central and South American (AMR), South Asian (SAS), and East Asian (EAS).

Data Analysis

Statistics were evaluated using R version 3.6.1. Fisher’s exact test was used for categorical variables, and P-values were corrected using Benjamini–Hochberg corrections for multiple comparisons when appropriate. The Wilcox and the Kruskal–Wallis tests were used to assess continuous, non-parametric variables including TMB and age.

Results

Patient Characteristics

A total of 396 patients were identified for inclusion in this study. The HR+/HER2− subtype was the most prevalent (n = 168), followed by TNBC (n = 142), and HER2+ (n = 18). In addition, 68 cases had unknown receptor status. Nearly all patients were female (99.5%, 394/396), and they had a median age of 62 years (range 29-89) which was similar across all subtypes (Table 1). The majority of patients were of predominantly European ancestry (66.4%, 263/396), while the remainder were mostly of African and American ancestry (17.7% and 11.6%, respectively). Notably, there was a numerical enrichment of patients with African ancestry in the TNBC subtype compared to the HR+/HER2− subtype (23.2% vs. 14.3%, P = .055).

Table 1.

Summary of patient characteristics molecular subtype using DAKO 22C3 PD-L1 IHC assay.

HER2+ (N = 18) HR+/HER2− (N = 168) Triple-negative (N = 142) Unknown (N = 68) Total (N = 396) P-value
Age, median [IQR] years62.5 [55-68]63 [52-69]60 [51-69]62.5 [52-74]62.588
Sex (% female) 17 (94.4%) 168 (100.0%) 142 (100.0%) 67 (98.5%) 394 (99.5%).024
Genetic ancestry.027
 AFR 2 (11.1%) 24 (14.3%) 33 (23.2%) 11 (16.2%) 70 (17.7%)
 AMR 1 (5.6%) 21 (12.5%) 15 (10.6%) 9 (13.2%) 46 (11.6%)
 EAS 5 (27.8%) 4 (2.4%) 3 (2.1%) 1 (1.5%) 13 (3.3%)
 EUR 10 (55.6%) 117 (69.6%) 89 (62.7%) 47 (69.1%) 263 (66.4%)
 SAS 0 (0.0%) 2 (1.2%) 2 (1.4%) 0 (0.0%) 4 (1.0%)
MSI-H0 (0%)0 (0%)0 (0%)0 (0%)0 (0%)1
TMB, median [IQR] mut/MB4.4 [2.5-6.3]2.5 [1.3-5.0]2.5 [1.3-5.0]2.5 [1.3-6.3]2.5.064
PD-L1 CDx positive 8 (44.4%) 26 (15.5%) 71 (50.0%) 23 (33.8%) 128 (32.3%) <.001
PD-L1 CPS, median [IQR]5 [1-18]1 [0-2]7.5 [1-20]1 [0-10]1 [0-10] <.001
HER2+ (N = 18) HR+/HER2− (N = 168) Triple-negative (N = 142) Unknown (N = 68) Total (N = 396) P-value
Age, median [IQR] years62.5 [55-68]63 [52-69]60 [51-69]62.5 [52-74]62.588
Sex (% female) 17 (94.4%) 168 (100.0%) 142 (100.0%) 67 (98.5%) 394 (99.5%).024
Genetic ancestry.027
 AFR 2 (11.1%) 24 (14.3%) 33 (23.2%) 11 (16.2%) 70 (17.7%)
 AMR 1 (5.6%) 21 (12.5%) 15 (10.6%) 9 (13.2%) 46 (11.6%)
 EAS 5 (27.8%) 4 (2.4%) 3 (2.1%) 1 (1.5%) 13 (3.3%)
 EUR 10 (55.6%) 117 (69.6%) 89 (62.7%) 47 (69.1%) 263 (66.4%)
 SAS 0 (0.0%) 2 (1.2%) 2 (1.4%) 0 (0.0%) 4 (1.0%)
MSI-H0 (0%)0 (0%)0 (0%)0 (0%)0 (0%)1
TMB, median [IQR] mut/MB4.4 [2.5-6.3]2.5 [1.3-5.0]2.5 [1.3-5.0]2.5 [1.3-6.3]2.5.064
PD-L1 CDx positive 8 (44.4%) 26 (15.5%) 71 (50.0%) 23 (33.8%) 128 (32.3%) <.001
PD-L1 CPS, median [IQR]5 [1-18]1 [0-2]7.5 [1-20]1 [0-10]1 [0-10] <.001

Abbreviations: AFR, African; AMR, Central and South American; CPS, combined positive score; EUR, European; MSI-H, microsatellite instability-High; PD-L1, programmed death ligand-1; SAS, South Asian; TMB, tumor mutational burden.

Table 1.

Summary of patient characteristics molecular subtype using DAKO 22C3 PD-L1 IHC assay.

HER2+ (N = 18) HR+/HER2− (N = 168) Triple-negative (N = 142) Unknown (N = 68) Total (N = 396) P-value
Age, median [IQR] years62.5 [55-68]63 [52-69]60 [51-69]62.5 [52-74]62.588
Sex (% female) 17 (94.4%) 168 (100.0%) 142 (100.0%) 67 (98.5%) 394 (99.5%).024
Genetic ancestry.027
 AFR 2 (11.1%) 24 (14.3%) 33 (23.2%) 11 (16.2%) 70 (17.7%)
 AMR 1 (5.6%) 21 (12.5%) 15 (10.6%) 9 (13.2%) 46 (11.6%)
 EAS 5 (27.8%) 4 (2.4%) 3 (2.1%) 1 (1.5%) 13 (3.3%)
 EUR 10 (55.6%) 117 (69.6%) 89 (62.7%) 47 (69.1%) 263 (66.4%)
 SAS 0 (0.0%) 2 (1.2%) 2 (1.4%) 0 (0.0%) 4 (1.0%)
MSI-H0 (0%)0 (0%)0 (0%)0 (0%)0 (0%)1
TMB, median [IQR] mut/MB4.4 [2.5-6.3]2.5 [1.3-5.0]2.5 [1.3-5.0]2.5 [1.3-6.3]2.5.064
PD-L1 CDx positive 8 (44.4%) 26 (15.5%) 71 (50.0%) 23 (33.8%) 128 (32.3%) <.001
PD-L1 CPS, median [IQR]5 [1-18]1 [0-2]7.5 [1-20]1 [0-10]1 [0-10] <.001
HER2+ (N = 18) HR+/HER2− (N = 168) Triple-negative (N = 142) Unknown (N = 68) Total (N = 396) P-value
Age, median [IQR] years62.5 [55-68]63 [52-69]60 [51-69]62.5 [52-74]62.588
Sex (% female) 17 (94.4%) 168 (100.0%) 142 (100.0%) 67 (98.5%) 394 (99.5%).024
Genetic ancestry.027
 AFR 2 (11.1%) 24 (14.3%) 33 (23.2%) 11 (16.2%) 70 (17.7%)
 AMR 1 (5.6%) 21 (12.5%) 15 (10.6%) 9 (13.2%) 46 (11.6%)
 EAS 5 (27.8%) 4 (2.4%) 3 (2.1%) 1 (1.5%) 13 (3.3%)
 EUR 10 (55.6%) 117 (69.6%) 89 (62.7%) 47 (69.1%) 263 (66.4%)
 SAS 0 (0.0%) 2 (1.2%) 2 (1.4%) 0 (0.0%) 4 (1.0%)
MSI-H0 (0%)0 (0%)0 (0%)0 (0%)0 (0%)1
TMB, median [IQR] mut/MB4.4 [2.5-6.3]2.5 [1.3-5.0]2.5 [1.3-5.0]2.5 [1.3-6.3]2.5.064
PD-L1 CDx positive 8 (44.4%) 26 (15.5%) 71 (50.0%) 23 (33.8%) 128 (32.3%) <.001
PD-L1 CPS, median [IQR]5 [1-18]1 [0-2]7.5 [1-20]1 [0-10]1 [0-10] <.001

Abbreviations: AFR, African; AMR, Central and South American; CPS, combined positive score; EUR, European; MSI-H, microsatellite instability-High; PD-L1, programmed death ligand-1; SAS, South Asian; TMB, tumor mutational burden.

DAKO 22C3 PD-L1 Expression Patterns

The overall PD-L1 positivity (CPS ≥10) rate was 32%. Median CPS for PD-L1 expression varied significantly by subtype (Fig. 1A). TNBC had the highest median CPS at 7.5 (IQR: 1.0-20) while HR+/HER2− had the lowest at 1.0 (IQR: 0-2.0) (P < .0001). TNBC also had greater median PD-L1 expression than the group of unknown subtype (CPS 1.0, IQR: 0-10.0) (P = .015), and the unknown subtype had greater median PD-L1 expression than the HR+/HER2− subtype (CPS 1.0 IQR: 0-2.0) (P = .0009). A similar trend was also observed when PD-L1 expression was classified as positive defined as a CPS ≥10 (Fig. 1B). TNBC had the highest rate of positivity at 50.0% (71/142) followed by the HER2+ group at 44.4%, (8/18) and the HR+/HER2− subset had the lowest rate of positivity at 15.5% (26/168).

(A) PD-L1 immunohistochemistry (IHC) combined positive scores (CPS) using the Dako 22C3 assay for breast cancers by molecular subtype. (B) % of breast cancer cases with PD-L1 IHC CPS ≥10 by molecular subtype.
Figure 1.

(A) PD-L1 immunohistochemistry (IHC) combined positive scores (CPS) using the Dako 22C3 assay for breast cancers by molecular subtype. (B) % of breast cancer cases with PD-L1 IHC CPS ≥10 by molecular subtype.

PD-L1 Positive versus PD-L1 Negative TNBC Cohort Based on DAKO 22C3 CDx Assay

We performed a comparison of PD-L1(+) (n = 71) and PD-L1(−) (n = 71) TNBC to determine if the difference in PD-L1 positivity correlated with clinicopathological or genomic characteristics (Table 2). There was no difference in age, sex, or genetic ancestry between the 2 groups. TNBC tissue samples from the breast did have an observed enrichment for PD-L1 positivity compared to TNBC tissue samples from a metastatic site (57% vs. 44%), but this was not statistically significant (P = .1766). The most common specimen sites are breast, lymph node, liver, skin, and lung representing 44%, 20%, 8%, 6%, and 5% of the entire TNBC cohort, respectively. PD-L1 positivity rates varied by metastatic site, highest in the lung and lymph nodes and lowest in the liver (Table 3). Median TMB and the rate of TMB ≥10 mut/MB were similar, and no patients had MSI-H status (Table 2). The landscape of GA was also similar between the PD-L1(+) and the PD-L1(−) cohorts. There were no significant differences in the frequency of the top of 30 most frequently altered genes of either group (Table 4). GA in TP53, MYC, RAD21, PIK3CA, and PTEN was most prevalent in the overall cohort. The frequency of the targetable genes BRCA1 and BRCA2 were also similar (7% vs. 10% and 8% vs. 3%, respectively). Amplification of CD274 was seen in 6 PD-L1 (+) cases (8%) and only 1 PD-L1(−) case (1%) (P = .12).

Table 2.

Comparison of PDL1+ and PDL1− TNBC using DAKO 22C3 PD-L1 IHC assay.

PD-L1+ (N = 71)PD-L1− (N = 71)P-value
Age, median (range) years57 (31-89+)61 (32-88).124
Sex (% female)71 (100%)71 (100%)1
Specimen site, breast/metastasis36/35 (51%/49%)27/44 (38%/62%).1766
Genetic ancestry (%).6003
 AFR14 (20%)19 (27%)
 AMR7 (10%)8 (11%)
 EAS2 (3%)1 (1%)
 EUR46 (65%)43 (61%)
 SAS2 (3%)0 (0%)
TMB, median [IQR] mut/Mb2.5 [1.25-5]2.5 [2.5-5.0].764
TMB ≥10 mut/Mb6 (8%)3 (4%).4934
MSI-H001
gLOH21.0 [12.0-26.2]15.2 [7.4-26.1].1486
PD-L1+ (N = 71)PD-L1− (N = 71)P-value
Age, median (range) years57 (31-89+)61 (32-88).124
Sex (% female)71 (100%)71 (100%)1
Specimen site, breast/metastasis36/35 (51%/49%)27/44 (38%/62%).1766
Genetic ancestry (%).6003
 AFR14 (20%)19 (27%)
 AMR7 (10%)8 (11%)
 EAS2 (3%)1 (1%)
 EUR46 (65%)43 (61%)
 SAS2 (3%)0 (0%)
TMB, median [IQR] mut/Mb2.5 [1.25-5]2.5 [2.5-5.0].764
TMB ≥10 mut/Mb6 (8%)3 (4%).4934
MSI-H001
gLOH21.0 [12.0-26.2]15.2 [7.4-26.1].1486

Abbreviations: AFR, African; AMR, Central and South American; CPS, combined positive score; EUR, European; gLOH, genomic loss of heterozygosity; PD-L1, programmed death ligand-1; SAS, South Asian; MSI-H, microsatellite instability-High; TMB, tumor mutational burden.

Table 2.

Comparison of PDL1+ and PDL1− TNBC using DAKO 22C3 PD-L1 IHC assay.

PD-L1+ (N = 71)PD-L1− (N = 71)P-value
Age, median (range) years57 (31-89+)61 (32-88).124
Sex (% female)71 (100%)71 (100%)1
Specimen site, breast/metastasis36/35 (51%/49%)27/44 (38%/62%).1766
Genetic ancestry (%).6003
 AFR14 (20%)19 (27%)
 AMR7 (10%)8 (11%)
 EAS2 (3%)1 (1%)
 EUR46 (65%)43 (61%)
 SAS2 (3%)0 (0%)
TMB, median [IQR] mut/Mb2.5 [1.25-5]2.5 [2.5-5.0].764
TMB ≥10 mut/Mb6 (8%)3 (4%).4934
MSI-H001
gLOH21.0 [12.0-26.2]15.2 [7.4-26.1].1486
PD-L1+ (N = 71)PD-L1− (N = 71)P-value
Age, median (range) years57 (31-89+)61 (32-88).124
Sex (% female)71 (100%)71 (100%)1
Specimen site, breast/metastasis36/35 (51%/49%)27/44 (38%/62%).1766
Genetic ancestry (%).6003
 AFR14 (20%)19 (27%)
 AMR7 (10%)8 (11%)
 EAS2 (3%)1 (1%)
 EUR46 (65%)43 (61%)
 SAS2 (3%)0 (0%)
TMB, median [IQR] mut/Mb2.5 [1.25-5]2.5 [2.5-5.0].764
TMB ≥10 mut/Mb6 (8%)3 (4%).4934
MSI-H001
gLOH21.0 [12.0-26.2]15.2 [7.4-26.1].1486

Abbreviations: AFR, African; AMR, Central and South American; CPS, combined positive score; EUR, European; gLOH, genomic loss of heterozygosity; PD-L1, programmed death ligand-1; SAS, South Asian; MSI-H, microsatellite instability-High; TMB, tumor mutational burden.

Table 3.

Rate of PD-L1 positivity by specimen site of tested tissue in TNBC using DAKO 22C3 PD-L1 IHC assay.

Specimen sitePD-L1 +Total
Lung5 (71%)7
Lymph node19 (66%)29
Breast36 (57%)63
Abdominal wall1 (50%)2
Pleura1 (50%)2
Skin4 (44%)9
Chest wall 2 (40%)5
Soft tissue1 (33%)3
Liver2 (17%)12
Brain 0 (0%)3
Bone0 (0%)2
Spine 0 (0%)1
Pelvis 0 (0%)1
Trachea 0 (0%)1
Head and neck 0 (0%)1
Bone marrow 0 (0%)1
Specimen sitePD-L1 +Total
Lung5 (71%)7
Lymph node19 (66%)29
Breast36 (57%)63
Abdominal wall1 (50%)2
Pleura1 (50%)2
Skin4 (44%)9
Chest wall 2 (40%)5
Soft tissue1 (33%)3
Liver2 (17%)12
Brain 0 (0%)3
Bone0 (0%)2
Spine 0 (0%)1
Pelvis 0 (0%)1
Trachea 0 (0%)1
Head and neck 0 (0%)1
Bone marrow 0 (0%)1
Table 3.

Rate of PD-L1 positivity by specimen site of tested tissue in TNBC using DAKO 22C3 PD-L1 IHC assay.

Specimen sitePD-L1 +Total
Lung5 (71%)7
Lymph node19 (66%)29
Breast36 (57%)63
Abdominal wall1 (50%)2
Pleura1 (50%)2
Skin4 (44%)9
Chest wall 2 (40%)5
Soft tissue1 (33%)3
Liver2 (17%)12
Brain 0 (0%)3
Bone0 (0%)2
Spine 0 (0%)1
Pelvis 0 (0%)1
Trachea 0 (0%)1
Head and neck 0 (0%)1
Bone marrow 0 (0%)1
Specimen sitePD-L1 +Total
Lung5 (71%)7
Lymph node19 (66%)29
Breast36 (57%)63
Abdominal wall1 (50%)2
Pleura1 (50%)2
Skin4 (44%)9
Chest wall 2 (40%)5
Soft tissue1 (33%)3
Liver2 (17%)12
Brain 0 (0%)3
Bone0 (0%)2
Spine 0 (0%)1
Pelvis 0 (0%)1
Trachea 0 (0%)1
Head and neck 0 (0%)1
Bone marrow 0 (0%)1
Table 4.

Frequency of gene alterations in TNBC by PD-L1 status using DAKO 22C3 PD-L1 IHC assay.

GenePDL1+ (N = 71)PDL1− (N = 71)P value, corrected
TP5366 (93%)56 (79%).8394407
MYC24 (34%)22 (31%)1
RAD2118 (25%)18 (25%)1
PIK3CA19 (27%)16 (23%)1
PTEN11 (15%)18 (25%)1
RB110 (14%)11 (15%)1
CDKN2A10 (14%)7 (10%)1
FGFR19 (13%)7 (10%)1
WHSC1L18 (11%)8 (11%)1
LYN8 (11%)5 (7%)1
ZNF7037 (10%)6 (8%)1
CDKN2B7 (10%)6 (8%)1
CCNE16 (8%)6 (8%)1
CCND16 (8%)6 (8%)1
BRCA17 (10%)5 (7%)1
MCL16 (8%)6 (8%)1
NOTCH26 (8%)6 (8%)1
FGF195 (7%)6 (8%)1
FGF35 (7%)6 (8%)1
PIK3R13 (4%)8 (11%)1
FGF45 (7%)6 (8%)1
CREBBP6 (8%)4 (6%)1
KRAS3 (4%)6 (8%)1
AKT23 (4%)6 (8%)1
NF14 (6%)5 (7%)1
JAK26 (8%)3 (4%)1
FGF233 (4%)5 (7%)1
MTAP3 (4%)5 (7%)1
KDM5A4 (6%)4 (6%)1
NOTCH16 (8%)2 (3%)1
GenePDL1+ (N = 71)PDL1− (N = 71)P value, corrected
TP5366 (93%)56 (79%).8394407
MYC24 (34%)22 (31%)1
RAD2118 (25%)18 (25%)1
PIK3CA19 (27%)16 (23%)1
PTEN11 (15%)18 (25%)1
RB110 (14%)11 (15%)1
CDKN2A10 (14%)7 (10%)1
FGFR19 (13%)7 (10%)1
WHSC1L18 (11%)8 (11%)1
LYN8 (11%)5 (7%)1
ZNF7037 (10%)6 (8%)1
CDKN2B7 (10%)6 (8%)1
CCNE16 (8%)6 (8%)1
CCND16 (8%)6 (8%)1
BRCA17 (10%)5 (7%)1
MCL16 (8%)6 (8%)1
NOTCH26 (8%)6 (8%)1
FGF195 (7%)6 (8%)1
FGF35 (7%)6 (8%)1
PIK3R13 (4%)8 (11%)1
FGF45 (7%)6 (8%)1
CREBBP6 (8%)4 (6%)1
KRAS3 (4%)6 (8%)1
AKT23 (4%)6 (8%)1
NF14 (6%)5 (7%)1
JAK26 (8%)3 (4%)1
FGF233 (4%)5 (7%)1
MTAP3 (4%)5 (7%)1
KDM5A4 (6%)4 (6%)1
NOTCH16 (8%)2 (3%)1
Table 4.

Frequency of gene alterations in TNBC by PD-L1 status using DAKO 22C3 PD-L1 IHC assay.

GenePDL1+ (N = 71)PDL1− (N = 71)P value, corrected
TP5366 (93%)56 (79%).8394407
MYC24 (34%)22 (31%)1
RAD2118 (25%)18 (25%)1
PIK3CA19 (27%)16 (23%)1
PTEN11 (15%)18 (25%)1
RB110 (14%)11 (15%)1
CDKN2A10 (14%)7 (10%)1
FGFR19 (13%)7 (10%)1
WHSC1L18 (11%)8 (11%)1
LYN8 (11%)5 (7%)1
ZNF7037 (10%)6 (8%)1
CDKN2B7 (10%)6 (8%)1
CCNE16 (8%)6 (8%)1
CCND16 (8%)6 (8%)1
BRCA17 (10%)5 (7%)1
MCL16 (8%)6 (8%)1
NOTCH26 (8%)6 (8%)1
FGF195 (7%)6 (8%)1
FGF35 (7%)6 (8%)1
PIK3R13 (4%)8 (11%)1
FGF45 (7%)6 (8%)1
CREBBP6 (8%)4 (6%)1
KRAS3 (4%)6 (8%)1
AKT23 (4%)6 (8%)1
NF14 (6%)5 (7%)1
JAK26 (8%)3 (4%)1
FGF233 (4%)5 (7%)1
MTAP3 (4%)5 (7%)1
KDM5A4 (6%)4 (6%)1
NOTCH16 (8%)2 (3%)1
GenePDL1+ (N = 71)PDL1− (N = 71)P value, corrected
TP5366 (93%)56 (79%).8394407
MYC24 (34%)22 (31%)1
RAD2118 (25%)18 (25%)1
PIK3CA19 (27%)16 (23%)1
PTEN11 (15%)18 (25%)1
RB110 (14%)11 (15%)1
CDKN2A10 (14%)7 (10%)1
FGFR19 (13%)7 (10%)1
WHSC1L18 (11%)8 (11%)1
LYN8 (11%)5 (7%)1
ZNF7037 (10%)6 (8%)1
CDKN2B7 (10%)6 (8%)1
CCNE16 (8%)6 (8%)1
CCND16 (8%)6 (8%)1
BRCA17 (10%)5 (7%)1
MCL16 (8%)6 (8%)1
NOTCH26 (8%)6 (8%)1
FGF195 (7%)6 (8%)1
FGF35 (7%)6 (8%)1
PIK3R13 (4%)8 (11%)1
FGF45 (7%)6 (8%)1
CREBBP6 (8%)4 (6%)1
KRAS3 (4%)6 (8%)1
AKT23 (4%)6 (8%)1
NF14 (6%)5 (7%)1
JAK26 (8%)3 (4%)1
FGF233 (4%)5 (7%)1
MTAP3 (4%)5 (7%)1
KDM5A4 (6%)4 (6%)1
NOTCH16 (8%)2 (3%)1

PD-L1 Positive versus PD-L1 Negative HR+/HER2− Cohort Based on DAKO 22C3 CDx Assay

HR+/HER2− cases were also compared based on their PD-L1 expression. Patients were of similar median age and genetic ancestry (Supplementary Table S1). Most tested samples were from a metastatic site in both the PD-L1(−) and PD-L1(+) groups (69% and 70%, respectively). The most prevalently tested metastatic sites were the liver (n = 46), lymph node (n = 21), spine (n = 8), bone (n = 7), and lung (n = 6) with varying PD-L1 expression by the site (Supplementary Table S2). Both the rate of TMB ≥10 mut/MB and the median TMB were similar in the 2 cohorts. Median gLOH was significantly higher in the PD-L1(+) patients (17.3% vs. 9.3%, P < .001). In addition, differences in the frequency of several gene alterations among the top 30 altered genes most frequently found in the whole cohort were identified (Supplementary Table S3). TP53, CREBBP, and CCNE1 alterations were more prevalent in the PD-L1(+) group compared to the PD-L1(−) group (P = .0027, P = .010, and P = .005, respectively).

Discussion

In this study, we evaluated the landscape of PD-L1 IHC expression using DAKO 22C3 assay in a large cohort of breast carcinoma patients. This landscape demonstrated variable expression based on receptor subtype. TNBC had the highest median expression of PD-L1 and the greatest (approximately 50%) PD-L1 positivity rate based on the CPS ≥10 threshold per the companion diagnostic label. We also observed an approximately 15% PD-L1 positivity rate in HR+/HER2− cases. Prospective clinical trials will be needed to determine if these HR+/HER2− patients will benefit from ICPI therapy and what the optimal cut-off is to predict benefit.

We also saw that the PD-L1(+) and PD-L1(−)TNBC groups were similar in both clinicopathologic and genomic characteristics. Patients were of similar age distributions, and there was a similar distribution of genetic ancestry. We also examined the prevalence of other ICPI biomarkers. Median TMB was similar in both groups and there was no significant difference between the PD-L1(+) and PD-L1(−) groups in the percent of patients with TMB ≥10 mut/Mb. The landscape of concurrent alterations also showed no differences. These results demonstrate that these other characteristics cannot be used as surrogates to predict whether PD-L1 expression is likely for TNBC patients and that evaluation of PD-L1 by immunohistochemistry is necessary to properly evaluate this biomarker for patients in the clinic and clinical trials. Of note, these results differed from studies in non-small cell lung cancer, urothelial carcinoma, and cervical carcinoma where the authors found significant clinical and molecular differences between the PD-L1(+) and PD-L1(−) groups.35-37

We noticed that TNBC tissue samples from the breast had a higher rate of PD-L1 positivity compared to TNBC tissue samples from a metastatic site (57% vs. 44%), though it did not reach statistical significance. This is consistent with a prior study that also showed lower PD-L1 expression in metastatic lesions and substantial variability in PD-L1 positivity rates by metastatic site.38 Taken together, these observations suggest that the immune microenvironment of metastatic lesions may differ from that of primary tissues in TNBC as hypothesized by other studies of the immune landscape specific to metastases.39-42 This data have important implications for the specimen sites to submit for testing and clinical trial design.

In the HR+/HER2− cohort, PD-L1 positivity did correlate with other genomic differences. Median gLOH was higher in the PD-L1(+) subgroup, and alterations in TP53, CREBBP, and CCNE1 were also enriched in this population. The elevated gLOH in the PD-L1(+) could provide a rationale for possible therapy combinations of poly (ADP-ribose) polymerase (PARP) inhibitors and immunotherapy for HR+ disease. TP53 has been shown to be enriched in metastatic HR+ breast cancer, but the interaction between TP53 alterations, PD-L1 expression, and prognosis merits further study based on these results.43

Notably, nearly a quarter of TNBC patients in this study were of predominantly African ancestry regardless of PD-L1 expression. This finding is consistent with reports that the TNBC phenotype is enriched in African American patients.6,7 Similar PD-L1 expression and highly comparable immune microenvironment between TNBC in African American and Caucasian patients have also been shown earlier.41 It is regrettable that patients identifying as being of Black race made up only 7% of enrollees in the IMpassion130 trial and only 4% of those enrolled in Keynote-355.10,19 Given this particular unmet need of African American patients and the evidence that PD-L1 expression is prevalent in this population, future clinical trials assessing PD-L1 as a biomarker of response to ICPI should aim to increase enrollment of these patients.

One limitation of this study is the lack of treatment history for the included patients. It is unknown if patients received prior therapy including hormone therapy and chemotherapy which could possibly have altered the clonal evolution and genomics of the tumor. The study is retrospective in nature, and prospective clinical trials will be needed to test further hypotheses in the active treatment of patients.

Conclusion

The subtypes of breast cancer have distinct patterns of PD-L1 expression, and thus investigations into the efficacy of ICPI in non-TNBC patients may consider including analysis of optimum cutoffs for non-TNBC patients. In TNBC, PD-L1 positivity is not associated with other clinicopathologic or genomic features and thus should continue to be integrated into future studies of the immune microenvironment and immunotherapy efficacy. TNBC tissue samples from the breast did have an observed but not statistically significant enrichment for PD-L1 positivity compared to TNBC tissue samples from a metastatic site, a finding that merits future research in a larger cohort.

Abbreviations

    Abbreviations
     
  • AFR

    African

  •  
  • AMR

    Central and South American

  •  
  • CGP

    comprehensive genomic profiling

  •  
  • CPS

    combined positive score

  •  
  • EAS

    East Asian

  •  
  • ER

    estrogen receptor

  •  
  • EUR

    European

  •  
  • FFPE

    formalin-fixed paraffin-embedded

  •  
  • gLOH

    genomic loss of heterozygosity

  •  
  • HER2

    human epidermal growth factor receptor

  •  
  • ICPI

    immune checkpoint inhibitor

  •  
  • Mb

    megabases

  •  
  • MSI

    microsatellite instability

  •  
  • OS

    overall survival

  •  
  • PD-1

    programmed death receptor-1

  •  
  • PD-L1

    programmed death ligand-1

  •  
  • PR

    progesterone receptor

  •  
  • SAS

    South Asian

  •  
  • TILs

    tumor-infiltrating leukocytes

  •  
  • TMB

    tumor mutational burden

  •  
  • TMB-High

    tumor mutational burden ≥10 mutations per megabase

  •  
  • TNBC

    triple-negative breast cancer

Ethics Approval

Approval for this study was obtained from the Western Institutional Review Board Protocol No. 20152817.

Conflict of Interest

Natalie Danziger, Ethan S. Sokal, Ryon P. Graf, Matthew C. Hiemenz, Jake Maule, Vamsi Parimi, Jeffrey S. Ross, and Richard S.P. Huang are employees of Foundation Medicine, Inc., which is a wholly-owned subsidiary of Roche and receives stock from Roche. Carlo Palmieri: reported grant funding from Pfizer and Daiichi Sankyo and honoraria from Pfizer, Roche, Daiichi Sankyo, Novartis, Exact Sciences, Gilead, SeaGen, and Eli Lilly. Lajos Pusztai has received consulting fees and honoraria from Seagen, Pfizer, AstraZeneca, Merck, Novartis, Bristol-Myers Squibb, Pfizer, Genentech, Eisai, Pieris, Immunomedics, Clovis, Syndax, H3Bio, Radius Health, Personalis, Daiichi, and Natera, as well as institutional research funding from Seagen, AstraZeneca, Merck, Pfizer, and Bristol Myers Squibb.

Author Contributions

Conception/design: N.D, R.S.P.H. Provision of study material or patients: M.H., J.M., V.P., J.R., R.S.P.H. Collection and/or assembly of data: N.D. Data analysis and interpretation: All authors. Manuscript writing: All authors. Final approval of manuscript: All authors.

Data Availability

The data underlying this article are available in the article and in its online supplementary material.

References

1.

Bianchini
G
,
De Angelis
C
,
Licata
L
,
Gianni
L.
Treatment landscape of triple-negative breast cancer—expanded options, evolving needs
.
Nat Rev Clin Oncol.
2021
;
19
(
2
):
1
-
23
.

2.

Siegel
RL
,
Miller
KD
,
Jemal
A.
Cancer statistics, 2020
.
CA Cancer J Clin.
2020
;
70
(
1
):
7
-
30
. https://doi.org/10.3322/caac.21590

3.

Foulkes
WD
,
Smith
IE
,
Reis-Filho
JS.
Triple-negative breast cancer
.
N Engl J Med.
2010
;
363
:
1938
-
1948
. https://doi.org/10.1056/NEJMra1001389

4.

Boyle
P.
Triple-negative breast cancer: epidemiological considerations and recommendations
.
Ann Oncol.
2012
;
23
:
vi7
-
v12
. https://doi.org/10.1093/annonc/mds187

5.

Dent
R
,
Trudeau
M
,
Pritchard
KI
, et al. .
Triple-negative breast cancer: clinical features and patterns of recurrence
.
Clin Cancer Res.
2007
;
13
(
15
):
4429
-
4434
. https://doi.org/10.1158/1078-0432.CCR-06-3045

6.

Newman
LA
,
Kaljee
LM.
Health disparities and triple-negative breast cancer in African American women: a review
.
JAMA Surg
.
2017
;
152
(
5
):
485
-
493
. https://doi.org/10.1001/jamasurg.2017.0005

7.

Huo
D
,
Hu
H
,
Rhie
SK
, et al. .
Comparison of breast cancer molecular features and survival by African and European ancestry in the cancer genome atlas
.
JAMA Oncol
.
2017
;
3
(
12
):
1654
-
1662
. https://doi.org/10.1001/jamaoncol.2017.0595

8.

Genentech Inc.
TECENTRIQ (atezolizumab) Prescribing Information
. Accessed April 2022. https://www.gene.com/download/pdf/tecentriq_prescribing.pdf.

9.

Research C for DE and.
FDA approves atezolizumab for PD-L1 positive unresectable locally advanced or metastatic triple-negative breast cancer.
FDA
. Accessed
December 3, 2019
. Published online March 18,
2019
. http://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-atezolizumab-pd-l1-positive-unresectable-locally-advanced-or-metastatic-triple-negative

10.

Schmid
P
,
Adams
S
,
Rugo
HS
, et al. .
Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer
.
N Engl J Med.
2018
;
379
(
8
):
2108
-
2121
. https://doi.org/10.1056/NEJMoa1809615

11.

Miles
D
,
Gligorov
J
,
André
F
, et al. .
IMpassion131 investigators
.
Primary results from IMpassion131, a double-blind, placebo-controlled, randomised phase III trial of first-line paclitaxel with or without atezolizumab for unresectable locally advanced/metastatic triple-negative breast cancer
.
Ann Oncol.
2021
;
32
(
8
):
994
-
1004
. https://doi.org/10.1016/j.annonc.2021.05.801

12.

Genentech: Press Releases.
Accessed
December 20, 2021
. Friday, Aug 27,
2021
. https://www.gene.com/media/press-releases/14927/2021-08-27/genentech-provides-update-on-tecentriq-u

13.

Research C for DE and FDA grants accelerated approval to pembrolizumab for locally recurrent unresectable or metastatic triple-negative breast cancer.
FDA
. Accessed
December 16, 2021
. Published online June 11,
2021
. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-locally-recurrent-unresectable-or-metastatic-triple

14.

Ohaegbulam
KC
,
Assal
A
,
Lazar-Molnar
E
,
Yao
Y
,
Zang
X.
Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway
.
Trends Mol Med.
2015
;
21
(
1
):
24
-
33
. https://doi.org/10.1016/j.molmed.2014.10.009

15.

Han
Y
,
Liu
D
,
Li
L.
PD-1/PD-L1 pathway: current researches in cancer
.
Am J Cancer Res
.
2020
;
10
(
3
):
727
-
742
.

16.

Huang
RSP
,
Haberberger
J
,
Severson
E
, et al. .
A pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases
.
Mod Pathol.
2020
;
34
(
2
):
1
-
12
. https://doi.org/10.1038/s41379-020-00664-y

17.

Huang
RSP
,
Li
X
,
Haberberger
J
, et al. .
Biomarkers in breast cancer: an integrated analysis of comprehensive genomic profiling and PD-L1 immunohistochemistry biomarkers in 312 patients with breast cancer.
The Oncologist.
https://doi.org/10.1634/theoncologist.2020-0449

18.

Huang
RSP
,
Haberberger
J
,
McGregor
K
, et al. .
Clinicopathologic and genomic landscape of breast carcinoma brain metastases
.
The Oncologist
.
2021
;
26
(
10
):
835
-
844
. https://doi.org/10.1002/onco.13855

19.

Cortes
J
,
Cescon
DW
,
Rugo
HS
, et al. .
Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial
.
The Lancet
.
2020
;
396
(
10265
):
1817
-
1828
. https://doi.org/10.1016/S0140-6736(20)32531-9

20.

Dako PD-L1 IHC 22C3 pharmDx. Package insert
. Accessed
December 21, 2021
. Published online
2019
. https://www.accessdata.fda.gov/cdrh_docs/pdf15/P150013S014C.pdf

21.

Rugo
HS
,
Cortes
J
,
Cescon
DW
, et al. .
LBA16 - KEYNOTE-355: final results from a randomized, double-blind phase III study of first-line pembrolizumab + chemotherapy vs placebo + chemotherapy for metastatic TNBC
.
Ann Oncol.
2021
;
32
.

22.

Research C for DE and.
FDA approves pembrolizumab for adults and children with TMB-H solid tumors.
FDA
. Accessed
August 13, 2021
. Published online June 17,
2020
. https://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-pembrolizumab-adults-and-children-tmb-h-solid-tumors

23.

Research C for DE and.
FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication.
FDA
. Accessed
August 13, 2021
. Published online February 9,
2019
. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-pembrolizumab-first-tissuesite-agnostic-indication

24.

Huang
RSP
,
Decker
B
,
Murugesan
K
, et al. .
Pan-cancer analysis of CD274 (PD-L1) mutations in 314,631 patient samples and subset correlation with PD-L1 protein expression
.
J ImmunoTher Cancer.
2021
;
9
(
6
):
e002558
. https://doi.org/10.1136/jitc-2021-002558

25.

Huang
RSP
,
Murugesan
K
,
Montesion
M
, et al. .
Pan-cancer landscape of CD274 (PD-L1) copy number changes in 244 584 patient samples and the correlation with PD-L1 protein expression
.
J ImmunoTher Cancer.
2021
;
9
(
5
):
e002680
. https://doi.org/10.1136/jitc-2021-002680

26.

Kelly
AD
,
Murugesan
K
,
Kuang
Z
, et al. .
Pan-cancer landscape of CD274 (PD-L1) rearrangements in 283,050 patient samples, its correlation with PD-L1 protein expression, and immunotherapy response
.
J ImmunoTher Cancer.
2021
;
9
(
11
):
e003550e003550
. https://doi.org/10.1136/jitc-2021-003550

27.

Bachelot
T
,
Filleron
T
,
Bieche
I
, et al. .
Durvalumab compared to maintenance chemotherapy in metastatic breast cancer: the randomized phase II SAFIR02-BREAST IMMUNO trial
.
Nat Med.
2021
;
27
(
2
):
250
-
255
. https://doi.org/10.1038/s41591-020-01189-2

28.

Frampton
GM
,
Fichtenholtz
A
,
Otto
GA
, et al. .
Development and validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing
.
Nat Biotechnol.
2013
;
31
(
11
):
1023
-
1031
. https://doi.org/10.1038/nbt.2696

29.

Lipson
D
,
Capelletti
M
,
Yelensky
R
, et al. .
Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies
.
Nat Med.
2012
;
18
(
3
):
382
-
384
. https://doi.org/10.1038/nm.2673

30.

Chalmers
ZR
,
Connelly
CF
,
Fabrizio
D
, et al. .
Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden
.
Genome Med.
2017
;
9
(
1
):
1
-
14
. https://doi.org/10.1186/s13073-017-0424-2

31.

Trabucco
SE
,
Gowen
K
,
Maund
SL
, et al. .
A novel next-generation sequencing approach to detecting microsatellite instability and pan-tumor characterization of 1000 microsatellite instability–high cases in 67,000 patient samples
.
J Mol Diagn.
2019
;
21
(
6
):
1053
-
1066
. https://doi.org/10.1016/j.jmoldx.2019.06.011

32.

Swisher
EM
,
Lin
KK
,
Oza
AM
, et al. .
Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial
.
Lancet Oncol.
2017
;
18
(
1
):
75
-
87
. https://doi.org/10.1016/S1470-2045(16)30559-9

33.

Coleman
RL
,
Oza
AM
,
Lorusso
D
, et al. .
Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial
.
The Lancet
.
2017
;
390
(
10106
):
1949
-
1961
. https://doi.org/10.1016/S0140-6736(17)32440-6

34.

Connelly
CF
,
Carrot-Zhang
J
,
Stephens
PJ
,
Frampton
GM.
Abstract 1227: somatic genome alterations in cancer as compared to inferred patient ancestry
.
Cancer Res.
2018
;
78
(
Suppl 13
):
1227
-
1227
. https://doi.org/10.1158/1538-7445.am2018-1227

35.

Huang
RSP
,
Severson
E
,
Haberberger
J
, et al. .
Landscape of biomarkers in non-small cell lung cancer using comprehensive genomic profiling and PD-L1 immunohistochemistry
.
Pathol Oncol Res.
2021
;
27
. https://doi.org/10.3389/pore.2021.592997

36.

Huang
RSP
,
Haberberger
J
,
Harries
L
, et al. .
Clinicopathologic and genomic characterization of PD‐L1 positive urothelial carcinomas
.
The Oncologist
.
2021
;
26
(
5
):
375
-
382
. https://doi.org/10.1002/onco.13753

37.

Huang
RSP
,
Haberberger
J
,
Murugesan
K
, et al. .
Clinicopathologic and genomic characterization of PD-L1-positive uterine cervical carcinoma
.
Mod Pathol.
2021
:
1
-
9
. https://doi.org/10.1038/s41379-021-00780-3

38.

Rozenblit
M
,
Huang
R
,
Danziger
N
, et al. .
Comparison of PD-L1 protein expression between primary tumors and metastatic lesions in triple-negative breast cancers
.
J ImmunoTher Cancer.
2020
;
8
(
2
):
e001558
. https://doi.org/10.1136/jitc-2020-001558

39.

Li
JJ
,
Tsang
JY
,
Tse
GM.
Tumor microenvironment in breast cancer—updates on therapeutic implications and pathologic assessment
.
Cancers
.
2021
;
13
(
16
):
42334233
. https://doi.org/10.3390/cancers13164233

40.

Liu
C
,
Wang
M
,
Xu
C
, et al. .
Immune checkpoint inhibitor therapy for bone metastases: specific microenvironment and current situation
.
J Immunol Res
.
2021
;
2021
:
8970173
. https://doi.org/10.1155/2021/8970173

41.

García-Mulero
S
,
Alonso
MH
,
Pardo
J
, et al. .
Lung metastases share common immune features regardless of primary tumor origin
.
J ImmunoTher Cancer.
2020
;
8
(
1
):
e000491
. https://doi.org/10.1136/jitc-2019-000491

42.

Moutafi
MK
,
Tao
W
,
Huang
R
, et al. .
Comparison of programmed death-ligand 1 protein expression between primary and metastatic lesions in patients with lung cancer
.
J ImmunoTher Cancer.
2021
;
9
(
4
):
e002230
. https://doi.org/10.1136/jitc-2020-002230

43.

Bertucci
F
,
Ng
CKY
,
Patsouris
A
, et al. .
Genomic characterization of metastatic breast cancers
.
Nature.
2019
;
569
:
560
-
564
. https://doi.org/10.1038/s41586-019-1056-z

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.