Purpose:

HER3 belongs to a family of receptor tyrosine kinases with oncogenic properties and is targeted by a variety of novel anticancer agents. There is a huge unmet medical need for systemic treatment options in patients with brain metastases (BM). Therefore, we aimed to investigate HER3 expression in BM of breast (BCa) and non–small cell lung cancer (NSCLC) as the basis for future clinical trial design.

Experimental Design:

We analyzed 180 BM samples of breast cancer or NSCLC and 47 corresponding NSCLC extracranial tissue. IHC was performed to evaluate protein expression of HER3, and immune cells based on CD3, CD8, and CD68. To identify dysregulated pathways based on differential DNA methylation patterns, we used Infinium MethylationEPIC microarrays.

Results:

A total of 99/132 (75.0%) of BCa-BM and 35/48 (72.9%) of NSCLC-BM presented with HER3 expression. Among breast cancer, HER2-positive and HER2-low BM showed significantly higher rates of HER3 coexpression than HER2-negative BM (87.1%/85.7% vs. 61.0%, P = 0.004). Among NSCLC, HER3 was more abundantly expressed in BM than in matched extracranial samples (72.9% vs. 41.3%, P = 0.003). No correlation of HER3 expression and intratumoral immune cell density was observed. HER3 expression did not correlate with overall survival from BM diagnosis. Methylation signatures differed according to HER3 status in BCa-BM samples. Pathway analysis revealed subtype-specific differences, such as TrkB and Wnt signaling pathways dysregulated in HER2-positive and triple-negative breast cancer BM, respectively.

Conclusions:

HER3 is highly abundant in BM of breast cancer and NSCLC. Given the promising results of antibody–drug conjugates in extracranial disease, BM-specific trials that target HER3 are warranted.

See related commentary by Kabraji and Lin, p. 2961

Translational Relevance

The development of brain metastases (BM) is a tremendous clinical challenge and the ultimate cause of death in many patients with cancer. Effective strategies to manage BM are an unmet clinical need, and novel approaches to treat BM are dearly needed. Recent advances and promising response rates in patients with BM treated with antibody–drug conjugates against the HER2 receptor encourage further studies on potential novel therapeutic targets. Here, we show that a high proportion of patients with BM of breast and non–small cell lung cancer (NSCLC) express the HER3 receptor in therapeutically relevant amounts. Importantly, the majority of HER2-negative tumors were still positive for HER3. Furthermore, we show that BM of NSCLC express HER3 more frequently compared with their matched extracranial tumors. Taken together, these findings suggest the clinical use of HER3 as a potential promising therapeutic target in BM of breast cancer and NSCLC.

HER3/ErbB3 belongs to a family of receptor tyrosine kinases with oncogenic properties (1). In contrast to other known family members, such as HER2 (ErbB2), HER3 has an impaired kinase activity (2). Therefore, heterodimerization with other HER members, preferentially with HER2, are necessary for its own oncogenic potential (3). HER3 overexpression has been observed in several tumor types such as breast, ovarian, prostate, gastric, melanoma, colon, lung, and head and neck cancer (4–11). A meta-analysis performed in 2012 by Ocana and colleagues evaluated 12 studies with a median percentage of HER3 overexpression in cancers of 42.2% (17.0–69.7; ref. 12). Solid tumors with the highest frequency of HER3 overexpression included colorectal (69.0%), melanoma (65.0%), and gastric (59.0%) cancer. Moreover, increased HER3 expression was postulated as a potential cause for therapy resistance against other HER-targeted therapies (13, 14). To overcome therapeutic failure, approaches to directly target HER3 are emerging including abrogating the kinase activity of its dimerization partners by using small-molecule inhibitors or by directly targeting its extracellular domain with mAbs (15, 16).

HER3 is of particular interest in the context of brain metastases (BM) formation as HER3 signaling has been shown to be involved in metastatic spread and colonization to the brain (17, 18). BM are a major clinical challenge that affect approximately 20% of all patients with cancer. Higher incidences occur in certain tumor types like EGFR-mutated non–small cell lung cancer (NSCLC) or triple-negative (TNBC) or HER2-positive (HER2+) breast cancer (BCa), where BM tend to develop in up to 40% of cases (19). Treatment options are still limited, focusing on radiotherapy and, if applicable, surgical resection of the tumor mass (20). While initially effective, these interventions are associated with frequent neurologic impairment, hence significantly reducing quality of life, though rarely capable to improve prognosis beyond 2 years. Therefore, additional therapeutic options including systemic treatment are warranted to improve prognosis.

Molecular targets for personalized treatment approaches are of particular interest for BM treatment. Indeed, up to 50% of BM were shown to harbor potentially clinically relevant molecular alterations, which were not detected in their matched primary tumors (21). Recently, targeted treatments including antibody–drug conjugates (ADC) resulted in promising clinical activity in patients with BM (22–24). Furthermore, patritumab-deruxtecan (HER3-DXd), a novel ADC against HER3, demonstrated activity and safety in a phase I clinical trial including patients with metastatic NSCLC harboring an EGFR-activating mutation (25). In addition, early results of a phase I/II clinical trial in patients with breast cancer of any subtype showed promising efficacy of HER3-DXd as well (26).

In the current study, we aimed to investigate the frequency of HER3 overexpression in BM of patients with breast cancer and NSCLC as those entities most frequently developing BM. Considering the influence of immune cells on the efficacy of mAb-based therapies, we also evaluated the tumor microenvironment for its immune cell composition within the BM. We further used methylation profiles of HER3-positive (HER3+) and -negative (HER3) specimens to identify relevant dysregulated pathways and gain further biological insight for future trial designs.

Patient cohort

Patients diagnosed with BM from breast cancer or NSCLC between 1990 and 2020 at the Medical University of Vienna (Vienna, Austria) were identified. Formalin-fixed and paraffin-embedded (FFPE) material available for scientific purposes was restored from the pathology and neuropathology department, respectively. Clinical data were collected retrospectively using available medical records. This study was approved by the Ethics Committees of the Medical University of Vienna (approval no. 1975/2021) in accordance with the Declaration of Helsinki. The need for informed consent was waived by the competent regulatory authorities due to the retrospective nature of the study.

IHC

A total of 4-μm-thick shavings were prepared from FFPE tissue blocks of BM and primary tumor samples. IHC staining was performed on a Ventana Benchmark ULTRA machine (Roche Diagnostics) using the following antibodies: HER3 (Cell Signaling Technology, #12708, RRID:AB_2721919, 1:100), HER2 [PATHWAY HER2 (4B5), Roche catalog no. 790-4493, RRID:AB_2921204, ready-to-use (RTU)], Estrogen Receptor [CONFIRM ER (SP1), Roche, catalog no. 790-4324, RRID:AB_2857956, RTU], Progesteron Receptor [CONFIRM PR (1E2), Ventana Medical Systems catalog no. 790-4296, RRID:AB_2335976, RTU], CD3 (Lab Vision catalog no. RM-9107-S, RRID:AB_149922, 1:200), CD8 (Agilent catalog no. M7103, RRID:AB_2075537, 1:100), CD68 (Agilent catalog no. M0814, RRID:AB_2314148, 1:500). Heat-induced antigen retrieval was achieved by incubation with the Tris-EDTA–based Cell Conditioning 1 (CC1) buffer (Roche Diagnostics) for all antibodies. HER3 signal intensity was amplified by the application of the Ventana Amplification Kit (Roche). Protein expression was visualized using the UltraView DAB IHC Detection Kit (Ventana) and counterstained with Hematoxylin and Bluing Reagent (Ventana). A protocol including specific conditions for each antibody is provided in Supplementary Table S1. Specificity of the HER3 antibody was further validated by staining two breast cancer cell lines with known HER3 expression levels (Supplementary Fig. S1A).

Evaluation of IHC staining

Slides stained for immune cell markers were digitalized on a slide scanner for semiquantitative analysis using the Definiens Tissue Studio software v3.0 (Definiens Inc., RRID:SCR_023001) as described previously (27). Hormone receptors and HER2 expression were reevaluated in brain metastasis specimens by microscopic evaluation using established criteria (28, 29). Briefly, nuclear staining of estrogen receptor (ER) and progesteron receptor (PR) in at least 1% of tumor cells was considered hormone receptor–positive. Luminal tumors had to show positive expression for either ER and/or PR but they were not further grouped into A and B subtypes. HER2 and HER3 stainings were only considered if complete, strong membranous staining pattern of at least 10% tumor cells was present. HER2 expression was further graded into IHC 0, 1+, 2+, and 3+ while only IHC3+ and IHC2+/ISH+ tumors were termed HER2+, IHC1+ and IHC2+/ISH− as HER2-low, and IHC0 as HER2-negative (HER2). We did not perform ISH analysis in IHC2+ BM samples instead adopted subtyping from primary samples. HER3 staining was graded according to complete, membranous staining intensity into negative, low and high based on staining intensity following aforementioned HER2 criteria (Fig. 1A). For analysis, HER3-low and -high tumors were combined into HER3+. Two independent observers evaluated the IHC-stained slides.

Figure 1.

Frequent overexpression of HER3 in BM of breast and lung cancer. A, Representative microscopic images of BM stained for HER3 by IHC grouped into negative (left), low (middle), and high (right) expression. Scale bar, 50 μm. B, The proportion of HER3 expression in breast (left) and lung (right) cancer BM. C, Distribution of HER3 expression in the three subtypes of breast cancer: luminal/hormone receptor–positive (left), HER2+ (middle), and TNBC (right). D, Correlation between HER2 IHC scoring and HER3 expression in BM of patients with breast cancer (n = 132). E, A sankey diagram demonstrating the transformation of HER3 expression between matched primary lung cancer and corresponding BM (n = 47).

Figure 1.

Frequent overexpression of HER3 in BM of breast and lung cancer. A, Representative microscopic images of BM stained for HER3 by IHC grouped into negative (left), low (middle), and high (right) expression. Scale bar, 50 μm. B, The proportion of HER3 expression in breast (left) and lung (right) cancer BM. C, Distribution of HER3 expression in the three subtypes of breast cancer: luminal/hormone receptor–positive (left), HER2+ (middle), and TNBC (right). D, Correlation between HER2 IHC scoring and HER3 expression in BM of patients with breast cancer (n = 132). E, A sankey diagram demonstrating the transformation of HER3 expression between matched primary lung cancer and corresponding BM (n = 47).

Close modal

DNA methylation

Genomic DNA was isolated from tumor cell–enriched (≥70%) FFPE blocks (n = 71) using the Maxwell FFPE Plus Kit (Promega) following the manufacture's instruction. Isolated DNA was quantified on a Qubit 2.0 Fluorometer (Thermo Fisher Scientific) using the associated assay kits and 500 ng were subsequently bisulfite converted using the EpiTect Fast Kit (Qiagen) according to the manufacturer's manual. Before each sample was applied to the Infinium EPIC Methylation BeadChip (Illumina) protocol, the Infinium HD FFPE Restore Kit (Illumina) was performed to improve sample quality. Processed microarray chips were scanned on an iScan device (Illumina, RRID:SCR_016388) to obtain raw fluorescence intensity (.idat) files.

Bioinformatical analysis

Intensity data were imported into R using the RnBeads package (v2.12.2, RRID:SCR_010958) following the suggested pipeline which performs assay quality validation, probe filtering, and normalization steps as well as differential methylation analysis (30). Probes on sex chromosomes were excluded from subsequent analysis as well as SNP-enriched and cross-reactive probes. The Greedycut algorithm was used to remove probes and samples of highest impurity based on a detection P value above 0.05. The SWAN algorithm was chosen to normalize microarray data (31). Differentially methylated sites were selected on the basis of calculated rank cutoffs by RnBeads. P values on the site level were computed using the limma method. Dimensional reduction of the methylation data was calculated and visualized with the umap package for R. Heatmaps were generated using the ClustVis online tool and pathway enrichment analyses was performed utilizing the graphical web application ShinyGO v.0.76.3 (32, 33).

Statistical analysis

Differences between two groups were tested using a two-tailed Mann–Whitney test while differences between multiple (≥3) groups were tested using Kruskal–Wallis test with Dunn post hoc test for multiple comparison. The independence of categorical variables was tested by either χ2 or Fisher exact test depending on sample size. An adjusted P value below 0.05 was considered statistically significant. Statistical analyses were performed using GraphPad Prism, Version 9.4.0 (RRID:SCR_002798), IBM SPSS Statistics for Windows, Version 26.0 (IBM Corp, RRID:SCR_019096), and the R statistical environment v.4.1.1 (R Core Team; R Foundation for Statistical Computing, RRID:SCR_001905). Survival analyses were computed with the R package survminer v.0.4.9. Overall survival (OS) was defined as the time of BM diagnosis to all-cause death or last follow-up. A log-rank test was performed to compare survival of groups and a Cox regression model was performed to estimate HRs.

Data availability

Raw data of DNA methylation analysis are publicly available at the Gene Expression Omnibus database respiratory under the accession number GSE220826. Further data will be shared upon reasonable request to the corresponding author.

Patients’ characteristics

We identified 696 patients with BM of breast cancer (n = 191) and NSCLC (n = 505) diagnosed at our institution in the last 30 years. From 180 patients, sufficient tissue material was available for scientific purposes and included into this study. The ethnicity of the overall cohort consisted exclusively of white Europeans. All patients with breast cancer were female, while 69.6% (32/48) of the patients with NSCLC were male. Of the 132 BCa-BM, 49 (37.1%) were of luminal subtype, 31 (23.5%) were HER2+, and 52 (39.4%) were TNBC. A total of 34/48 (70.8%) NSCLC-BM derived from lung adenocarcinomas, 12/48 (25.0%) had squamous, and 2/48 (4.2%) a mixed histology. While 87% of patients with NSCLC had only one BM at time of diagnosis, this only applied to 44.7% of patients with breast cancer. Matched extracranial samples were available from 46/48 patients with NSCLC which all derived from primary lung tumors except of one pleural carcinosis biopsy. Of the patients with NSCLC, 15/48 patients received adjuvant systemic treatment and 5/48 adjuvant radiotherapy of the primary, extracranial tumor (4/5 a combined radiochemotherapy of the primary tumor). In the breast cancer cohort, 87/132 patients received adjuvant systemic therapy and 92/132 adjuvant radiotherapy of the primary, extracranial tumor (69/132 a combined radiochemotherapy of the primary tumor). Further patients’ characteristics are summarized in Table 1. Patients’ characteristics according to HER3 expression levels are summarized separately in Table 2.

Table 1.

Baseline patients’ characteristics of the whole BM cohort including patients with breast and lung cancer.

Breast (n = 132)Lung (n = 48)Total (n = 180)
Gender, n (%) 
 Male 33 (68.8%) 33 (18.3%) 
 Female 132 (100%) 15 (31.3%) 147 (81.6%) 
Age at BM diagnosis, years 
 Median (IQR) 52 (44–61) 56 (51–62) 53 (45–62) 
 Range 27–78 43–72 27–78 
 Unknown 
IHC, n (%) 
 ER    
  Positive 54 (40.9%) NA 54 (40.9%) 
  Negative 78 (59.1%) NA 78 (59.1%) 
 PR    
  Positive 55 (41.7%) NA 55 (41.7%) 
  Negative 76 (57.6%) NA 76 (57.6%) 
 HER2    
  IHC0 61 (46.2%) 26 (54.2%) 87 (48.3%) 
  IHC1+ 25 (18.9%) 18 (37.5%) 43 (23.8%) 
  IHC2+ 18 (13.6%) 4 (8.3%) 22 (12.2%) 
  IHC3+ 28 (21.2%) 28 (15.6%) 
 HER3    
  Negative 33 (25%) 13 (27.1%) 46 (25.6%) 
  Strong 55 (41.7%) 24 (50.0%) 79 (43.9) 
  Weak 44 (33.3%) 11 (22.9%) 55 (30.6%) 
Year of BM diagnosis, n (%) 
 1990–2000 38 (28.8%) 15 (31.3%) 53 (29.4%) 
 2001–2010 42 (31.8%) 33 (68.8%) 75 (41.7%) 
 2011–2020 52 (39.4%) 52 (28.9%) 
Karnofsky, n (%) 
 <70 49 (37.1%) 5 (10.4%) 54 (30.0%) 
 80 38 (28.8%) 12 (25.0%) 50 (27.8%) 
 90 31 (23.5%) 17 (35.4%) 48 (26.7%) 
 100 11 (8.3%) 14 (29.2%) 25 (13.9%) 
 Missing 3 (2.3%) 3 (1.7%) 
Stage, n (%) 
 I 39 (29.6%) 16 (33.3%) 55 (30.6%) 
 II 53 (40.2%) 13 (27.1%) 66 (36.7) 
 III 27 (20.5%) 8 (16.7%) 35 (19.4%) 
 IV 13 (9.8%) 11 (22.9%) 24 (13.3%) 
Surgery prior BM, n (%) 123 (93.2%) 37 (77.1%) 160 (88.9%) 
 Missing 2 (1.5%) 2 (1.1%) 
Chemotherapy prior BM, n (%) 108 (81.8%) 16 (33.3%) 124 (68.9%) 
 Missing 5 (3.8%) 5 (2.8%) 
Adjuvant chemotherapy, n (%) 82 (62.1%) 15 (31.3%) 97 (53.9%) 
 Missing 8 (6.1%) 8 (4.4%) 
Adjuvant radiotherapy, n (%) 79 (59.8%) 5 (10.4%) 84 (46.7%) 
 Missing 10 (7.6%) 10 (5.6%) 
BM only, n (%) 59 (44.7%) 41 (85.4%) 100 (55.0%) 
 Missing 2 (1.5%) 2 (1.1%) 
Number of BM at diagnosis, n (%) 
 1 76 (57.6%) 42 (87.5%) 118 (64.4%) 
 2–3 34 (25.8%) 6 (6.3%) 40 (22.2%) 
 >3 17 (12.9%) 0 (6.3%) 17 (9.4%) 
 Missing 5 (3.8%) 5 (2.8%) 
TTP, primary tumor to BM diagnosis, months 
 Median (IQR) 32 (25.2–38.8) 11 (5.4–16.6) 26 (22.5–29.5) 
 Range 0–275 0–74 0—275 
 Unknown 
TTP, primary tumor to extracranial metastasis, months 
 Median (IQR) 24 (16.3–31.7) 23 (9.4–36.6) 23 (19.0–27.0) 
 Range 0–166 7–65 0–166 
 Unknown 70 42 112 
TTP, extracranial metastasis to BM diagnosis, months 
 Median (IQR) 16 (10.9–21.1) 2 (0–11.6) 14 (9.2–18.8) 
 Range 0–123 0–16 0–123 
 Unknown 70 42 112 
Overall survival, BM diagnosis, months 
 Median (IQR) 10 (3–21) 14 (6–45) 11 (4–24) 
 Range 0–134 0–183 0–183 
 1 year (%) 44.20% 54.30% 47.00% 
 Unknown 
Breast (n = 132)Lung (n = 48)Total (n = 180)
Gender, n (%) 
 Male 33 (68.8%) 33 (18.3%) 
 Female 132 (100%) 15 (31.3%) 147 (81.6%) 
Age at BM diagnosis, years 
 Median (IQR) 52 (44–61) 56 (51–62) 53 (45–62) 
 Range 27–78 43–72 27–78 
 Unknown 
IHC, n (%) 
 ER    
  Positive 54 (40.9%) NA 54 (40.9%) 
  Negative 78 (59.1%) NA 78 (59.1%) 
 PR    
  Positive 55 (41.7%) NA 55 (41.7%) 
  Negative 76 (57.6%) NA 76 (57.6%) 
 HER2    
  IHC0 61 (46.2%) 26 (54.2%) 87 (48.3%) 
  IHC1+ 25 (18.9%) 18 (37.5%) 43 (23.8%) 
  IHC2+ 18 (13.6%) 4 (8.3%) 22 (12.2%) 
  IHC3+ 28 (21.2%) 28 (15.6%) 
 HER3    
  Negative 33 (25%) 13 (27.1%) 46 (25.6%) 
  Strong 55 (41.7%) 24 (50.0%) 79 (43.9) 
  Weak 44 (33.3%) 11 (22.9%) 55 (30.6%) 
Year of BM diagnosis, n (%) 
 1990–2000 38 (28.8%) 15 (31.3%) 53 (29.4%) 
 2001–2010 42 (31.8%) 33 (68.8%) 75 (41.7%) 
 2011–2020 52 (39.4%) 52 (28.9%) 
Karnofsky, n (%) 
 <70 49 (37.1%) 5 (10.4%) 54 (30.0%) 
 80 38 (28.8%) 12 (25.0%) 50 (27.8%) 
 90 31 (23.5%) 17 (35.4%) 48 (26.7%) 
 100 11 (8.3%) 14 (29.2%) 25 (13.9%) 
 Missing 3 (2.3%) 3 (1.7%) 
Stage, n (%) 
 I 39 (29.6%) 16 (33.3%) 55 (30.6%) 
 II 53 (40.2%) 13 (27.1%) 66 (36.7) 
 III 27 (20.5%) 8 (16.7%) 35 (19.4%) 
 IV 13 (9.8%) 11 (22.9%) 24 (13.3%) 
Surgery prior BM, n (%) 123 (93.2%) 37 (77.1%) 160 (88.9%) 
 Missing 2 (1.5%) 2 (1.1%) 
Chemotherapy prior BM, n (%) 108 (81.8%) 16 (33.3%) 124 (68.9%) 
 Missing 5 (3.8%) 5 (2.8%) 
Adjuvant chemotherapy, n (%) 82 (62.1%) 15 (31.3%) 97 (53.9%) 
 Missing 8 (6.1%) 8 (4.4%) 
Adjuvant radiotherapy, n (%) 79 (59.8%) 5 (10.4%) 84 (46.7%) 
 Missing 10 (7.6%) 10 (5.6%) 
BM only, n (%) 59 (44.7%) 41 (85.4%) 100 (55.0%) 
 Missing 2 (1.5%) 2 (1.1%) 
Number of BM at diagnosis, n (%) 
 1 76 (57.6%) 42 (87.5%) 118 (64.4%) 
 2–3 34 (25.8%) 6 (6.3%) 40 (22.2%) 
 >3 17 (12.9%) 0 (6.3%) 17 (9.4%) 
 Missing 5 (3.8%) 5 (2.8%) 
TTP, primary tumor to BM diagnosis, months 
 Median (IQR) 32 (25.2–38.8) 11 (5.4–16.6) 26 (22.5–29.5) 
 Range 0–275 0–74 0—275 
 Unknown 
TTP, primary tumor to extracranial metastasis, months 
 Median (IQR) 24 (16.3–31.7) 23 (9.4–36.6) 23 (19.0–27.0) 
 Range 0–166 7–65 0–166 
 Unknown 70 42 112 
TTP, extracranial metastasis to BM diagnosis, months 
 Median (IQR) 16 (10.9–21.1) 2 (0–11.6) 14 (9.2–18.8) 
 Range 0–123 0–16 0–123 
 Unknown 70 42 112 
Overall survival, BM diagnosis, months 
 Median (IQR) 10 (3–21) 14 (6–45) 11 (4–24) 
 Range 0–134 0–183 0–183 
 1 year (%) 44.20% 54.30% 47.00% 
 Unknown 

Abbreviations: BM, brain metastases; ER, estrogen receptor; HER, human epidermal growth factor receptor; n, number; NA, not available; PR, progesterone receptor; IQR, interquartile range.

Table 2.

Baseline characteristics of patients, stratified according to their HER3 expression.

HER3 negative (n = 46)HER3 low (n = 55)HER3 high (n = 79)PaTest statistic (df)
Gender, n (%)    0.2105 χ²(2) = 3.117 
 Male 9 (19.6%) 6 (10.9%) 18 (22.8%)   
 Female 37 (80.4%) 49 (89.1%) 61 (77.2%)   
Age at BM diagnosis, years    0.566 χ²(2) = 1.138 
 Median (IQR) 51 (43–60) 55 (46–63) 54 (45–62)   
 Range 27–78 27–76 30–75   
Primary tumor, n (%)    0.3932 χ²(2) = 1.867 
 Lung 13 (28.3%) 11 (20.0%) 24 (30.4%)   
 Breast 33 (71.7%) 44 (80.0%) 55 869.6%)   
Subtype, n (%)    0.0084 χ²(6) = 17.238 
 Luminal 8 (17.4%) 22 (40.0%) 21 (26.6%)   
 HER2 4 (8.7%) 8 (14.5%) 19 (24.1%)   
 TNBC 21 (45.7%) 15 (27.3%) 16 (20.3%)   
 NSCLC 13 (28.3%) 10 (18.2%) 23 (29.1%)   
IHC, n (%) 
 ER    0.0145 χ²(2) = 8.747 
  Positive 7 (15.2%) 18 (32.7%) 29 (36.7%)   
  Negative 26 (56.5%) 26 (47.3%) 26 (32.9%)   
 PR    0.4874 χ²(2) = 1.437 
  Positive 11 (23.9%) 20 (36.4%) 24 (30.4%)   
  Negative 22 (47.8%) 23 (41.8%) 31 (39.2%)   
 HER2    0.003 χ²(6) = 19.806 
  IHC0 33 (71.7%) 20 (36.4%) 34 (43.0%)   
  IHC1+ 8 (17.4%) 19 (34.5%) 16 (20.3%)   
  IHC2+ 4 (8.7%) 7 (12.7%) 11 (13.9%)   
  IHC3+ 1 (2.2%) 9 (16.4%) 18 (22.8%)   
Year of BM diagnosis, n (%)    0.3133 χ²(4) = 4.755 
 1990–2000 18 (32.6%) 16 (25.9) 19 (24.4%)   
 2001–2010 19 (44.2%) 23 (42.6%) 32 (41.0%)   
 2011–2020 9 (20.9%) 16 (29.6%) 28 (35.9%)   
Karnofsky, n (%)    0.5729 χ²(6) = 4.776 
 <70 15 (32.6%) 18 (32.7%) 21 (26.7%)   
 80 15 (32.6%) 16 (29.1%) 19 (24.1%)   
 90 12 (26.1%) 14 (25.5%) 22 (27.8%)   
 100 3 (6.5%) 7 (12.7%) 15 (19.0%)   
 Missing 1 (2.2%) 2 (2.5%)   
Stage, n (%)    0.6011 χ²(2) = 1.018 
 I—III 37 (80.4%) 48 (87.3%) 69 (87.3%)   
 IV 8 (17.4%) 6 (10.9%) 10 (12.7%)   
 Missing 1 (2.2%) 1 (1.8%)   
Surgery prior BM, n (%) 40 (87.0%) 49 (89.1%) 71 (89.9%) 0.857 † 
 Missing 1 (2.2%) 1 (1.3%)   
Chemotherapy prior BM, n (%) 30 (65.2%) 40 (72.7%) 54 (68.4%) 0.8838 χ²(2) = 0.247 
 Missing 2 (4.3%) 3 (3.8%)   
Adjuvant chemotherapy, n (%) 27 (58.7%) 27 (49.1%) 43 (54.4%) 0.4245 χ²(2) = 1.713 
 Missing 4 (8.7%) 2 (3.6%) 2 (2.5%)   
Adjuvant radiotherapy, n (%) 17 (37.0%) 27 (49.1%) 40 (50.6%) 0.5052 χ²(2) = 1.366 
 Missing 5 (10.9%) 3 (5.5%) 2 (2.5%)   
BM only, n (%) 21 (45.7%) 28 (50.9%) 51 (64.6%) 0.0838 χ²(2) = 4.959 
 Missing 1 (2.2%) 1 (1.3%)   
Relapse BM, n (%) 24 (52.2%) 29 (52.7%) 39 (49.4%) 0.9224 χ²(2) = 0.162 
 Missing 1 (2.2%) 1 (1.3%)   
Relapse tumor, n (%) 16 (34.8%) 26 (47.3%) 35 (44.3%) 0.465 χ²(2) = 1.532 
 Missing 1 (2.2%) 1 (1.3%)   
TTP, primary tumor to BM diagnosis, months 
 Median (IQR) 20 (10.1–29.9) 30 (20.8–39.2) 26 (19.3–32.7) 0.061 χ²(2) = 5.590 
 Range 0—100 0–200 0–275   
 Unknown   
TTP, primary tumor to extracranial metastasis, months 
 Median (IQR) 18 (1.9–34.1) 24 (13.0–35.0) 25 (16.8–33.2) 0.441 χ²(2) = 1.638 
 Range 0–57 0–76 0–166   
 Unknown 26 32 54   
Overall survival, BM diagnosis    0.118 χ²(2) = 4.268 
 Median, months 6 (2, 20) 11 (4, 24) 12 (4, 31)   
 Range 0–183 1—122 0–141   
 1 year (%) 39.10% 45.00% 54.50%   
 Unknown   
HER3 negative (n = 46)HER3 low (n = 55)HER3 high (n = 79)PaTest statistic (df)
Gender, n (%)    0.2105 χ²(2) = 3.117 
 Male 9 (19.6%) 6 (10.9%) 18 (22.8%)   
 Female 37 (80.4%) 49 (89.1%) 61 (77.2%)   
Age at BM diagnosis, years    0.566 χ²(2) = 1.138 
 Median (IQR) 51 (43–60) 55 (46–63) 54 (45–62)   
 Range 27–78 27–76 30–75   
Primary tumor, n (%)    0.3932 χ²(2) = 1.867 
 Lung 13 (28.3%) 11 (20.0%) 24 (30.4%)   
 Breast 33 (71.7%) 44 (80.0%) 55 869.6%)   
Subtype, n (%)    0.0084 χ²(6) = 17.238 
 Luminal 8 (17.4%) 22 (40.0%) 21 (26.6%)   
 HER2 4 (8.7%) 8 (14.5%) 19 (24.1%)   
 TNBC 21 (45.7%) 15 (27.3%) 16 (20.3%)   
 NSCLC 13 (28.3%) 10 (18.2%) 23 (29.1%)   
IHC, n (%) 
 ER    0.0145 χ²(2) = 8.747 
  Positive 7 (15.2%) 18 (32.7%) 29 (36.7%)   
  Negative 26 (56.5%) 26 (47.3%) 26 (32.9%)   
 PR    0.4874 χ²(2) = 1.437 
  Positive 11 (23.9%) 20 (36.4%) 24 (30.4%)   
  Negative 22 (47.8%) 23 (41.8%) 31 (39.2%)   
 HER2    0.003 χ²(6) = 19.806 
  IHC0 33 (71.7%) 20 (36.4%) 34 (43.0%)   
  IHC1+ 8 (17.4%) 19 (34.5%) 16 (20.3%)   
  IHC2+ 4 (8.7%) 7 (12.7%) 11 (13.9%)   
  IHC3+ 1 (2.2%) 9 (16.4%) 18 (22.8%)   
Year of BM diagnosis, n (%)    0.3133 χ²(4) = 4.755 
 1990–2000 18 (32.6%) 16 (25.9) 19 (24.4%)   
 2001–2010 19 (44.2%) 23 (42.6%) 32 (41.0%)   
 2011–2020 9 (20.9%) 16 (29.6%) 28 (35.9%)   
Karnofsky, n (%)    0.5729 χ²(6) = 4.776 
 <70 15 (32.6%) 18 (32.7%) 21 (26.7%)   
 80 15 (32.6%) 16 (29.1%) 19 (24.1%)   
 90 12 (26.1%) 14 (25.5%) 22 (27.8%)   
 100 3 (6.5%) 7 (12.7%) 15 (19.0%)   
 Missing 1 (2.2%) 2 (2.5%)   
Stage, n (%)    0.6011 χ²(2) = 1.018 
 I—III 37 (80.4%) 48 (87.3%) 69 (87.3%)   
 IV 8 (17.4%) 6 (10.9%) 10 (12.7%)   
 Missing 1 (2.2%) 1 (1.8%)   
Surgery prior BM, n (%) 40 (87.0%) 49 (89.1%) 71 (89.9%) 0.857 † 
 Missing 1 (2.2%) 1 (1.3%)   
Chemotherapy prior BM, n (%) 30 (65.2%) 40 (72.7%) 54 (68.4%) 0.8838 χ²(2) = 0.247 
 Missing 2 (4.3%) 3 (3.8%)   
Adjuvant chemotherapy, n (%) 27 (58.7%) 27 (49.1%) 43 (54.4%) 0.4245 χ²(2) = 1.713 
 Missing 4 (8.7%) 2 (3.6%) 2 (2.5%)   
Adjuvant radiotherapy, n (%) 17 (37.0%) 27 (49.1%) 40 (50.6%) 0.5052 χ²(2) = 1.366 
 Missing 5 (10.9%) 3 (5.5%) 2 (2.5%)   
BM only, n (%) 21 (45.7%) 28 (50.9%) 51 (64.6%) 0.0838 χ²(2) = 4.959 
 Missing 1 (2.2%) 1 (1.3%)   
Relapse BM, n (%) 24 (52.2%) 29 (52.7%) 39 (49.4%) 0.9224 χ²(2) = 0.162 
 Missing 1 (2.2%) 1 (1.3%)   
Relapse tumor, n (%) 16 (34.8%) 26 (47.3%) 35 (44.3%) 0.465 χ²(2) = 1.532 
 Missing 1 (2.2%) 1 (1.3%)   
TTP, primary tumor to BM diagnosis, months 
 Median (IQR) 20 (10.1–29.9) 30 (20.8–39.2) 26 (19.3–32.7) 0.061 χ²(2) = 5.590 
 Range 0—100 0–200 0–275   
 Unknown   
TTP, primary tumor to extracranial metastasis, months 
 Median (IQR) 18 (1.9–34.1) 24 (13.0–35.0) 25 (16.8–33.2) 0.441 χ²(2) = 1.638 
 Range 0–57 0–76 0–166   
 Unknown 26 32 54   
Overall survival, BM diagnosis    0.118 χ²(2) = 4.268 
 Median, months 6 (2, 20) 11 (4, 24) 12 (4, 31)   
 Range 0–183 1—122 0–141   
 1 year (%) 39.10% 45.00% 54.50%   
 Unknown   

Abbreviations: †, nonparametric test; BM, brain metastases; CI, confidence interval; df, degrees of freedom; ER, estrogen receptor; HER, human epidermal growth factor receptor; n, number; NSCLC, non–small cell lung cancer; PR, progesterone receptor; TNBC, triple-negative breast cancer.

aPearson χ² test, Kruskal–Wallis rank-sum test, Fisher exact test, log-rank test.

HER3 is frequently expressed in BM of patients with breast and lung cancer

The expression pattern of HER3 varied between tumor samples from completely negative to low and high intensity (Table 1; Fig. 1A). Overall, 99/132 (75.0%) BCa-BM and 33/48 (68.8%) NSCLC-BM presented with HER3 positivity (Fig. 1B; Table 1). A total of 10/14 (71.4%) of NSCLC-BM with a squamous or mixed histology and 23/34 (67.6%) of the adenocarcinoma-based NSCLC-BM were HER3+. HER3 expression significantly differed between breast cancer subtypes: 27/31 (87.1%) HER2+ BCa-BM patients presented with HER3 expression, in contrast to 41/49 (83.7%) luminal breast cancer and 31/52 (59.6%) TNBC BM (P < 0.01; χ2 test; Fig. 1C). A total of 37/61 (60.6%) patients with HER2 breast cancer presented with HER3 expression and 31/37 (83.8%) of HER2-low patients were positive for HER3 (Fig. 1D; Supplementary Fig. S1B). Importantly, specimens with high levels of HER3 expression were also different according to breast cancer subtype: 19/31 (61.3%) in HER2+, 20/49 (40.8%) in luminal, and 16/52 (30.8%) in TNBC (P = 0.02; χ2 test) BMs. While the amount of HER3 expression indeed decreased in association with HER2, 50% of HER2 (IHC score 0) BCa-BM were still positive for HER3, with almost a third of them even expressing high levels of HER3 (Supplementary Fig. S1C).

HER3 expression is more frequent in NSCLC-BM than in matched extracranial tumors

From 46/48 patients (95.8%) with NSCLC, matched extracranial tumor material for the analysis of HER3 expression was available. HER3 expression was more commonly observed in BM than in extracranial tumors (34/47 vs. 19/47; P = 0.003; Fisher exact test; Fig. 1E); high HER3 expression was more frequently observed in BM than in extracranial tumors as well (48.9% vs. 8.5%; P < 0.001, Fisher exact test). A similar increase of expression was observed for HER2 in BM samples. Here, 22/48 (45.8%) of NSCLC-BM where HER2-low compared with 7/46 (15.2%) of NSCLC primary tumors (Supplementary Fig. S1D). We did not detect any HER2 IHC 3+ cases in the NSCLC cohort. Of the HER2-low NSCLC-BM, 19/22 (86.4%) were also positive for HER3 (Supplementary Fig. S1E). Given the long inclusion period of our study we investigated whether the year of sample acquisition and the results of HER3 IHC staining interact with each other. However, the decade of diagnosis did not differ significantly between HER3-high, HER3-low, and HER3 samples (P = 0.3133, χ2 test; Table 2).

Correlation of HER3 with survival time after BM diagnosis

Median OS from BM diagnosis was 10 months [interquartile range (IQR) = 3–21] in patients with breast cancer and 14 months (IQR = 6–45) in patients with NSCLC (Table 1). Regarding different breast cancer subtypes, patients with luminal breast cancer and HER3 expression presented with improved OS compared with patients without HER3 expression [P = 0.032, HR = 0.39, 95% confidence interval (CI) = 0.17–0.92], while no difference was observed in other breast cancer subtypes (Fig. 2AC). By including the extent of expression into the analysis, only patients with high HER3 levels had longer OS (Table 2; Supplementary Fig. S2A–S2C). In patients with NSCLC-BM, no difference in survival was evident according to HER3 expression (P = 0.73, HR = proportional hazard assumption does not hold; Fig. 2D; Supplementary Fig. S2D).

Figure 2.

Survival analysis according to HER3 expression in BM of patients with breast and lung cancer. Patients were stratified into HER3+ (high and low HER3 expression) and HER3 subgroups. Time from first diagnosis of BM until death was plotted by the Kaplan–Meier method, and P value was calculated by the log-rank test. Unlike patients with BM derived from HER2+ (B) or triple-negative BCa (C), and NSCLC (D), only those with BM from luminal BCa (A) showed a significant (P = 0.028) difference in OS depending on HER3 expression. No Cox regression was performed for the HER2 dataset because of insufficient number of events and NSCLC because proportional hazard could not be assumed. HR, hazard ratio; CI, confidence interval.

Figure 2.

Survival analysis according to HER3 expression in BM of patients with breast and lung cancer. Patients were stratified into HER3+ (high and low HER3 expression) and HER3 subgroups. Time from first diagnosis of BM until death was plotted by the Kaplan–Meier method, and P value was calculated by the log-rank test. Unlike patients with BM derived from HER2+ (B) or triple-negative BCa (C), and NSCLC (D), only those with BM from luminal BCa (A) showed a significant (P = 0.028) difference in OS depending on HER3 expression. No Cox regression was performed for the HER2 dataset because of insufficient number of events and NSCLC because proportional hazard could not be assumed. HR, hazard ratio; CI, confidence interval.

Close modal

HER3 expression is not correlated with the inflammatory tumor microenvironment composition

Overall, there was no significant difference between CD3+ (P = 0.4774, Kruskal–Wallis test), CD8+ (P = 0.4464), and CD68+ (P = 0.3834) immune cell density in BM of patients with breast cancer with different HER3 expression (Fig. 3A). No statistically significant difference was observed in breast cancer subtypes either (Supplementary Fig. S3A–S3C). Similarly, no differences in immune cell infiltration based on HER3 status were observed in BM of patients with NSCLC, including CD3+ (P = 0.0821), CD8+ (P = 0.2848), and CD68+ (P = 0.9725) leukocytes (Fig. 3B). However, overall, the number of immune cells in the tumor microenvironment was higher in BM of patients with NSCLC compared with patients with breast cancer: CD3 (362 vs. 156 cells/mm2, P < 0.001, estimated difference between means (est. MD) = 187.414, CI = 120.184–263.515, Mann–Whitney test), CD8 (131 vs. 67 cells/mm2, P = 0.003, est. MD = 22.223, CI = 7.774–43.786), and CD68 (1,988 vs. 672 cells/mm2, P < 0.001, est. MD = 1044.598, CI = 757.613–1277.654).

Figure 3.

Immune cell infiltration is not associated with HER3 expression. The number of CD3+ T lymphocytes, CD8+ cytotoxic T cells, and CD68+ macrophages in BCa-BM (A) and NSCLC-BM (B). No differences in either HER3 group could be detected (P > 0.05, Kruskal–Wallis test); therefore, no further pairwise comparisons have been conducted.

Figure 3.

Immune cell infiltration is not associated with HER3 expression. The number of CD3+ T lymphocytes, CD8+ cytotoxic T cells, and CD68+ macrophages in BCa-BM (A) and NSCLC-BM (B). No differences in either HER3 group could be detected (P > 0.05, Kruskal–Wallis test); therefore, no further pairwise comparisons have been conducted.

Close modal

Methylation signatures differ according to HER3 expression

To gain further insight on potential molecular drivers of HER3 expression, we performed methylation analysis as an omics method providing a broad insight and directions for further in-depth analysis. To identify relevant pathways potentially affected by differing HER3 expression, we interrogate the DNA methylome of HER3+ and HER3 BM. Overall, we analyzed 71 samples comprising of 16 patients with NSCLC (8 HER3+ and 8 HER3 BM) and 55 patients with breast cancer (27 HER3+, 20 HER3, and 8 HER3-low BM). A detailed description of the patient characteristics used for methylation analysis is shown in Supplementary Table S2. After data processing and clearing, 641,418 CpG sites remained for genome-wide methylation analysis. By performing a uniform manifold approximation and projection (UMAP) model for dimensional reduction using the top 10,000 CpG sites based on variance of beta values across the whole dataset, the BM samples separated into three major clusters: (i) a luminal/HER2+, (ii) a TNBC, and (iii) a NSCLC cluster (Fig. 4A). In addition, while HER3+ and HER3 samples of TNBC and NSCLC-BM are not divisible within their clusters, only the HER3 samples of HER2+ and luminal BM misclustered within TNBC and NSCLC-BM. Subsequently, we were focusing on a subtype-specific analysis to identify differentially methylated positions (DMP) between HER3+ and HER3 BM. For luminal BM, we identified 66,999 (44,809 hypermethylated and 22,190 hypomethylated) DMPs, 56,673 (21,151 hypermethylated and 35,522 hypomethylated) for HER2+ BM, and 47,482 (21,901 hypermethylated and 25,581 hypomethylated) for TNBC-BM, but only 15 (six hypermethylated and nine hypomethylated) DMPs for NSCLC-BM indicating an inferior effect of HER3 expression in NSCLC (Fig. 4B). Next, we focused on the top 1,000 differentially methylated sites to define HER3-specific methylation signatures in each breast cancer subtype. A list of these sites for each subtype including relevant annotation is provided in Supplementary Tables S3–S5. Hierarchical clustering for each subtype separated HER3+ from HER3 BM with only a single outlier in HER2+ and luminal breast cancer BM (Fig. 4C). Each detected CpG site was then annotated to its designated gene that were further used for pathway enrichment analyses to detect dysregulated pathways (Fig. 4D). Supplementary Table S6 summarizes these pathways giving additional relevant information. A recurrent hit, appearing in both, HER2+ and TNBC BM, was associated with Wnt signaling. Furthermore, EGF/EGFR signaling pathways including its known downstream PI3K/AKT signaling cascade appears deregulated in TNBC BM together with G protein–coupled receptor involved pathways. In Luminal BM, TGFβ signaling seems affected by differential methylation of selected target genes.

Figure 4.

DNA methylation profiling of patients with BM of breast and lung cancer. A, Dimension reduction by UMAP to compare DNA methylation signatures across different breast cancer subtypes and NSCLC-derived BM. Different colors indicate subtype identity, whereas shape reveals HER3 status. B, Scatter plot comparing the mean beta values of each CpG site between HER3+ and HER3 samples of each subtype, respectively. Red dots indicate the top 1,000 differentially methylated CpG sites. C, Heatmaps with hierarchical clustering of each breast cancer subtype using the top 1,000 differently methylated CgG sites between HER3+ (red) and HER3 (blue) samples. Heatmap colors represent the unscaled and/or uncentered β values. D, Pathway enrichment analysis query WikiPathways identified deregulated pathways in each breast cancer subtype depending on HER3 expression. FDR, false discovery rate.

Figure 4.

DNA methylation profiling of patients with BM of breast and lung cancer. A, Dimension reduction by UMAP to compare DNA methylation signatures across different breast cancer subtypes and NSCLC-derived BM. Different colors indicate subtype identity, whereas shape reveals HER3 status. B, Scatter plot comparing the mean beta values of each CpG site between HER3+ and HER3 samples of each subtype, respectively. Red dots indicate the top 1,000 differentially methylated CpG sites. C, Heatmaps with hierarchical clustering of each breast cancer subtype using the top 1,000 differently methylated CgG sites between HER3+ (red) and HER3 (blue) samples. Heatmap colors represent the unscaled and/or uncentered β values. D, Pathway enrichment analysis query WikiPathways identified deregulated pathways in each breast cancer subtype depending on HER3 expression. FDR, false discovery rate.

Close modal

Furthermore, to determine whether HER3 expression may be regulated by DNA methylation, we specifically evaluated the methylation level of 33 selected CpG sites associated with the genomic locus of HER3 (ERBB3) on the Infinium Methylation EPIC microarray. However, we did not detect any differences between HER3+ and HER3 BM including all analyzed samples indicating a DNA methylation independent regulation of HER3 expression in those samples (Supplementary Fig. S4A).

HER3 is of particular interest in the context of BM, as its expression is associated with BM development and serves as a promising therapeutic target for personalized therapy approaches. In the current study, we could detect widespread expression of HER3 in BM of patients with breast cancer and NSCLC. Indeed, more than two-thirds of BM samples presented with detectable HER3 expression of any intensity whereas almost half of them had high expression as defined by complete membranous staining of over 10% of tumor cells. BM from the HER2+ subtype presented most frequently with HER3 expression. However, also the majority of TNBC and NSCLC-BM samples showed HER3 expression. Given the lack of systemic treatment options particularly in TNBC BM so far, our data strongly support further development of HER3-directed therapies in the context of BM.

To gain more insight on functional aspects of HER3 expression in BM, we performed genome-wide DNA methylation profiling and pathway analysis. Here, we could verify subtype-specific methylation signatures as recently demonstrated by Capper and colleagues (34). This supports the diverse tissue origin of our BM samples and is an important quality measure of our data. In addition, methylation profiles of HER3+ and HER3 samples did differ according to the underlying tumor subtype. The identified differentially methylation patterns are strongly associated with pathways known to facilitate and support brain metastatic outgrowth. For example, brain-derived neutrophic factor signaling including TRKB (NTRK2) appears dysregulated in HER3+/HER2+ BM and is associated with neurogenesis and synaptic development (35). The neutrophin receptor TRKB was shown to be highly activated in patient tissue and breast cancer cell lines and heterodimerization of HER2 and TRKB was postulated as a potential mediator for BM progression (36, 37). However, if TRKB could also serve as a potential binding partner of HER3 is yet unknown. Furthermore, we identified Wnt signaling to be dysregulated in BM of HER2+ and TNBC BM according to HER3 expression. Previously, the Wnt signaling pathway was identified as driver of BM in basal-like breast cancer (38). Indeed, HER3 HER2+ BM samples clustered within the TNBC BM due to their similar methylation profile, supporting their potential biological relation. While we could not show direct pathways causing HER3 overexpression, we found no evidence that DNA methylation is involved in the regulation of HER3 expression in BM of patients with breast cancer and NSCLC.

HER3 was expressed more frequently in BM compared with the matched primary tumors of patients with NSCLC, supporting the importance of HER3 signaling for BM development. Interestingly, the rate of HER3 expression in our cohort of extracranial NSCLC samples appeared lower than that from other sources, ranging from 81% to 90% (25, 39). However, in their studies, Yonesaka and colleagues and Jänne and colleagues only included NSCLC with mutated EGFR indicating a correlation of the expression of different HER family members. Consistently, our observation that HER2+ breast tumors show the highest expression of HER3 coincide with other studies (40). Unfortunately, we were not able to include matched primary tumor samples from patients with breast cancer to compare HER3 expression in time and space of this tumor entity. Other reports investigating the expression of HER3 in primary breast tumors vary greatly, ranging from 15% to 67% (4, 41). However, preliminary data from other groups support our finding of increased levels of HER3 in BM (42). Da Silva and colleagues evaluated BM of patients with breast cancer in a smaller cohort in 2010 and found that 11/37 (29.7%) of the primary tumors, 22/37 (59%) of the matched metastases, and 13/21 (62%) of the unmatched BM (P = 0.019) showed positivity for HER3 by IHC (17). Taken together, these observations indicate HER3 as a critical mediator in brain metastatic spread, which could serve as a selective pressure to promote the outgrowth of HER3+ cells at this particular metastatic site. However, further mechanistic insight on the role of HER3 in the development of BM is warranted, particular in the context of new HER3-targeting ADCs. In line, preclinical data further support the importance of HER3 in the brain metastatic cascade, particularly in the context of coexpression with HER2. While HER2 represents an orphan receptor that lacks a ligand, neuregulin-1 (NRG1 or heregulin-1) binds to HER3 and leads to its dimerization and further activation (17). The heregulin-HER3-HER2 signaling was shown to promote the matrix metalloproteinase–dependent transendothelial migration of BM initiating cancer cells through the blood–brain barrier and thereby facilitate BM establishment (18). Indeed, levels of NRG-1 are particularly high in the brain and are secreted by both, microglial and neuronal cells (43–45). Therefore, further preclinical studies concentrating on the role of HER3 inhibition to prevent BM development would be of high interest.

A novel interesting therapeutic approach in cancer therapy involves conjugates of specific mAbs coupled to cytotoxic drugs (ADCs). HER3-directed ADCs like patritumab-deruxtecan have shown promising activity experimentally and in early clinical development (25, 46–48). Indeed, the correlation of HER3 expression and response to HER3-targeting ADCs is currently under investigation. The first published clinical study suggested a high response rate for the HER3 ADC patritumab-deruxtecan in patients with EGFR-mutated NSCLC (25). Although, this early clinical trial did not require HER3 expression as an inclusion criterion, membranous HER3 expression was detected retrospectively in all included patients. Therefore, the observed response rates toward the HER3-targeting ADC could be explained at least by an enrichment for HER3 expression in this particular study population. In the meantime, other current clinical trials define at least a low expression of HER3 as a mandatory biomarker for inclusion (26). Interestingly, response to the HER2-directed ADC trastuzumab-deruxtecan was observed for patients with very low HER2 expression as well, leading to its approval for patients with HER2-low breast cancer (49). Therefore, further investigation is required to define the predictive role of HER3 expression in the context of HER3-targeting ADCs. The next generation of ADCs has in comparison with earlier generation improved cleavable linker-payload systems, which enable stable drug delivery to cancer cells causing selective destruction of antigen-expressing cells (50). In addition, by passive diffusion of cytotoxic drug molecules into neighboring antigen-negative tumor cells a so-called bystander effect further increases the efficacy of novel ADCs (51). Given the recent reports of high intracranial activity of the HER2-directed ADC trastuzumab-deruxtecan in patients with HER2+ BCa-BM, ADCs might become a particular promising approach in this disease setting (22–24). The current data of high HER3 expression in BM and in particular of higher expression as compared with matched primary tumors, strongly support HER3 as a target for ADCs in systemic treatment of BM.

It has been shown by various clinical trials that the presence of tumor-infiltrating lymphocytes in HER2+ early and advanced breast cancer is associated with a favorable prognosis and predicts a beneficial anti-HER2 therapeutic response (52–54). We did not observe any HER3 expression–associated differences in the respective inflammatory profiles, instead a steady state across all cohorts was found. Despite this observation, the sufficient number of immune cells required for antibody-dependent immunogenicity remains unknown and the mechanism of ADCs may depend less upon antibody-dependent cellular cytotoxicity but on the activity of the cytotoxic agent.

HER3 is a kinase-defective receptor; therefore, its sole expression might have only limited oncogenic potential and therefore no impact on OS. More important than a prognostic impact is a potential predictive impact as a biomarker for response to HER3-targeting ADC. However, previous reports suggested a prognostic significance for HER3 in solid tumors, such as gastric cancer and prostate cancer (46, 55, 56). In contrast, we observed no consistent correlation of HER3 expression with survival prognosis. While certain reports indeed found HER3 levels do serve as prognostic factors in hormone receptor–negative breast cancer (57–59), we were not able to verify its role as a negative predictor for patients with BM of breast and lung cancer as only hormone receptor–positive (luminal) metastatic patients with breast cancer with high expression of HER3 were associated with better prognosis. An association of HER3 expression with OS was adjusted for established risk factors including age, gender, and graded prognostic assessment class. Here, the association of HER3 expression remained statistically significant in the subgroup of luminal breast cancer (HR = 0.39; 95% CI, 0.17–092; P = 0.032), while no association of the other investigated factors was observed. Interestingly, Lee and colleagues observed a similar favorable clinical outcome in patients with hormone receptor–positive breast cancer based on disease-free survival (60). Frankly, the long observational period covering three decades might have introduced confounder that limit definitive survival analyses. For example, standard lines of anticancer therapy have changed throughout the years for both, breast cancer and NSCLC. However, while we could not detect an influence of adjuvant chemotherapy given on OS in any of the investigated subgroups, we cannot deny that modern targeted therapy has generally prolonged survival of patients with cancer. Taken this heterogeneity into consideration including the rather small sample sizes per subgroup, any conclusion and generalization using a rather historical cohort mustbe taken with caution. Nevertheless, to the best of our knowledge, this study represents the most comprehensive work on HER3 expression in BM so far.

In conclusion, our data highlight the role of HER3 expression in BM and encourage further clinical development of HER3-targeting therapeutics in patients with BCa-BM and NSCLC-BM.

F. Eckert reports personal fees from Dr. Sennewald GmbH outside the submitted work. R. Bartsch reports personal fees from AstraZeneca, Eisai, Eli Lilly, Gilead, Grunenthal, MSD, Novartis, Pfizer, Pierre-Fabre, Roche, and Seagen and grants and personal fees from Daiichi outside the submitted work. G. Heller reports personal fees from X4 Pharmaceuticals outside the submitted work. M. Preusser reports personal fees and non-financial support from Bayer, Bristol-Myers Squibb, Novartis, Gerson Lehrman Group (GLG), CMC Contrast, GlaxoSmithKline, Mundipharma, Roche, BMJ Journals, MedMedia, AstraZeneca, AbbVie, Lilly, Medahead, Daiichi Sankyo, Sanofi, Merck Sharp & Dome, Tocagen, Adastra, Gan & Lee Pharmaceuticals, and Servier outside the submitted work. A.S. Berghoff reports grants from Daiichi Sankyo during the conduct of the study as well as grants, personal fees, non-financial support, and other support from Roche; personal fees and non-financial support from Merck, AstraZeneca, CeCaVa, Bristol Myers Squibb, and Daiichi Sankyo; and other support from Amgen and AbbVie outside the submitted work. No disclosures were reported by the other authors.

E. Tomasich: Conceptualization, data curation, software, formal analysis, validation, investigation, visualization, methodology, writing–original draft, project administration, writing–review and editing. A. Steindl: Data curation, formal analysis, validation, investigation, writing–review and editing. C. Paiato: Data curation, software, formal analysis, investigation, methodology. T. Hatziioannou: Data curation, software, formal analysis, validation, visualization, methodology, writing–review and editing. M. Kleinberger: Software, formal analysis, investigation, methodology, writing–review and editing. L. Berchtold: Software, formal analysis, validation, writing–review and editing. R. Puhr: Software, formal analysis, validation, investigation. J.A. Hainfellner: Resources, formal analysis, validation. L. Müllauer: Resources, formal analysis, validation, methodology, writing–review and editing. G. Widhalm: Formal analysis, investigation. F. Eckert: Formal analysis, writing–review and editing. R. Bartsch: Conceptualization, formal analysis, investigation, writing–review and editing. G. Heller: Conceptualization, data curation, software, formal analysis, validation, visualization, methodology, writing–review and editing. M. Preusser: Conceptualization, resources, supervision, funding acquisition, writing–review and editing. A.S. Berghoff: Conceptualization, resources, formal analysis, supervision, funding acquisition, investigation, writing–original draft, project administration.

The Austrian Federal Ministry for Digital and Economic Affairs, the National Foundation for Research, Technology and Development, and the Christian Doppler Research Association financially supported this study by research grants to M. Preusser. This study was further supported by a research grant of Daiichi Sankyo to A.S. Berghoff.

The financial support by the Austrian Federal Ministry for Digital and Economic Affairs, the National Foundation for Research, Technology and Development, and the Christian Doppler Research Association is gratefully acknowledged. This study was further supported by an unrestricted grant of Daiichi Sankyo.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Kumagai
S
,
Koyama
S
,
Nishikawa
H
.
Antitumour immunity regulated by aberrant ERBB family signalling
.
Nat Rev Cancer
2021
;
21
:
181
97
.
2.
Citri
A
,
Skaria
KB
,
Yarden
Y
.
The deaf and the dumb: the biology of ErbB-2 and ErbB-3
.
Exp Cell Res
2003
;
284
:
54
65
.
3.
Gandullo-Sánchez
L
,
Ocaña
A
,
Pandiella
A
.
HER3 in cancer: from the bench to the bedside
.
J Exp Clin Cancer Res
2022
;
41
:
310
.
4.
Travis
A
,
Pinder
SE
,
Robertson
JFR
,
Bell
JA
,
Wencyk
P
,
Gullick
WJ
, et al
.
C-erbB-3 in human breast carcinoma: expression and relation to prognosis and established prognostic indicators
.
Br J Cancer
1996
;
74
:
229
33
.
5.
Tanner
B
,
Hasenclever
D
,
Stern
K
,
Schormann
W
,
Bezler
M
,
Hermes
M
, et al
.
ErbB-3 predicts survival in ovarian cancer
.
J Clin Oncol
2006
;
24
:
4317
23
.
6.
Koumakpayi
IH
,
Diallo
JS
,
Le Page
C
,
Lessard
L
,
Gleave
M
,
Bégin
LR
, et al
.
Expression and nuclear localization of ErbB3 in prostate cancer
.
Clin Cancer Res
2006
;
12
:
2730
7
.
7.
Hayashi
M
,
Inokuchi
M
,
Takagi
Y
,
Yamada
H
,
Kojima
K
,
Kumagai
J
, et al
.
High expression of HER3 is associated with a decreased survival in gastric cancer
.
Clin Cancer Res
2008
;
14
:
7843
9
.
8.
Reschke
M
,
Mihic-Probst
D
,
Van Horst EH Der
KP
,
Wild
PJ
,
Hutterer
M
, et al
.
HER3 is a determinant for poor prognosis in melanoma
.
Clin Cancer Res
2008
;
14
:
5188
97
.
9.
Beji
A
,
Horst
D
,
Engel
J
,
Kirchner
T
,
Ullrich
A
.
Toward the prognostic significance and therapeutic potential of HER3 receptor tyrosine kinase in human colon cancer
.
Clin Cancer Res
2012
;
18
:
956
68
.
10.
Siegfried
JM
,
Lin
Y
,
Diergaarde
B
,
Lin
HM
,
Dacic
S
,
Pennathur
A
, et al
.
Expression of PAM50 genes in lung cancer: evidence that interactions between hormone receptors and HER2/HER3 contribute to poor outcome
.
Neoplasia
2015
;
17
:
817
25
.
11.
Qian
G
,
Jiang
N
,
Wang
D
,
Newman
S
,
Kim
S
,
Chen
Z
, et al
.
Heregulin and HER3 are prognostic biomarkers in oropharyngeal squamous cell carcinoma
.
Cancer
2015
;
121
:
3600
11
.
12.
Ocana
A
,
Vera-Badillo
F
,
Seruga
B
,
Templeton
A
,
Pandiella
A
,
Amir
E
.
HER3 overexpression and survival in solid tumors: a meta-analysis
.
J Natl Cancer Inst
2013
;
105
:
266
73
.
13.
Kruser
TJ
,
Wheeler
DL
.
Mechanisms of resistance to HER family targeting antibodies
.
Exp Cell Res
2010
;
316
:
1083
100
.
14.
Vlacich
G
,
Coffey
RJ
.
Resistance to EGFR-targeted therapy: a family affair
.
Cancer Cell
2011
;
20
:
423
5
.
15.
Mishra
R
,
Patel
H
,
Alanazi
S
,
Yuan
L
,
Garrett
JT
.
HER3 signaling and targeted therapy in cancer
.
Oncol Rev
2018
;
12
:
355
.
16.
Haikala
HM
,
Jänne
PA
.
Thirty years of HER3: from basic biology to therapeutic interventions
.
Clin Cancer Res
2021
;
27
:
3528
39
.
17.
Da Silva
L
,
Simpson
PT
,
Smart
CE
,
Cocciardi
S
,
Waddell
N
,
Lane
A
, et al
.
HER3 and downstream pathways are involved in colonization of brain metastases from breast cancer
.
Breast Cancer Res
2010
;
12
:
R46
.
18.
Momeny
M
,
Saunus
JM
,
Marturana
F
,
McCart Reed
AE
,
Black
D
,
Sala
G
, et al
.
Heregulin-HER3-HER2 signaling promotes matrix metalloproteinase-dependent blood-brain-barrier transendothelial migration of human breast cancer cell lines
.
Oncotarget
2015
;
6
:
3932
46
.
19.
Achrol
AS
,
Rennert
RC
,
Anders
C
,
Soffietti
R
,
Ahluwalia
MS
,
Nayak
L
, et al
.
Brain metastases
.
Nat Rev Dis Prim
2019
;
5
:
5
.
20.
Le Rhun
E
,
Guckenberger
M
,
Smits
M
,
Dummer
R
,
Bachelot
T
,
Sahm
F
, et al
.
EANO–ESMO clinical practice guidelines for diagnosis, treatment and follow-up of patients with brain metastasis from solid tumours
.
Ann Oncol
2021
;
32
:
1332
47
.
21.
Brastianos
PK
,
Carter
SL
,
Santagata
S
,
Cahill
DP
,
Taylor-Weiner
A
,
Jones
RT
, et al
.
Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets
.
Cancer Discov
2015
;
5
:
1164
77
.
22.
Bartsch
R
,
Berghoff
AS
,
Furtner
J
,
Marhold
M
,
Bergen
ES
,
Roider-Schur
S
, et al
.
Trastuzumab deruxtecan in HER2-positive breast cancer with brain metastases: a single-arm, phase 2 trial
.
Nat Med
2022
;
28
:
1840
7
.
23.
Pérez-García
JM
,
Vaz Batista
M
,
Cortez
P
,
Ruiz-Borrego
M
,
Cejalvo
JM
,
de la Haba-Rodriguez
J
, et al
.
Trastuzumab deruxtecan in patients with central nervous system involvement from HER2-positive breast cancer: The DEBBRAH trial
.
Neuro Oncol
2023
;
25
:
157
66
.
24.
Kabraji
S
,
Ni
J
,
Sammons
S
,
Li
T
,
Van Swearingen
AED
,
Wang
Y
, et al
.
Preclinical and clinical efficacy of trastuzumab deruxtecan in breast cancer brain metastases
.
Clin Cancer Res
2023
;
29
:
174
82
.
25.
Jänne
PA
,
Baik
C
,
Su
WC
,
Johnson
ML
,
Hayashi
H
,
Nishio
M
, et al
.
Efficacy and safety of patritumab deruxtecan (HER3-DXd) in EGFR inhibitor-resistant, EGFR-mutated non-small cell lung cancer
.
Cancer Discov
2022
;
12
:
74
89
.
26.
Krop
IE
,
Masuda
N
,
Mukohara
T
,
Takahashi
S
,
Nakayama
T
,
Inoue
K
, et al
.
Results from the phase 1/2 study of patritumab deruxtecan, a HER3-directed antibody-drug conjugate (ADC), in patients with HER3-expressing metastatic breast cancer (MBC)
.
J Clin Oncol
40
:
16s
,
2022
(
suppl; abstr
1002
).
27.
Starzer
AM
,
Berghoff
AS
,
Hamacher
R
,
Tomasich
E
,
Feldmann
K
,
Hatziioannou
T
, et al
.
Tumor DNA methylation profiles correlate with response to anti-PD-1 immune checkpoint inhibitor monotherapy in sarcoma patients
.
J Immunother Cancer
2021
;
9
:
e001458
.
28.
Hammond
MEH
,
Hayes
DF
,
Wolff
AC
,
Mangu
PB
,
Temin
S
.
American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer
.
J Oncol Pract
2010
;
6
:
195
7
.
29.
Wolff
AC
,
Hammond
MEH
,
Schwartz
JN
,
Hagerty
KL
,
Allred
DC
,
Cote
RJ
, et al
.
American Society of Clinical Oncology/College of American Pathologists guideline recommendations for human epidermal growth factor receptor 2 testing in breast cancer
.
J Clin Oncol
2007
;
25
:
118
45
.
30.
Müller
F
,
Scherer
M
,
Assenov
Y
,
Lutsik
P
,
Walter
J
,
Lengauer
T
, et al
.
RnBeads 2.0: comprehensive analysis of DNA methylation data
.
Genome Biol
2019
;
20
:
55
.
31.
Maksimovic
J
,
Gordon
L
,
Oshlack
A
.
SWAN: subset-quantile within array normalization for illumina infinium HumanMethylation450 BeadChips
.
Genome Biol
2012
;
13
:
R44
.
32.
Metsalu
T
,
Vilo
J
.
ClustVis: a web tool for visualizing clustering of multivariate data using principal component analysis and heatmap
.
Nucleic Acids Res
2015
;
43
:
W566
70
.
33.
Ge
SX
,
Jung
D
,
Jung
D
,
Yao
R
.
ShinyGO: a graphical gene-set enrichment tool for animals and plants
.
Bioinformatics
2020
;
36
:
2628
9
.
34.
Jurmeister
P
,
Weber
K
,
Villegas
S
,
Karn
T
,
Untch
M
,
Thieme
A
, et al
.
DNA methylation profiling identifies two distinct subgroups in breast cancers with low hormone receptor expression, mainly associated with HER2 amplification status
.
Clin Epigenetics
2021
;
13
:
184
.
35.
Colucci-D'amato
L
,
Speranza
L
,
Volpicelli
F
.
Neurotrophic factor BDNF, physiological functions and therapeutic potential in depression, neurodegeneration and brain cancer
.
Int J Mol Sci
2020
;
21
:
7777
.
36.
Choy
C
,
Ansari
KI
,
Neman
J
,
Hsu
S
,
Duenas
MJ
,
Li
H
, et al
.
Cooperation of neurotrophin receptor TrkB and Her2 in breast cancer cells facilitates brain metastases
.
Breast Cancer Res
2017
;
19
:
51
.
37.
Zhang
R
,
Zhao
J
,
Zhao
L
.
TRKB acts as a prognostic predictive marker in Her-2 positive breast cancer
.
Ann Diagn Pathol
2022
;
60
:
152017
.
38.
Klemm
F
,
Bleckmann
A
,
Siam
L
,
Chuang
HN
,
Rietkötter
E
,
Behme
D
, et al
.
β-catenin-independent WNT signaling in basal-like breast cancer and brain metastasis
.
Carcinogenesis
2011
;
32
:
434
42
.
39.
Yonesaka
K
,
Tanizaki
J
,
Maenishi
O
,
Haratani
K
,
Kawakami
H
,
Tanaka
K
, et al
.
HER3 augmentation via blockade of EGFR/AKT signaling enhances anticancer activity of HER3-targeting patritumab deruxtecan in EGFR-mutated non–small cell lung cancer
.
Clin Cancer Res
2022
;
28
:
390
403
.
40.
Naidu
R
,
Yadav
M
,
Nair
S
,
Kutty
MK
.
Expression of c-erbB3 protein in primary breast carcinomas
.
Br J Cancer
1998
;
78
:
1385
90
.
41.
Barnes
NLP
,
Khavari
S
,
Boland
GP
,
Cramer
A
,
Knox
WF
,
Bundred
NJ
.
Absence of HER4 expression predicts recurrence of ductal carcinoma in situ of the breast
.
Clin Cancer Res
2005
;
11
:
2163
8
.
42.
Kusuhara
S
,
Kogawa
T
,
Shimokawa
M
,
Funasaka
C
,
Kondoh
CN
,
Harano
K
, et al
.
264P Increased membrane HER3 expression in brain metastases compared to primary tumors in breast cancer
.
Ann Oncol
2022
;
33
:
S658
.
43.
Law
AJ
,
Shannon Weickert
C
,
Hyde
TM
,
Kleinman
JE
,
Harrison
PJ
.
Neuregulin-1 (NRG-1) mRNA and protein in the adult human brain
.
Neuroscience
2004
;
127
:
125
36
.
44.
Mei
L
,
Xiong
WC
.
Neuregulin 1 in neural development, synaptic plasticity and schizophrenia
.
Nat Rev Neurosci
2008
;
9
:
437
52
.
45.
Liew
H
,
Kim
Y-M
,
Soon Choi
H
,
Ram Jang
A
,
Churchill
D
,
Hyung Lee
S
, et al
.
Soluble neuregulin-1 from microglia enhances amyloid beta-induced neuronal death
.
CNS Neurol Disord Drug Targets
2016
;
15
:
918
26
.
46.
Gil
V
,
Miranda
S
,
Riisnaes
R
,
Gurel
B
,
D'Ambrosio
M
,
Vasciaveo
A
, et al
.
HER3 is an actionable target in advanced prostate cancer
.
Cancer Res
2021
;
81
:
6207
18
.
47.
Koyama
K
,
Ishikawa
H
,
Abe
M
,
Shiose
Y
,
Ueno
S
,
Qiu
Y
, et al
.
Patritumab deruxtecan (HER3-DXd), a novel HER3 directed antibody drug conjugate, exhibits in vitro activity against breast cancer cells expressing HER3 mutations with and without HER2 overexpression
.
PLoS One
2022
;
17
:
e0267027
.
48.
Pascual
T
,
Oliveira
M
,
Ciruelos
E
,
Bellet Ezquerra
M
,
Saura
C
,
Gavilá
J
, et al
.
SOLTI-1805 TOT-HER3 study concept: a window-of-opportunity trial of patritumab deruxtecan, a HER3 directed antibody drug conjugate, in patients with early breast cancer
.
Front Oncol
2021
;
11
:
638482
.
49.
Modi
S
,
Jacot
W
,
Yamashita
T
,
Sohn
J
,
Vidal
M
,
Tokunaga
E
, et al
.
Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer
.
N Engl J Med
2022
;
387
:
9
20
.
50.
Yonesaka
K
,
Takegawa
N
,
Watanabe
S
,
Haratani
K
,
Kawakami
H
,
Sakai
K
, et al
.
An HER3-targeting antibody–drug conjugate incorporating a DNA topoisomerase I inhibitor U3–1402 conquers EGFR tyrosine kinase inhibitor-resistant NSCLC
.
Oncogene
2019
;
38
:
1398
409
.
51.
Desai
A
,
Abdayem
P
,
Adjei
AA
,
Planchard
D
.
Antibody-drug conjugates: a promising novel therapeutic approach in lung cancer
.
Lung Cancer
2022
;
163
:
96
106
.
52.
Salgado
R
,
Denkert
C
,
Campbell
C
,
Savas
P
,
Nuciforo
P
,
Aura
C
, et al
.
Tumor-infiltrating lymphocytes and associations with pathological complete response and event-free survival in HER2-positive early-stage breast cancer treated with lapatinib and trastuzumab: a secondary analysis of the NeoALTTO trial
.
JAMA Oncol
2015
;
1
:
448
55
.
53.
Luen
SJ
,
Salgado
R
,
Fox
S
,
Savas
P
,
Eng-Wong
J
,
Clark
E
, et al
.
Tumour-infiltrating lymphocytes in advanced HER2-positive breast cancer treated with pertuzumab or placebo in addition to trastuzumab and docetaxel: a retrospective analysis of the CLEOPATRA study
.
Lancet Oncol
2017
;
18
:
52
62
.
54.
Denkert
C
,
von Minckwitz
G
,
Darb-Esfahani
S
,
Lederer
B
,
Heppner
BI
,
Weber
KE
, et al
.
Tumour-infiltrating lymphocytes and prognosis in different subtypes of breast cancer: a pooled analysis of 3771 patients treated with neoadjuvant therapy
.
Lancet Oncol
2018
;
19
:
40
50
.
55.
Wang
Y
,
Yang
H
,
Duan
G
,
Wang
Y
,
Yang
H
,
Duan
G
.
HER3 over-expression and overall survival in gastrointestinal cancers
.
Oncotarget
2015
;
6
:
42868
78
.
56.
Li
Q
,
Zhang
R
,
Yan
H
,
Zhao
P
,
Wu
L
,
Wang
H
, et al
.
Prognostic significance of HER3 in patients with malignant solid tumors
.
Oncotarget
2017
;
8
:
67140
51
.
57.
Chiu
CG
,
Masoudi
H
,
Leung
S
,
Voduc
DK
,
Gilks
B
,
Huntsman
DG
, et al
.
HER-3 overexpression is prognostic of reduced breast cancer survival: a study of 4046 patients
.
Ann Surg
2010
;
251
:
1107
16
.
58.
Bae
SY
,
La Choi
Y
,
Kim
S
,
Kim
M
,
Kim
J
,
Jung
SP
, et al
.
HER3 status by immunohistochemistry is correlated with poor prognosis in hormone receptor-negative breast cancer patients
.
Breast Cancer Res Treat
2013
;
139
:
741
50
.
59.
Berghoff
AS
,
Bartsch
R
,
Preusser
M
,
Ricken
G
,
Steger
GG
,
Bago-Horvath
Z
, et al
.
Co-overexpression of HER2/HER3 is a predictor of impaired survival in breast cancer patients
.
Breast
2014
;
23
:
637
43
.
60.
Lee
Y
,
Cho
S
,
Jae
HS
,
Bong
KS
,
Han
KK
,
Kim
I
, et al
.
Correlated expression of erbB-3 with hormone receptor expression and favorable clinical outcome in invasive ductal carcinomas of the breast
.
Am J Clin Pathol
2007
;
128
:
1041
9
.