Next Article in Journal
Dexamethasone Suppression Testing in a Contemporary Cohort with Adrenal Incidentalomas in Two U.S. Integrated Healthcare Systems
Next Article in Special Issue
Early Detection of Disease Progression in Metastatic Cancers: Could CTCs Improve RECIST Criteria?
Previous Article in Journal
Comparison of Targeted Biopsy and Combined Biopsy to Avoid Unnecessary Systematic Biopsy in Patients with PI-RADS 5 Lesions
Previous Article in Special Issue
Role of Fork-Head Box Genes in Breast Cancer: From Drug Resistance to Therapeutic Targets
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Integrated Molecular Characterization of HER2-Low Breast Cancer Using Next Generation Sequencing (NGS)

1
Biopathology Department, Institut de Cancérologie de Lorraine—Alexis Vautrin, CNRS UMR7039 CRAN Université de Lorraine, 6 Avenue de Bourgogne, 54519 Vandœuvre-lès-Nancy, France
2
Methodology Biostatistics Unit, Institut de Cancérologie de Lorraine—Alexis Vautrin, 54519 Vandoeuvre-lès-Nancy, France
3
Medical Oncology Department, Institut de Cancérologie de Lorraine—Alexis Vautrin, 54519 Vandœuvre-lès-Nancy, France
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(12), 3164; https://doi.org/10.3390/biomedicines11123164
Submission received: 7 November 2023 / Revised: 17 November 2023 / Accepted: 26 November 2023 / Published: 28 November 2023

Abstract

:
Based on immunohistochemistry (IHC) and in situ hybridization (ISH), HER2-low breast cancers (BC) subtype—defined as IHC1+ or IHC2+/ISH− tumors—emerged and represent more than half of all BC. We evaluated the performance of NGS for integrated molecular characterization of HER2-low BC, including identification of actionable molecular targets, copy number variation (CNV), and microsatellite instability (MSI) analysis. Thirty-one BC specimens (11 HER2+, 10 HER2−, and 10 HER2-low) were routinely analyzed using IHC and ISH, and were selected and analyzed using NGS for gene mutations including ESR1, PIK3CA, AKT1, ERBB2, TP53, BRCA1, and BRCA2, CNV, and MSI. CNV values for the ERBB2 gene were significantly (p < 0.001) different between HER2+, and either HER2-low or HER2− tumors with mean values of 7.8 (SD = 6.8), 1.9 (SD = 0.3), and 2.0 (SD = 0.3), respectively. Using 3.25 as the cutoff value, 96.8% overall concordance of HER2 status was achieved between IHC and NGS compared to IHC and ISH. Using NGS, gene mutations and amplifications were detected in 68% (21/31) and 19% (6/31) of the cases, respectively. One case of MSI was detected in a HER2-negative and ISH unamplified case. Beside IHC, NGS allows the identification of HER2-low subtype simultaneously, with the detection of multiple actionable gene mutations being helpful for molecular board treatment selection.

1. Introduction

Together with hormone receptors (HR) status, based on estrogen (ER) and progesterone receptor (PR) expression, HER2 status is standard for the diagnosis of breast cancer (BC). It is evaluated using immunohistochemistry (IHC), and the expression of HER2 protein, encoded by ERBB2 gene, is scored on a scale of 0 to 3+ depending on staining intensity and the percentage of tumor cells positive for HER2 [1]. In cases with intermediate overexpression (HER2 2+) obtained by IHC, in situ hybridization (ISH) is used to detect the amplification of the HER2 gene using fluorescent or chromogenic probes which bind to the ERBB2 gene. As a reference, labeling of chromosome 17 centromere (CEP17) is performed and tumors are declared ISH positive when a high level of gene amplification is detected (ERBB2/CEP17 ratio > 2.0).
As illustrated by Figure 1, HER2-positive status is based on strong (3+) or intermediate (2+) IHC overexpression with gene amplification (ISH+). Ten to 20% of BC are HER2-positive [2,3], and patients with HER2-positive BC are eligible for targeted therapies directed against HER2 protein; either monoclonal antibodies (MAbs) such as trastuzumab and pertuzumab, or kinase inhibitors such as lapatinib and, more recently, antibody–drug conjugates (ADC) such as trastuzumab deruxtecan.
There is growing interest in understanding the importance of low HER2 expression in BC, defined as IHC1+ or 2+ and ISH-negative (HER2/CEP17 ratio < 2.0), as recently highlighted in the last update of the guidelines of the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) [4].
However, identification of HER2-low BC can be equivocal. Recently, the results of a multi-institutional assessment of pathologists scoring HER2 using immunohistochemistry were reported. Using the 4-category HER2 IHC scoring system [5], substantial discordance within the intermediate categories (IHC1+ and IHC2+) was observed with less than 1% and 3.6% agreement, respectively. Within the IHC score of 0 cases, the discordance was also substantial, with an overall agreement of only 25%. Adjustment of the criteria has been proposed to reduce the interobserver variability, suggesting that the modified 2007 ASCO/CAP criteria were more reproducible in distinguishing HER2-negative from HER2-low cases than the 2018 ASCO/CAP criteria [6]. Even in such conditions, the reproducibility was not fully adequate and was not improved by adding ISH. Since the threshold between HER2 IHC 0 and 1+ is now becoming clinically actionable, this could lead to a suboptimal identification of patients eligible for HER2-low targeted therapies. Since differentiating between the HER2 IHC score 0 and 1 + has not been clinically significant until recently, the updated recommendations for the standardized pathology report for HER2 testing were published in compliance with 2023 ASCO/CAP updates and 2023 ESMO consensus statements [7]. These recommendations mainly include the adoption of standardized procedures and guidelines, the promotion of specialized training for pathologists in interpreting HER2 expression, specifically in the lower expression level, and the development of artificial intelligence-based procedures.
The rate of HER2-low BC is about 40–65% of HR+ tumors, and 23–40% of HR-negative tumors express low levels of HER2 [8]. HER2-low cancers are reported to be more frequently observed in clinical stages I–II, ER+, ductal, histologic grade 1 or 2, and luminal molecular subtypes BC. When compared to HER2− (IHC 0) and HER2+ (IHC3+ or IHC2+ ISH amplified) BC, HER2-low BC had lower Ki-67 and higher ER positivity [9,10]. In a recent paper [11] examining 12,965 BC patients, among which 635 were stage IV patients, HER2-low BC represented more than half of both cohorts (59% overall, 53% stage IV). HER2-low BC were more frequently HR-positive than IHC 0 BC. Further, no survival differences were found between HER2-low and IHC 0 among stage IV patients.
HER2 expression is dynamic, with a higher conversion rate from HER2-low to HER2− than the reverse, and discordant between primary tumors and advanced disease [12]. These results highlight that, when feasible and safe, it is useful to obtain repeated biopsies at the time of disease progression, as this may affect treatment options.
The first HER2-targeting therapy that was widely studied for HER2-low disease was trastuzumab. The NSABP B-47 trial, a randomized phase 3 trial including 3270 women with high-risk and HER2-low BC comparing chemotherapy with or without trastuzumab [13], showed no difference in invasive disease-free survival between the groups. Later, a phase 2 study showed little benefit with pertuzumab [14] for the treatment of patients with HER2-low BC. Based on these results, the use of anti-HER2 remained limited to patients with HER2+ tumors, until the development of HER2-directed antibody–drug conjugates (ADC-HER2) such as trastuzumab deruxtecan (T-DXd).
ADC-HER2s consist of a monoclonal antibody that delivers a chemotherapy molecule directly to HER2-expressing tumor cells, resulting in the targeted killing of cancer cells while minimizing damage to healthy cells [15]. Historically, the first ADC-HER2 put on the market was trastuzumab emtansine or TDM-1, whose interest in HER2+ breast cancers has been widely demonstrated whether in metastatic [16,17,18], adjuvant [19,20], or neoadjuvant [21] settings.
Trastuzumab deruxtecan (T-DXd) is an ADC composed of a HER2-targeting monoclonal antibody, a cleavable ligand. The cytotoxic payload is deruxtecan, a potent topoisomerase 1 inhibitor. T-DXd has a high drug-to-antibody ratio of 8:1 [22]. Results from phase II [23] and phase III [24] clinical trials have shown the efficacy of T-DXd in patients with metastatic HER2+ breast cancer, with a higher response rate, longer PFS, and higher survival rate at 1 year than TDM-1 [25].
Recently, the results of the phase 2 DAISY trial [26], evaluating the efficacy of T-DXd in patients with HER2+, HER2-low, and HER2- metastatic BC revealed that, although T-DXd efficacy increased when HER2 expression was high, a mild activity was also observed in patients with HER2− tumors. This trial highlighted that although HER2 expression is a determinant of T-DXd efficacy, very low levels of HER2 could drive T-DXd cellular uptake and/or other mechanisms independent of HER2, which may also be partially involved.
Beside HR and HER2 status, additional molecular targets are emerging in BC, revealing the need for extensive molecular diagnosis to envisage precision therapy. ESMO scale for actionability of molecular targets (ESCAT) ranges 6 classes based on the level of evidence currently available (Table 1), and the clinical indication of actionability in a specific cancer type to assist oncologists in making a therapeutic decision [27]. In this scale, the genomic alterations are ranked according to their clinical benefit for patients. Until now, in BC, only three alterations have been reported to provide a clinical benefit in prospective randomized trials: HER2 amplification, PIK3CA mutations, and BRCA1 and BRCA2 germline mutations.
Lastly, beside the gene mutation and amplification profiles of BC, MSI status is also gaining in interest as a biomarker for immunotherapy [28]. Many clinical trials are actually evaluating the interest of combining immunotherapy with either endocrine therapy in HR+ BC, or anti-HER2 therapies in HER2+ BC [29].
The present study is a proof of concept, retrospective study of integrated NGS characterization of HER2 low tumors, evaluating the feasibility of using NGS for determination of the ERBB2 gene amplification in BC tumor specimens that have already undergone routine IHC for ER, PR, Ki67, and HER2 expression. This study also focuses on the potency of NGS analysis to identify simultaneously additional actionable molecular targets as well as MSI. The routine (Figure 1A) and the experimental (Figure 1B) workflows are illustrated in Figure 1.

2. Material and Methods

2.1. Tumor Specimens

Thirty-one FFPE BC tumor specimens were selected from the institutional tumor bank of the Lorraine Cancer Institute (authorization nr. DC-2013-1910 from the French Ministry of Research). According to the French regulation (art L-1211-2 of the Public Health Code, law nr. 2004-800, 6 August 2004).

2.2. Routine Diagnostic Biomarkers Analyses

The tumors were fixed in 4% buffered aqueous formaldehyde for at least 12 h (biopsies) or 24 h (surgical specimens), then paraffin-embedded. Five µm slides were then prepared, and BC tumors specimens were stained by hematoxylin and eosin (CoverStainer Dako, Agilent Technologies, Les Ulis, France). CK19 (clone RCK 108, Agilent Technologies), estrogen receptor (ER, clone SP1, Ventana Roche Diagnostics, Meylan, France), progesterone receptor (PR, clone 1E2, Ventana Roche Diagnostics), HER2 (clone 4B5, Ventana Roche Diagnostics), and Ki-67 (clone MIB-1, Dako, Agilent Technologies) expression were determined using immunohistochemistry. All IHC assays were automated (Benchmark Ultra Ventana, Roche Diagnostics or Dako Omnis, Agilent Technologies) and processed according to the manufacturer’s recommendations.
The routine diagnostic analyses for BC were performed as recommended by the ASCO-CAP [30] and the French Unicancer GEFPICS group «Groupe d’Etude des Facteurs Pronostiques Immunohistochimiques dans le Cancer du Sein» [31].
The histopathological grading of the BC was performed according to the classification of the Scarff–Bloom–Richardson modified score. According to the recommendations of the GEFPICS [31], the scoring criteria that were used exactly followed the international recommendations. The threshold for positivity of ER and PR expression was set at 10% of labelled cells. Ki-67 expression was expressed as a percentage of the labelled cells. HER2-positive status was defined on the basis of complete intense membrane staining of more than 10% of the tumor cells (3+) or intermediate IHC overexpression (2+), defined as weak–moderate membrane staining in >10% tumor cells or intense membrane staining in <10% tumor cells with gene amplification (ISH+). IHC2+ tumors were analyzed using the IVD HER2 Dual ISH DNA Assay (D-DISH) (Ventana, Roche Diagnostics) to detect the amplification of the ERBB2 gene using chromogenic probes that bind to the ERBB2 gene. As a reference, labeling of chromosome 17 centromere (Chr17) was performed. According to the ASCO/CAP and GEFPICS recommendations, the amplification status is defined as amplified if the ERBB2/Chr17 ratio > 2.0, and as non-amplified if the HER2/Chr17 ratio < 2.0. If the ERBB2/Chr17 ratio falls between 1.8 to 2.2, an additional 20 nuclei should be enumerated, and the ratio re-calculated on the basis of all 40 nuclei.

2.3. Next Generation Sequencing

FFPE tissues were macrodissected after hematoxylin-eosin slide examination and after the tumor content was assessed by an experienced pathologist. DNA was then extracted using the QIAamp GeneRead DNA FFPE tissue kit (Qiagen, Courtaboeuf, France), and DNA concentrations were measured using the Qubit dsDNA HS assay kit and Qubit 3.0 Fluorometer instrument (ThermoFisher Scientific, Illkirch, France) according to the manufacturer’s recommendations. DNA quality was evaluated by electrophoretic profiles analysis using the high sensitivity large fragment analysis kit (Fragment Analyzer, Agilent Technologies). Genomic DNA Quality Number (GQN) score was calculated for each sample, corresponding to the proportion of DNA fragments with a size greater than the threshold set at 300 bp. Highly degraded DNA have a GQN close to 0, indicating that none of the fragments exceeds the threshold. Conversely, a good quality DNA have a GQN close to 10, with 100% of the DNA fragments being above the threshold value. However, considering the preciousness of the samples, no threshold was applied for GQN, and GQN values were considered during the post-analytical procedure, among all other quality criteria, to validate the results.
NGS libraries were prepared from 100 ng of extracted tumour DNA using a custom-designed, capture-based “Solid Tumour Solution” kit (Sophia Genetics, Geneva, Switzerland) that covers the regions of clinical interest of 51 cancer-associated genes as described elsewhere [32], among which are BC molecular target genes such as ESR1, PIK3CA, AKT1, ERBB2, TP53, BRCA1, and BRCA2. Targeted sequencing was performed using the MiSeq sequencer (Illumina). Generated raw NGS data were analyzed using the Sophia DDM software (v.5.10.37) (Sophia Genetics). Briefly, the bioinformatics pipeline (v. 5.5.79) consists of an alignment of the fastq files to generate bam files (hg19 reference genome), followed by a variant calling for the determination of SNV and indels. All results are finally available in the Sophia DDM software as retained variants and low-confidence variants. Quality control limits were required for each sample tested by NGS: >1 M reads/sample, >95% mapped reads, >70% on target, >85% at 500× depth. All pathogenic (class 5) and likely pathogenic (class 4) mutations detected among the 51 gene panel were reported. Copy Number Variation (CNV) was also calculated using CNV module from Sophia DDM software working in the gene amplification mode, i.e., only whole-gene duplications with a sufficiently large average copy number were reported. The threshold for gene amplification was set at 3.25, as recommended by the supplier of Sophia DDM (Sophia Genetics). Deletions and CNVs affecting only parts of genes were ignored. The gene amplification analysis included 24 genes (NRAS, ALK, SF3B1, RAF1, PIK3CA, FGFR3, PDGFRA, KIT, FBXW7, TERT, ROS1, EGFR, MET, BRAF, FGFR1, CDKN2A, RET, FGFR2, HRAS, MYOD1, KRAS, CDK4, TP53, ERBB2). The coverage levels used for gene amplification detection included a linear GC correction deduced from all of the regions included in the panel. MSI was determined using an algorithm module provided by Sophia DDM software based on reads analysis at 6 unique MS regions, for which 3 were split into forward (FWD) and reverse (REV) strands (BAT25_FWD, BAT25_REV, BAT26_REV, CAT25_REV, NR-21_FWD, NR-21_REV, NR-22_FWD, NR-22_REV, and NR-27_REV), as previously validated in FFPE tissues from endometrial and colon cancers [32].

2.4. Statistical Analysis

The sample size was calculated consistently with the recommendations of the French Committee for Accreditation (COFRAC Comité Français d’accréditation) in its technical guidelines for accreditation and validation of methods in medical biology (Guide technique d’accréditation de validation des méthodes de biologie médicale SH GTA 04—Revision 02 https://tools.cofrac.fr/documentation/sh-gta-04 (accessed on 15 November 2022)). The sample size of this study was justified considering the Cohen’s effect size using the pairwise and unpaired comparison test. In such conditions, sample groups (HER2+ vs. HER2-low and HER2+ versus HER2−) were compared to identify a significant standardized difference. The ranges for Cohen’s effect size were, respectively, 0.10 < d < 0.30 (small effect), 0.30 < d < 0.50 (moderate effect), and d ≥ 0.50 (large effect). Data were compared using the non-parametric Wilcoxon rank sum test. Statistical analyses were performed using R-studio software (version 2022.07.2+576) with rstatix and Wilcox test () functions. A p-value < 0.05 was used as a significativity limit.

3. Results

Thirty-one FFPE BC tumor specimens were selected from the tumor bank based on IHC and ISH results of HER2 status obtained during routine first line BC testing, together with hormone receptors (ER and PR) and Ki67 IHC analyses.
Among the 31 tumors, 5 were classified as grade 1 (SBR-EE classification), 17 as grade 2, and 9 as grade 3. Most of the tumors were hormone receptor positive (29/31), defined as positive for ER or PR expression with 29/31 ER+ and 23/31 PR+. All tumors had ER/PR expression above 10%, therefore making no difference between ASCO/CAP and GEFFPICS classification.
HER2 IHC results show that 10/31 tumors were IHC 0, 5 IHC1+, 9 IHC2+, and 7 IHC3+. The 9 HER2 IHC2+ tumors were further analyzed using ISH, and HER2 amplification was observed in 4/9 tumors (44%). In total, the analyzed BC population consisted of 10 HER2 negative, 10 HER2-low, and 11 HER2 positive tumors. HER2-negative and HER2-low tumors were mostly grade 2 tumors (6/10 in HER2-negative and 7/10 in HER2-low), as opposed to HER2-positive tumors that were mostly composed of grade 3 tumors (7/11).
KI67 labeling ranged from 2 to 80%, and was significantly higher (p < 0.001) in HER2-positive (IHC3+ or IHC2+ and ISH+) than in HER2-negative (IHC0) and HER2-low (IHC1+ or IHC2+, ISH−) BC, with mean values of 34.2 (SD = 19.2), 15.8 (SD = 13.4) and 13.5 (SD = 6.5), respectively (Figure 2A).
All analyzed macrodissected samples had tumor cell content exceeding 10% (range 15–80%) after examination of HE-stained slides by an expert pathologist. No difference was observed between the different groups.
Using NGS, CNV values for the ERBB2 gene were significantly different (p < 0.001) between HER2-positive, either HER2-low or HER2-negative tumors with mean values of 7.8 (SD = 6.8), 1.9 (SD = 0.3), and 2.0 (SD = 0.3), respectively (Figure 2B). With a cutoff value for CNV set at 3.25, HER2 amplification status was consistent between ISH and NGS in all but one sample (sample #24). This sample has discordant results with a CNV value of 2.9, while having a weak positive HER2/Chr17 ISH ratio of 2.2 and a tumor cell content of 40%, close to the threshold values. In addition, the DNA extract from this sample had a low GQN value of 1.5, illustrating a low quality DNA that could explain the outlying. As a whole, the results for HER2 status were achieved following the experimental workflow (IHC combined with NGS produced results that were consistent with those achieved using the reference workflow (IHC combined with ISH) in 30/31 cases i.e., 96.8%).
Considering 21 samples for comparison of HER2+ (N = 11) and HER2− (N = 10) or HER2+ (N = 11) and HER2-low (N = 10), evidenced in each case was an effect size of 0.85 (large effect), therefore validating the sample size.
Additional gene amplifications were simultaneously detected in 19% (6/31) of the cases, either in HER2-negative tumors (2 cases with FGFR1 amplifications and 2 with MYOD1 amplifications) and in HER2-positive tumors (1 case with FGFR1 amplification, 1 with PIK3CA amplification and 1 with ROS1amplification) (Supplementary Figure S1).
Additional pathogenic and likely pathogenic gene mutations were detected among the 51 gene panel in 68% (21/31) of the cases with 12 PIK3CA, 6 TP53, 2 AKT1, and 4 BRCA2 mutations.
Further, one case of MSI status was detected in a HER2-negative, ISH unamplified, HR-positive tumor bearing a PIK3CA gene mutation (c.1624G > A, p.(Glu542Lys)).
All data are summarized in Table 2.

4. Discussion

Based on the expression of hormone receptors (HR) and HER2, BC used to be classified into four intrinsic subtypes: luminal A (HR+ and HER2−), luminal B (HR+ and HER2+), HER2-enriched (HR− and HER2+), and triple-negative BC (TNBC, HR− and HER2−) [33], among which TNBC is generally considered to have the worst prognosis.
The identification of HER2-low BC has recently reached great clinical significance [34]. Usually reported as HER2-negative and classified as TNBC or luminal-like in clinical practice, HER2-low BC is a heterogeneous disease and shows distinct molecular profile and prognosis compared with tumors negative for HER2 by IHC [35].
However, since negative results have been reported with the addition of trastuzumab or pertuzumab to chemotherapy in patients with HER2-low BC [13], HER2-low BC were not eligible for anti-HER2 monoclonal antibodies.
The DESTINY-Breast04 phase 3 trial revealed the efficacy of T-DXd in HER2-low metastatic breast cancers previously treated with one or two lines of chemotherapy. As compared to physician-selected chemotherapy, T-DXd significant increased PFS and OS in the HR+ cohort [36].
In the DAISY trial [26], even very low levels of HER2 expression appeared to drive T-DXd cellular uptake and other mechanisms independent of HER2, and were suggested to be also partially involved in the efficacy of T-DXd.
The identification of HER2-low status is therefore becoming crucial in the management of BC, but remains challenging due to methodological and analytical variables that might influence the sensitivity and reproducibility of HER2 IHC and ISH routine testing. Additionally, the heterogeneity of HER2 expression and/or amplification should be considered for improving HER2-low identification. Since HER2 heterogeneity could be seen in both HER2 gene copy number and/or protein expression, guidelines have been updated by the ASCO/CAP [4], but the consensus to define HER2 heterogeneity for IHC is still lacking [37,38]. This sounds particularly important since HER2 IHC heterogeneity has been reported to be more frequent in the HER2-low samples [39] and might contribute to variability of HER2-low interpretation, and consequently influence the response to ADCs [40].
The development of NGS technologies has raised hopes for precision medicine treatment strategies in breast cancer (BC) and triple-negative breast cancer. Multiple additional molecular targets can now be screened in BC, as illustrated by the ESCAT classification [27]. Alterations that have been recently added in international guidelines [41] include the ESR1 gene mutations and those that still need confirmatory prospective studies, but for which evidence of efficacy is available, including the PIK3CA, AKT1, HER2, TP53, BRCA1, and BRCA2 gene mutations that were included in the custom capture-based 51-gene NGS panel used in this study.
Besides mutations, the identification of gene amplifications is of increasing interest for diagnostic, pronostic, and theranostic purposes in BC. This is the case for some amplifications that were identified in our study. MYOD1 amplification was reported to be of pronostic interest in BC, with the greatest prevalence in breast invasive lobular carcinoma as well as ROS1 amplification [40]. FGFR1 amplification was reported to be associated with bad prognosis in hormone-positive BC and resistance to hormonal monotherapy, or in combination with palbociclib. Resistances can be reverted with triple ER, CDK4/6, and FGFR1 blockade [42]. PIK3CA mutations and amplifications have been reported in approximately 10% of HR-positive and HER2-negative primary BC and among patients with PIK3CA-mutated BC; those with amplified PIK3CA have worse outcomes [43].
Finally, MSI status can serve as a biomarker to initiate immunotherapy in TNBC and also in HER2-negative HR-positive metastatic BC [28]. In this context, although MSI status is detected in a low proportion of BC [44], its simultaneous determination using NGS can be valuable to allow the initiation of immunotherapy, alone or in combination with endocrine therapy in clinical trials upon molecular board decision [29].
Facing the availability of tumor tissue in limited quantity, which is very often already consumed by multiple IHC and ISH procedures, molecular pathologists are often confronted to choose between rapid PCR-based diagnostic with short time-to-results (TTR) delay (1–3 days in routine practice) limited to a relatively small number of target genes. In such a context, extensive NGS with large gene panels in the several hundreds can prove very helpful, although generating a longer TTR (1–3 weeks in routine practice). While optimizing the use of small biopsies, using NGS could allow for the selection of innovative therapeutic options based on molecular tumor board decision guided by the ESCAT scale [27], and/or to propose to the patient that they be included in a clinical trial [45].
In the present study we show that, following first-line IHC routine analysis alone, NGS would provide biomarkers and identify multiple targets as well as MSI status, which is helpful in HER2-low and TNBC. Using NGS can spare biological material and time, and can prove useful for tumor stratification upon molecular board. This study warrants further confirmation in a larger patient sample population.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/biomedicines11123164/s1.

Author Contributions

Conceptualization, J.-L.M., A.H. and M.H.; Methodology, N.S.; Validation, J.-L.M., M.H., P.G. and V.M.; Formal Analysis, J.-L.M. and N.S.; Investigation, J.-L.M. and M.H.; Data Curation, J.-L.M. and M.H.; Writing—Original Draft Preparation, J.-L.M.; Writing—Review & Editing, J.-L.M., M.H., P.G., A.L., A.H. and V.M.; Supervision, J.-L.M.; Project Administration, J.-L.M.; Funding Acquisition, J.-L.M. and V.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by a research grant from Seagen France (Rueil Malmaison France) and the private research funds of the Institut de Cancérologie de Lorraine.

Institutional Review Board Statement

This study was approved by the Institutional Review Board of the Institut de Cancérologie de Lorraine (Comité Scientifique Interne) on 16 November 2021, decision 20221116-5.

Informed Consent Statement

All patients were duly informed and signed a statement of non-objection to storage and use of their tumor tissue specimens for translational research purposes.

Data Availability Statement

The data presented in this study are available on reasonable request from the corresponding author. The data are not publicly available due to ethical reasons.

Conflicts of Interest

J.-L.M. received honoraria, financial support for meetings and research grants from Seagen and Astra Zeneca, A.H. received honoraria, financial support for meetings and research grants from Seagen, Astra Zeneca and Sophia Genetics, V.M. received honoraria, financial support for meetings and research grants from Seagen and Astra Zeneca. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest These results were presented at the 2023 European Society of Medical Oncology meeting in Madrid, Spain.

References

  1. Deluche, É.; Vincent-Salomon, A. «HER2-faible», un nouveau concept dans la prise en charge des cancers du sein: HER2-low breast cancer: A new concept in breast cancer treatment strategy. Bull. Cancer 2021, 108, 11S1–11S7. [Google Scholar] [CrossRef] [PubMed]
  2. Cronin, K.A.; Harlan, L.C.; Dodd, K.W.; Abrams, J.S.; Ballard-Barbash, R. Population-based Estimate of the Prevalence of HER-2 Positive Breast Cancer Tumors for Early Stage Patients in the US. Cancer Investig. 2010, 28, 963–968. [Google Scholar] [CrossRef] [PubMed]
  3. Slamon, D.J.; Clark, G.M.; Wong, S.G.; Levin, W.J.; Ullrich, A.; McGuire, W.L. Human breast cancer: Correlation of relapse and survival with amplification of the HER-2/neu oncogene. Science 1987, 235, 177–182. [Google Scholar] [CrossRef] [PubMed]
  4. Wolff, A.C.; Somerfield, M.R.; Dowsett, M.; Hammond, M.E.H.; Hayes, D.F.; McShane, L.M.; Saphner, T.J.; Spears, P.A.; Allison, K.H. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology-College of American Pathologists Guideline Update. Arch. Pathol. Lab. Med. 2023, 147, 993–1000. [Google Scholar] [CrossRef] [PubMed]
  5. Robbins, C.J.; Fernandez, A.I.; Han, G.; Wong, S.; Harigopal, M.; Podoll, M.; Singh, K.; Ly, A.; Kuba, M.G.; Wen, H.; et al. Multi-institutional Assessment of Pathologist Scoring HER2 Immunohistochemistry. Mod. Pathol. 2023, 36, 100032. [Google Scholar] [CrossRef]
  6. Baez-Navarro, X.; van Bockstal, M.R.; Nawawi, D.; Broeckx, G.; Colpaert, C.; Doebar, S.C.; Hogenes, M.C.; Koop, E.; Lambein, K.; Peeters, D.J.; et al. Interobserver Variation in the Assessment of Immunohistochemistry Expression Levels in HER2-Negative Breast Cancer: Can We Improve the Identification of Low Levels of HER2 Expression by Adjusting the Criteria? An International Interobserver Study. Mod. Pathol. 2023, 36, 100009. [Google Scholar] [CrossRef] [PubMed]
  7. Ivanova, M.; Porta, F.M.; D’ercole, M.; Pescia, C.; Sajjadi, E.; Cursano, G.; De Camilli, E.; Pala, O.; Mazzarol, G.; Venetis, K.; et al. Standardized pathology report for HER2 testing in compliance with 2023 ASCO/CAP updates and 2023 ESMO consensus statements on HER2-low breast cancer. Virchows Arch. 2023. Epub ahead of print. [Google Scholar] [CrossRef]
  8. Tarantino, P.; Jin, Q.; Tayob, N.; Jeselsohn, R.M.; Schnitt, S.J.; Vincuilla, J.; Parker, T.; Tyekucheva, S.; Li, T.; Lin, N.U.; et al. Prognostic and Biologic Significance of ERBB2-Low Expression in Early-Stage Breast Cancer. JAMA Oncol. 2022, 8, 1177–1183. [Google Scholar] [CrossRef]
  9. Zhang, H.; Katerji, H.; Turner, B.M.; Audeh, W.; Hicks, D.G. HER2-low breast cancers: Incidence, HER2 staining patterns, clinicopathologic features, MammaPrint and BluePrint genomic profiles. Mod. Pathol. 2022, 35, 1075–1082. [Google Scholar] [CrossRef]
  10. Schettini, F.; Chic, N.; Brasó-Maristany, F.; Paré, L.; Pascual, T.; Conte, B.; Martínez-Sáez, O.; Adamo, B.; Vidal, M.; Barnadas, E.; et al. Clinical, pathological, and PAM50 gene expression features of HER2-low breast cancer. NPJ Breast Cancer 2021, 7, 1, Erratum in NPJ Breast Cancer 2023, 9, 32. [Google Scholar] [CrossRef]
  11. Check, D.K.; Jackson, B.E.; Reeder-Hayes, K.E.; Dinan, M.A.; Faherty, E.; Kwong, J.; Mehta, S.; Spees, L.; Wheeler, S.B.; Wilson, L.E.; et al. Characteristics, healthcare utilization, and outcomes of patients with HER2-low breast cancer. Breast Cancer Res. Treat. 2023. Epub ahead of print. [Google Scholar] [CrossRef] [PubMed]
  12. Anderson, S.; Bartow, B.B.; Siegal, G.P.; Huang, X.; Wei, S. The dynamics of HER2-low expression during breast cancer progression. Breast Cancer Res. Treat. 2023, 201, 437–446. [Google Scholar] [CrossRef] [PubMed]
  13. Fehrenbacher, L.; Cecchini, R.S.; Geyer, C.E., Jr.; Rastogi, P.; Costantino, J.P.; Atkins, J.N.; Crown, J.P.; Polikoff, J.; Boileau, J.-F.; Provencher, L.; et al. NSABP B-47/NRG Oncology Phase III Randomized Trial Comparing Adjuvant Chemotherapy with or without Trastuzumab in High-Risk Invasive Breast Cancer Negative for HER2 by FISH and with IHC1+ or 2+. J. Clin. Oncol. 2020, 38, 444–453. [Google Scholar] [CrossRef] [PubMed]
  14. Gianni, L.; Lladó, A.; Bianchi, G.; Cortes, J.; Kellokumpu-Lehtinen, P.-L.; Cameron, D.A.; Miles, D.; Salvagni, S.; Wardley, A.; Goeminne, J.-C.; et al. Open-Label, Phase II, Multicenter, Randomized Study of the Efficacy and Safety of Two Dose Levels of Pertuzumab, a Human Epidermal Growth Factor Receptor 2 Dimerization Inhibitor, in Patients with Human Epidermal Growth Factor Receptor 2–Negative Metastatic Breast Cancer. J. Clin. Oncol. 2010, 28, 1131–1137. [Google Scholar] [CrossRef]
  15. Hafeez, U.; Parakh, S.; Gan, H.K.; Scott, A.M. Antibody-Drug Conjugates for Cancer Therapy. Molecules 2020, 25, 4764. [Google Scholar] [CrossRef]
  16. Verma, S.; Miles, D.; Gianni, L.; Krop, I.E.; Welslau, M.; Baselga, J.; Pegram, M.; Oh, D.-Y.; Diéras, V.; Guardino, E.; et al. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791, Erratum in N. Engl. J. Med. 2013, 368, 2442. [Google Scholar] [CrossRef]
  17. Diéras, V.; Miles, D.; Verma, S.; Pegram, M.; Welslau, M.; Baselga, J.; Krop, I.E.; Blackwell, K.; Hoersch, S.; Xu, J.; et al. Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously treated HER2-positive advanced breast cancer (EMILIA): A descriptive analysis of final overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 732–742, Erratum in Lancet Oncol. 2017, 18, e433; Erratum in Lancet Oncol. 2018, 19, e667. [Google Scholar] [CrossRef]
  18. Krop, I.E.; Kim, S.-B.; Martin, A.G.; LoRusso, P.M.; Ferrero, J.-M.; Badovinac-Crnjevic, T.; Hoersch, S.; Smitt, M.; Wildiers, H. Trastuzumab emtansine versus treatment of physician’s choice in patients with previously treated HER2-positive metastatic breast cancer (TH3RESA): Final overall survival results from a randomised open-label phase 3 trial. Lancet Oncol. 2017, 18, 743–754. [Google Scholar] [CrossRef]
  19. Von Minckwitz, G.; Huang, C.-S.; Mano, M.S.; Loibl, S.; Mamounas, E.P.; Untch, M.; Wolmark, N.; Rastogi, P.; Schneeweiss, A.; Redondo, A.; et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N. Engl. J. Med. 2019, 380, 617–628. [Google Scholar] [CrossRef]
  20. Krop, I.E.; Im, S.-A.; Barrios, C.; Bonnefoi, H.; Gralow, J.; Toi, M.; Ellis, P.A.; Gianni, L.; Swain, S.M.; Im, Y.-H.; et al. Trastuzumab Emtansine Plus Pertuzumab Versus Taxane Plus Trastuzumab Plus Pertuzumab After Anthracycline for High-Risk Human Epidermal Growth Factor Receptor 2–Positive Early Breast Cancer: The Phase III KAITLIN Study. J. Clin. Oncol. 2022, 40, 438–448. [Google Scholar] [CrossRef]
  21. Hurvitz, S.A.; Martin, M.; Jung, K.H.; Huang, C.-S.; Harbeck, N.; Valero, V.; Stroyakovskiy, D.; Wildiers, H.; Campone, M.; Boileau, J.-F.; et al. Neoadjuvant Trastuzumab Emtansine and Pertuzumab in Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer: Three-Year Outcomes From the Phase III KRISTINE Study. J. Clin. Oncol. 2019, 37, 2206–2216. [Google Scholar] [CrossRef] [PubMed]
  22. Nakada, T.; Sugihara, K.; Jikoh, T.; Abe, Y.; Agatsuma, T. The Latest Research and Development into the Antibody–Drug Conjugate, [fam-] Trastuzumab Deruxtecan (DS-8201a), for HER2 Cancer Therapy. Chem. Pharm. Bull. 2019, 67, 173–185. [Google Scholar] [CrossRef] [PubMed]
  23. Modi, S.; Saura, C.; Yamashita, T.; Park, Y.H.; Kim, S.-B.; Tamura, K.; Andre, F.; Iwata, H.; Ito, Y.; Tsurutani, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Breast Cancer. N. Engl. J. Med. 2020, 382, 610–621. [Google Scholar] [CrossRef] [PubMed]
  24. André, F.; Park, Y.H.; Kim, S.-B.; Takano, T.; Im, S.-A.; Borges, G.; Lima, J.P.; Aksoy, S.; Gregori, J.G.; De Laurentiis, M.; et al. Trastuzumab deruxtecan versus treatment of physician’s choice in patients with HER2-positive metastatic breast cancer (DESTINY-Breast02): A randomised, open-label, multicentre, phase 3 trial. Lancet 2023, 401, 1773–1785. [Google Scholar] [CrossRef] [PubMed]
  25. Cortés, J.; Kim, S.-B.; Chung, W.-P.; Im, S.-A.; Park, Y.H.; Hegg, R.; Kim, M.H.; Tseng, L.-M.; Petry, V.; Chung, C.-F.; et al. Trastuzumab Deruxtecan versus Trastuzumab Emtansine for Breast Cancer. N. Engl. J. Med. 2022, 386, 1143–1154. [Google Scholar] [CrossRef] [PubMed]
  26. Mosele, F.; Deluche, E.; Lusque, A.; Le Bescond, L.; Filleron, T.; Pradat, Y.; Ducoulombier, A.; Pistilli, B.; Bachelot, T.; Viret, F.; et al. Trastuzumab deruxtecan in metastatic breast cancer with variable HER2 expression: The phase 2 DAISY trial. Nat. Med. 2023, 29, 2110–2120. [Google Scholar] [CrossRef] [PubMed]
  27. Wolff, L.; Kiesewette, B. Applicability of ESMO-MCBS and ESCAT for molecular tumor boards. MEMO—Mag. Eur. Med. Oncol. 2022, 15, 190–195. [Google Scholar] [CrossRef]
  28. Sivapiragasam, A.; Kumar, P.A.; Sokol, E.S.; Albacker, L.A.; Killian, J.K.; Ramkissoon, S.H.; Huang, R.S.P.; Severson, E.A.; Brown, C.A.; Danziger, N.; et al. Predictive Biomarkers for Immune Checkpoint Inhibitors in Metastatic Breast Cancer. Cancer Med. 2021, 10, 53–61. [Google Scholar] [CrossRef]
  29. Debien, V.; De Caluwé, A.; Wang, X.; Piccart-Gebhart, M.; Tuohy, V.K.; Romano, E.; Buisseret, L. Immunotherapy in breast cancer: An overview of current strategies and perspectives. NPJ Breast Cancer 2023, 9, 7. [Google Scholar] [CrossRef]
  30. Allison, K.H.; Hammond, M.E.H.; Dowsett, M.; McKernin, S.E.; Carey, L.A.; Fitzgibbons, P.L.; Hayes, D.F.; Lakhani, S.R.; Chavez-MacGregor, M.; Perlmutter, J.; et al. Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Guideline Update. J. Clin. Oncol. 2020, 38, 1346–1366. [Google Scholar] [CrossRef]
  31. GEFPICS-FNCLCC. Recommendations for the immunohistochemistry of the hormonal receptors on paraffin sections in breast cancer. Update 1999. Group for Evaluation of Prognostic Factors using Immunohistochemistry in Breast Cancer (GE-FPICS-FNCLCC). Ann. Pathol. 1999, 19, 336–343, Erratum in Ann. Pathol. 1999, 19, 486. [Google Scholar]
  32. Gilson, P.; Levy, J.; Rouyer, M.; Demange, J.; Husson, M.; Bonnet, C.; Salleron, J.; Leroux, A.; Merlin, J.-L.; Harlé, A. Evaluation of 3 molecular-based assays for microsatellite instability detection in formalin-fixed tissues of patients with endometrial and colorectal cancers. Sci. Rep. 2020, 10, 16386. [Google Scholar] [CrossRef] [PubMed]
  33. Crimini, E.; Repetto, M.; Aftimos, P.; Botticelli, A.; Marchetti, P.; Curigliano, G. Precision medicine in breast cancer: From clinical trials to clinical practice. Cancer Treat. Rev. 2021, 98, 102223. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, J.; Liao, D.; Zhang, X.; Miao, C.; Chen, K. Can Patients with HER2-Low Breast Cancer Benefit from Anti-HER2 Therapies? A Review. Breast Cancer Targets Ther. 2023, 15, 281–294. [Google Scholar] [CrossRef]
  35. Berrino, E.; Annaratone, L.; Bellomo, S.E.; Ferrero, G.; Gagliardi, A.; Bragoni, A.; Grassini, D.; Guarrera, S.; Parlato, C.; Casorzo, L.; et al. Integrative genomic and transcriptomic analyses illuminate the ontology of HER2-low breast carcinomas. Genome Med. 2022, 14, 98. [Google Scholar] [CrossRef]
  36. Modi, S.; Jacot, W.; Yamashita, T.; Sohn, J.; Vidal, M.; Tokunaga, E.; Tsurutani, J.; Ueno, N.T.; Prat, A.; Chae, Y.S.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Low Advanced Breast Cancer. N. Engl. J. Med. 2022, 387, 9–20. [Google Scholar] [CrossRef]
  37. Ahn, S.; Woo, J.W.; Lee, K.; Park, S.Y. HER2 status in breast cancer: Changes in guidelines and complicating factors for interpretation. J. Pathol. Transl. Med. 2020, 54, 34–44. [Google Scholar] [CrossRef]
  38. Sajjadi, E.; Guerini-Rocco, E.; De Camilli, E.; Pala, O.; Mazzarol, G.; Venetis, K.; Ivanova, M.; Fusco, N. Pathological identification of HER2-low breast cancer: Tips, tricks, and troubleshooting for the optimal test. Front. Mol. Biosci. 2023, 10, 1176309. [Google Scholar] [CrossRef]
  39. Wu, Y.; Zhong, R.; Ma, F. HER2-low breast cancer: Novel detections and treatment advances. Crit. Rev. Oncol. 2023, 181, 103883. [Google Scholar] [CrossRef]
  40. AACR Project GENIE Consortium. AACR Project GENIE: Powering Precision Medicine through an International Consortium. Cancer Discov. 2017, 7, 818–831. [Google Scholar] [CrossRef]
  41. Burstein, H.J.; DeMichele, A.; Somerfield, M.R.; Henry, N.L.; Biomarker Testing and Endocrine and Targeted Therapy in Metastatic Breast Cancer Expert Panels. Testing for ESR1 Mutations to Guide Therapy for Hormone Receptor–Positive, Human Epidermal Growth Factor Receptor 2–Negative Metastatic Breast Cancer: ASCO Guideline Rapid Recommendation Update. J. Clin. Oncol. 2023, 41, 3423–3425. [Google Scholar] [CrossRef] [PubMed]
  42. Mouron, S.; Manso, L.; Caleiras, E.; Rodriguez-Peralto, J.L.; Rueda, O.M.; Caldas, C.; Colomer, R.; Quintela-Fandino, M.; Bueno, M.J. FGFR1 amplification or overexpression and hormonal resistance in luminal breast cancer: Rationale for a triple blockade of ER, CDK4/6, and FGFR1. Breast Cancer Res. 2021, 23, 21. [Google Scholar] [CrossRef] [PubMed]
  43. Migliaccio, I.; Paoli, M.; Risi, E.; Biagioni, C.; Biganzoli, L.; Benelli, M.; Malorni, L. PIK3CA co-occurring mutations and copy-number gain in hormone receptor positive and HER2 negative breast cancer. NPJ Breast Cancer 2022, 8, 24. [Google Scholar] [CrossRef] [PubMed]
  44. Vidula, N.; Lipman, A.; Kato, S.; Weipert, C.; Hesler, K.; Azzi, G.; Elkhanany, A.; Juric, D.; Rodriguez, E.; Faulkner, C.; et al. Detection of microsatellite instability high (MSI-H) status by targeted plasma-based genotyping in metastatic breast cancer. NPJ Breast Cancer 2022, 8, 117. [Google Scholar] [CrossRef]
  45. Irelli, A.; Ranieri, S.C.; Di Giacomo, D.; Malatesta, S.; Patruno, L.V.; Tessitore, A.; Alesse, E.; Cannita, K. Role of the Molecular Tumor Board for the Personalized Treatment of Patients with Metastatic Breast Cancer: A Focus on the State of the Art in Italy. Cancers 2023, 15, 1727. [Google Scholar] [CrossRef]
Figure 1. Workflow diagrams illustrating the aim of this study: (A) routine workflow including IHC +/− ISH +/− NGS; (B) experimental workflow including IHC and NGS.
Figure 1. Workflow diagrams illustrating the aim of this study: (A) routine workflow including IHC +/− ISH +/− NGS; (B) experimental workflow including IHC and NGS.
Biomedicines 11 03164 g001
Figure 2. Distribution of Ki67 expression rate (A) and ERBB2 gene copy number variation (CNV) values (B) in the 31 HER2-negative, HER2-low and HER2-positive samples. (ns: non significant difference).
Figure 2. Distribution of Ki67 expression rate (A) and ERBB2 gene copy number variation (CNV) values (B) in the 31 HER2-negative, HER2-low and HER2-positive samples. (ns: non significant difference).
Biomedicines 11 03164 g002
Table 1. ESCAT ranking of molecular classification.
Table 1. ESCAT ranking of molecular classification.
Tier IClinically
relevant
A: survival benefit in prospective, randomised trials
B: clinical benefit in prospective, single-arm trials
C: clinical benefit in basket trials
Tier IIPotentially
clinically
relevant
A: clinical benefit in retrospective trials
B: increased responsiveness and outcome in prospective trials
Tier IIIPotentially
clinically
relevant
A: clinical benefit in other tumor entities
B: biomarker is located in the same gene/pathway as Tier I-IIIA targets
Tier IVEvidence from
preclinical
studies
A: improved drug sensitivity in in-vitro or in-vivo models
B: actionability shown in in silico models
Tier Vco-targeting approachesImproved objective response in prospective trials
Tier XNo evidenceBiomarkers are not actionable
Table 2. Histological and molecular characteristics of the tumors: Breat cancers specimens were routinely characterized using Immunohistochemistry (IHC) and In situ hybridization (ISH) for hormonal status, defined from estrogen receptor (ER) and Progesterone Receptor (PR) expression, Ki67 labeling index, HER2 expression and HER2 gene amplification. The tumor cell content was determined by expert pathologist to validate the macrodissection area. Next Generation Sequencing (NGS) was further used in this study for determination of gene amplifications defined by Copy number Variation (CNV ) > 3.25, gene mutations analysis with variant allelic fraction (VAF) determination. Only pathogenic (class 5) and likely pathogenic (class 4) mutations were recorded. Microsatellite instability (MSI) status defined as microsatellite instable (MSI) or microsatellite stable (MSS).
Table 2. Histological and molecular characteristics of the tumors: Breat cancers specimens were routinely characterized using Immunohistochemistry (IHC) and In situ hybridization (ISH) for hormonal status, defined from estrogen receptor (ER) and Progesterone Receptor (PR) expression, Ki67 labeling index, HER2 expression and HER2 gene amplification. The tumor cell content was determined by expert pathologist to validate the macrodissection area. Next Generation Sequencing (NGS) was further used in this study for determination of gene amplifications defined by Copy number Variation (CNV ) > 3.25, gene mutations analysis with variant allelic fraction (VAF) determination. Only pathogenic (class 5) and likely pathogenic (class 4) mutations were recorded. Microsatellite instability (MSI) status defined as microsatellite instable (MSI) or microsatellite stable (MSS).
Grade Hormone ReceptorsKi67Tumor Cells ContentHER2/ERBB2Additional Gene AmplificationsAdditional Gene Mutations MSI
SampleSBR-EEERPRHR Status(%)(%)IHC/ISH HER2 StatusCNV ERBB2GeneCNVGenec.P.VAFPathogenicityMSS/MSI
12ER+PR+HR+1270IHC 0HER2−2.4 PIK3CAc.1633G>Ap.(Glu545Lys)34%5MSS
23ER+PR-HR+5070IHC 0HER2−2.3 TP53c.267_269delinsTTp.(Trp91Glyfs*32)39%4MSS
32ER+PR+HR+980IHC 0HER2−2.1 MSS
42ER+PR+HR+1860IHC 0HER2−1.3 PIK3CAc.1624G>Ap.(Glu542Lys)31%5MSI
51ER+PR+HR+680IHC 0HER2−2.4MYOD13.4PIK3CAc.1633G>Ap.(Glu545Lys)36%5MSS
62ER+PR+HR+1870IHC 0HER2−2.1 TP53c.548C>Gp.(Ser183*)6%4MSS
AKT1c.49G>Ap.(Glu17Lys)59%5
71ER+PR+HR+970IHC 0HER2−1.9 PIK3CAc.3140A>Gp.(His1047Arg)26%5MSS
AKT1c.49G>Ap.(Glu17Lys)32%5
82ER+PR+HR+2260IHC 0HER2−1.9 MSS
92ER+PR-HR+220IHC 0HER2−2.0 PIK3CAc.3140A>Gp.(His1047Arg)5%5MSS
101ER+PR+HR+1215IHC 0HER2−2.0 MSS
111ER+PR-HR+1760IHC1+HER2− (HER2 low)1.8FGFR15.8 MSS
122ER+PR+HR+2070IHC1+HER2− (HER2 low)1.9 PIK3CAc.1633G>Ap.(Glu545Lys)27%5MSS
132ER+PR+HR+1270IHC1+HER2− (HER2 low)1.8 MSS
142ER+PR+HR+930IHC1+HER2− (HER2 low)1.8 PIK3CAc.3140A>Gp.(His1047Arg)2%5MSS
151ER+PR+HR+330IHC1+HER2− (HER2 low)1.9 RETc.2372A>Tp.(Tyr791Phe)49%4MSS
162ER+PR+HR+1580IHC2+ ISH-HER2− (HER2 low)1.8 MSS
172ER+PR-HR+870IHC2+ ISH-HER2− (HER2 low)2.4 BRCA2c.9376C>Tp.(Gln3126*)35%5MSS
182ER+PR+HR+1780IHC2+ ISH-HER2− (HER2 low)1.5 MSS
193ER+PR+HR+2575IHC2+ ISH-HER2− (HER2 low)2.5 MSS
202ER+PR+HR+960IHC2+ ISH-HER2− (HER2 low)2.0MYOD14.2BRCA2c.3830delp.(Asn1277Ilefs*7)49%5MSS
213ER+PR+HR+3580IHC2+ ISH+HER2+3.3 PIK3CAc.1624G>Ap.(Glu542Lys)8%5MSS
223ER+PR+HR+5070IHC2+ ISH+HER2+4.2 PIK3CAc.1633G>Ap.(Glu545Lys)44%5MSS
BRCA2c.5702_5703delp.(Glu1901Glyfs*5)40%5
233ER-PR-HR-4030IHC2+ ISH+HER2+3.3 TP53c.614A>Gp.(Tyr205Cys)7%4MSS
PIK3CAc.3140A>Gp.(His1047Arg)9%5
242ER+PR+HR+1040IHC2+ ISH+HER2+2.9 PIK3CAc.3140A>Gp.(His1047Arg)24%5MSS
252ER+PR+HR+1840IHC3+HER2+5.6 MSS
262ER+PR+HR+1870IHC3+HER2+8.2 TP53c.637C>Tp.(Arg213*)30%4MSS
273ER+PR+HR+3570IHC3+HER2+22.9 TP53c.832C>Tp.(Pro278Ser)37%4MSS
282ER+PR+HR+2530IHC3+HER2+7.1PIK3CA ROS15.5 6.2BRCA2c.5073dupp.(Trp1692Metfs*3)13%5MSS
293ER-PR-HR-8060IHC3+HER2+5.3 TP53c.584T>Cp.(Ile195Thr)17%4MSS
303ER+PR-HR+2560IHC3+HER2+4.3FGFR14.4SF3B1c.2098A>Gp.(Lys700Glu)29%4MSS
PIK3CAc.3140A>Gp.(His1047Arg)25%4
313ER+PR-HR+4025IHC3+HER2+19.1FGFR14.4 MSS
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Merlin, J.-L.; Husson, M.; Sahki, N.; Gilson, P.; Massard, V.; Harlé, A.; Leroux, A. Integrated Molecular Characterization of HER2-Low Breast Cancer Using Next Generation Sequencing (NGS). Biomedicines 2023, 11, 3164. https://doi.org/10.3390/biomedicines11123164

AMA Style

Merlin J-L, Husson M, Sahki N, Gilson P, Massard V, Harlé A, Leroux A. Integrated Molecular Characterization of HER2-Low Breast Cancer Using Next Generation Sequencing (NGS). Biomedicines. 2023; 11(12):3164. https://doi.org/10.3390/biomedicines11123164

Chicago/Turabian Style

Merlin, Jean-Louis, Marie Husson, Nassim Sahki, Pauline Gilson, Vincent Massard, Alexandre Harlé, and Agnès Leroux. 2023. "Integrated Molecular Characterization of HER2-Low Breast Cancer Using Next Generation Sequencing (NGS)" Biomedicines 11, no. 12: 3164. https://doi.org/10.3390/biomedicines11123164

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop