Next Article in Journal
Prognostic Impact of Copy Number Alterations’ Profile and AID/RAG Signatures in Acute Lymphoblastic Leukemia (ALL) with BCR::ABL and without Recurrent Genetic Aberrations (NEG ALL) Treated with Intensive Chemotherapy
Previous Article in Journal
C-Reactive Protein as an Early Predictor of Efficacy in Advanced Non-Small-Cell Lung Cancer Patients: A Tumor Dynamics-Biomarker Modeling Framework
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies

by
Carlo Pescia
1,2,
Elena Guerini-Rocco
1,3,
Giuseppe Viale
1 and
Nicola Fusco
1,3,*
1
Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
2
School of Pathology, University of Milan, 20141 Milan, Italy
3
Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(22), 5430; https://doi.org/10.3390/cancers15225430
Submission received: 15 October 2023 / Revised: 5 November 2023 / Accepted: 14 November 2023 / Published: 15 November 2023
(This article belongs to the Special Issue Breast Cancer Biomarkers and Clinical Translation)

Abstract

:

Simple Summary

Risk stratification for early breast cancer (BC) is extremely relevant for tailoring clinical decisions but challenging due to the absence of comprehensive guidelines. Traditional criteria like tumor size, lymph node involvement, histological type, grade, lymphovascular invasion, and immune cell infiltration serve as significant prognostic indicators. Alongside hormone receptor, HER2, and BRCA1/2 testing, molecular subtyping through gene expression profiling offers valuable insights for personalized clinical decisions. “Omics” technologies, applicable to tissue and liquid biopsy samples, have expanded risk evaluation capabilities, with limitations due to the lack of prospective data in early BC. This research overview paper highlights the need for standardized methodologies and integrated pathological models across multiple analytical dimensions for earlu BC risk stratification. The aim is to provide a practical guide for histopathologists and molecular pathologists involved in early BC profiling.

Abstract

Early breast cancer (BC) is the definition applied to breast-confined tumors with or without limited involvement of locoregional lymph nodes. While risk stratification is essential for guiding clinical decisions, it can be a complex endeavor in these patients due to the absence of comprehensive guidelines. Histopathological analysis and biomarker assessment play a pivotal role in defining patient outcomes. Traditional histological criteria such as tumor size, lymph node involvement, histological type and grade, lymphovascular invasion, and immune cell infiltration are significant prognostic indicators. In addition to the hormone receptor, HER2, and—in specific scenarios—BRCA1/2 testing, molecular subtyping through gene expression profiling provides valuable insights to tailor clinical decision-making. The emergence of “omics” technologies, applicable to both tissue and liquid biopsy samples, has broadened our arsenal for evaluating the risk of early BC. However, a pressing need remains for standardized methodologies and integrated pathological models that encompass multiple analytical dimensions. In this study, we provide a detailed examination of the existing strategies for early BC risk stratification, intending to serve as a practical guide for histopathologists and molecular pathologists.

1. Introduction

Early breast cancer (BC) is defined by the presence of tumors confined to the breast, including tumors with an involvement of less than three locoregional lymph nodes [1]. This definition aligns with the TNM stages T1-3, N0-1, and M0, as established by the American Joint Committee on Cancer (AJCC) [2,3]. Despite the rarity of recurrences, effectively managing early BC poses a significant challenge: precise risk stratification [4]. Despite efforts in clinical studies to identify high-risk early BC, there is a lack of consistency in defining such cases. In the monarchE trial, high-risk early BC was defined as the presence of either four or more positive axillary lymph nodes or one to three positive axillary lymph nodes with at least one of the following features: grade 3 disease, a tumor size of 5 cm or larger, and a Ki-67 proliferative index of at least 20% [5]. A consensus paper by the IRIDE working group [6] attempted a definition of high-risk early BC to be applied to hormone-responsive cases, encompassing grade 3 histology, pT3-pT4 and/or pN2-pN3 staging, Ki67 > 30%, expression of estrogen receptors < 10% and/or progesterone receptors < 20%, high residual cancer burden after neoadjuvant therapy, and high-risk class based on gene profiling assays. However, while each of these factors has the potential to influence the overall risk of relapse, there is currently no available prediction tool that incorporates and assigns appropriate weights to each of them.
Accurately evaluating the risk of both early and late recurrence is extremely important for informing clinical management strategies [7,8]. In this respect, histopathological analysis, combined with biomarker testing, plays a pivotal role in improving patient outcomes [9,10,11,12,13,14,15]. Traditional histology criteria, including tumor size, lymph node involvement, histological grade, lymphovascular invasion, and immune cell infiltration, have long been acknowledged as significant prognostic indicators in early BC [14,16]. The evaluation of hormone receptor (HR) status, specifically estrogen receptor (ER) and progesterone receptor (PgR), along with HER2 status, constitutes another essential aspect of BC profiling [17,18,19]. Molecular subtyping via gene expression profiling offers supplementary insights beyond conventional histopathological criteria, aiding treatment decisions [20,21,22,23]. Another significant biomarker in a subset of early BC is represented by BRCA1/2 mutational status, whose testing holds importance for risk stratification, patients’ selection for treatment with PARP inhibitors, and genetic screening in high-risk individuals [8].
Advancements in omics technologies, applicable to both tissue and liquid biopsy samples, have significantly broadened our tools for accurately assessing the risk associated with early BC [24]. Despite these advancements, several challenges persist for prognostication and therapy prediction in these patients. It is crucial to establish standardized methodologies and incorporate a multitude of factors into comprehensive pathological models. In this study, we provide an in-depth examination of the currently available biomarkers for early BC risk stratification. Each biomarker is thoroughly discussed, along with tailored testing strategies, aiming to offer a practical guide for histopathologists and molecular pathologists.

2. Dissecting the Pathology Report

2.1. Histopathology

Morphological analysis stands as the bedrock of BC clinical decision-making [25]. Consequently, histopathological parameters continue to provide invaluable insights into tumor characteristics and behavior, aiding in treatment determinations and risk stratification, albeit primarily offering prognostic insights. The conventional histopathological factors for early BC encompass tumor type, size, histological grade, lymph node involvement, and lymphovascular invasion. All these histopathological factors can be integrated into prognostic scoring systems, such as the Nottingham prognostic index (NPI), which integrates tumor size, grading, and lymph node status, as well as other online resources (e.g., predictPLUS and Adjuvant! Online) [26,27,28,29,30]. While traditional histopathological factors provide valuable prognostic information, they should be discussed in the context of a more comprehensive clinical and biological profiling.

2.1.1. Tumor Type

Histologic type describes the specific morphological and architectural characteristics that define the microscopic appearance of BC [31]. Low-risk BCs encompass a range of tumor types, including low-grade non-special-type (NST) carcinomas, and special-type tumors such as tubular carcinoma, cribriform carcinoma, mucinous carcinoma, salivary-gland-type carcinomas, and tumors with apocrine differentiation [31,32,33]. These malignancies are characterized by a low frequency of genomic instability and often exhibit an indolent clinical course with a low rate of relapses [34]. Nonetheless, in cases like these, therapeutic approaches can be complex, especially when dealing with NST or special-type triple-negative breast cancer (TNBC).

2.1.2. Tumor Size

The primary tumor’s largest dimension profoundly influences the prognosis, as larger tumor sizes are associated with a higher risk of metastasis and poorer outcomes [32]. The AJCC staging system incorporates tumor size (represented by the T specifier) to define BC stages. Considerations for determining the size of invasive carcinomas should be based on an assessment that combines macroscopic observations with microscopic examination and should be compared to clinical staging information [35,36,37]. According to the CAP cancer reporting protocol of invasive breast cancer (v4.9.0.0, available at https://www.cap.org/protocols-and-guidelines/cancer-reporting-tools/cancer-protocol-templates, accessed on 13 November 2023), for carcinomas larger than 1.0 mm but less than 1.5 mm, it is crucial not to round down to 1.0 mm; instead, round up to 2.0 mm. This precaution prevents the misclassification of tumors as pT1mi, which display survival rates very similar to those of ductal carcinoma in situ (DCIS)

2.1.3. Histological Grade

All invasive breast carcinomas should undergo grading, specifically using the Nottingham combined histologic grade, which is an Elston–Ellis modification of the Scarff–Bloom–Richardson grading system [38,39]. High-grade tumors are associated with increased aggressiveness and poorer outcomes, while low-grade tumors tend to have a more favorable prognosis. The Nottingham combined histologic grade assesses three main factors: the extent of tubule formation, the degree of nuclear pleomorphism, and the mitotic count (or mitotic rate). This grading system should be employed for reporting purposes, as it establishes a correlation between the histologic grade and clinical outcomes within each stage grouping.

2.1.4. Lymph Node Status

Lymph node involvement is another well-established prognostic factor in BC, determining, along with tumor size, the clinical and pathological stage of the disease (T and N specifiers) [40,41]. The presence of tumor cells within regional lymph nodes indicates a higher likelihood of systemic spread and is associated with a worse prognosis. The number of involved lymph nodes further refines the prognostic information, as well as the presence of extranodal extensions, which has been linked to inferior overall survival and disease-recurrence-free survival and should be incorporated into the pathology report [42]. The number of metastatic lymph nodes that define high-risk individuals in early BC can vary depending on the clinical guidelines and individual patient factors. However, commonly, the presence of four or more metastatic lymph nodes (pN2-N3) is considered a threshold for classifying a patient as high-risk in early BC [6], although such definition may be subject to changes and variations in both trials and clinical practice.

2.1.5. Lymphovascular Invasion

Lymphovascular invasion (LVI) is defined by the presence of cancer cells within an endothelial-lined space outside the border of the invasive carcinoma, regardless of the vessel type (i.e., blood or lymphatics) [43]. The presence of LVI correlates with an increased risk of both lymph node involvement and metastatic dissemination, ultimately being associated with adverse outcomes, both in adjuvant and neoadjuvant settings [43,44,45,46,47,48,49,50,51], and should always be indicated in the pathology report.

2.2. Biomarkers

2.2.1. Hormone Receptors

Assessing estrogen receptor (ER) and progesterone receptor (PgR) status using immunohistochemistry (IHC) is pivotal in BC management [52,53,54]. ER and PgR are key in hormonal signaling pathways, significantly influencing prognoses and treatment responses. ER-positive BCs (with >10% nuclear-positive neoplastic cells) constitute approximately 70% of cases, tied to milder disease progression and higher hormone sensitivity [55]. ER positivity implies tumor reliance on estrogen for growth and survival. Patients with ER-positive tumors have shown significant benefits from endocrine therapies [56]. PgR status is evaluated alongside ER status, as the expression of PgR is regulated via estrogen signaling [8,57]. Positive PgR status, defined as ≥1% neoplastic cells exhibiting nuclear staining, further refines the prognostic information and may have implications for endocrine therapy selection [58]. Tumors positive for both ER and PgR fall into the luminal group and generally have a better prognosis than dual-negative receptor tumors [59]. Importantly, the category of ER-low BC, introduced by 2020 ASCO/CAP guidelines and characterized by low ER expression (positivity in 1–10% of tumor cells), poses management and treatment challenges [60,61,62,63]. These tumors often exhibit aggressive characteristics, such as higher grade, larger size, and increased proliferation, and are associated with younger age, lymph node involvement, and poorer prognoses [62,64].

2.2.2. Ki-67 Labeling Index

The Ki-67 index is a measure of cellular proliferation and is essential in the assessment of early BC risk [65,66]. High Ki-67 levels are associated with a more aggressive tumor phenotype and poorer prognosis [67]. Patients with a high Ki-67 index may have a higher risk of disease recurrence and benefit from more intensive treatment approaches, such as adjuvant chemotherapy [68]. Hence, the Ki-67 index helps in distinguishing between patients who may benefit from additional adjuvant therapy and those who could be spared from unnecessary interventions, although an internationally established final consensus on the Ki67 cut-off for therapeutic decisions has not been reached yet [69]. Nevertheless, a Ki67 20% cut-off is currently used to identify luminal B BC among HR+/HER2- cases, with obvious therapeutic implications [70,71,72]. According to the American Society of Clinical Oncology (ASCO) guidelines, in postmenopausal patients diagnosed with stage I-II BC, Ki67 expression might be used in combination with other clinical pathological factors to arrive at informed decisions regarding adjuvant endocrine and chemotherapy treatments when multigene assays are not accessible. Ki67 expression levels become particularly valuable for prognostic assessment when they fall below 5% (indicating low proliferation) or exceed 30% (indicating high proliferation), while distinguishing values within this range is essentially technically unreliable [73]. Interestingly, the American Food and Drug Administration (FDA) has recently approved abemaciclib, a CDK4/6 inhibitor, in combination with endocrine therapy in the adjuvant setting for HR+/HER2- and node-positive BC at high risk for recurrence with a Ki67 proliferative index of ≥20% (immunohistochemistry validated on Agilent platforms with the MIB1 clone). This approval stems from the results of the monarchE trial (NCT03155997), which, however, revealed a clinical benefit from the addition of abemaciclib to this subset of patients regardless of the proliferative status [5]. Consequently, Ki67 mainly serves as a prognostic factor for high-risk scenarios, rather than a response predictor [74]. It is important to acknowledge that using Ki67 as a predictive and prognostic marker is not devoid of debate [11]. Disputes involve antibody clone selection, scoring methods, reproducibility across labs, and the potential benefits of computer-assisted analysis or AI for Ki67 assessment. Efforts persist in establishing uniform guidelines and cut-offs for Ki67 assessment to enhance its consistency and reliability in clinical practice [70,75,76].

2.2.3. HER2

Approximately 15–20% of BCs exhibit HER2 overexpression or amplification [77]. HER2-positive tumors tend to have a more aggressive clinical course, characterized by a higher risk of recurrence, and overall poorer prognosis [78]. However, targeted therapies like trastuzumab and pertuzumab have notably improved HER2-positive BC outcomes by inhibiting HER2 signaling pathways [79,80]. HER2 status is assessed using IHC for protein expression and reflex in situ hybridization (ISH) techniques for gene amplification. Standardized testing is used clinically to determine the eligibility for targeted treatments and guide decisions [81]. The integration of hormone receptor and HER2 testing into treatment algorithms has transformed the management of BC [32,82]. Additionally, HER2-low status, defined as HER2 FISH-negative cases with HER2 IHC 1+ or 2+ scores, has gained much interest lately due to the availability of new antibody–drug conjugates, such as trastuzumab–deruxtecan (T-Dx), which have been approved for previously treated metastatic BC [83,84]. Precise HER2 expression levels are vital, especially as the minimal level for T-DXd effectiveness is being studied. Given T-DXd’s potential benefits in HER2-zero (IHC 0) cases [85], the definition of HER2-low might change. Though HER2-low status currently does not impact early BC treatment, accurately defining HER2 status and reassessing it during progression and metastasis is essential for therapeutic strategies.

2.3. Tumor-Infiltrating Lymphocytes (TILs)

Tumor-infiltrating lymphocytes (TILs) have emerged as a critical component in the context of TNBC and HER2+ BC, where they hold a prognostic value [86]. According to the International TILs Working Group guidelines [87], TIL evaluation should encompass all mononuclear cells within the stromal/peritumoral compartment, expressed as a percentage relative to the stromal area itself. This evaluation should exclude intratumoral TILs, TILs outside the tumor border, TILs associated with intraductal carcinomas, as well as areas of necrosis or artifacts.
The abundance of so-called “stromal” TILs has been correlated to HER2+ and basal-like BC evaluated with gene expression profiling [88]. Moreover, higher TILs, with a threshold set at 20%, significantly predict pathological complete response (pCR) to neoadjuvant chemotherapy and better survival in HER2+ and TNBC [50,51,89,90,91,92]. Additionally, increased TILs have shown a correlation with improved outcomes following adjuvant therapy [93]. In detail, findings from the BIG-2-98 trial demonstrated that each 10% increase in TILs correlated with a reduced risk of relapse and death in node-positive TNBC, and conferred significant benefit in terms of disease-free survival in node-positive HER2+ patients treated with anthracycline-only chemotherapy [94]. Analyses from the FinHER trial revealed that TNBC with higher TILs at diagnosis experienced decreased distant recurrence rates, while HER2+ BC with higher TILs derived increased trastuzumab benefit [95]. Intriguingly, a 30% cut-off for TILs has been found to strongly up- and downstage traditional histopathological staging in BC, along with rendering histological grading irrelevant in terms of its prognostic implication in a pooled analysis comprising TILs; in light of these results, some authors have advocated for the integration of TILs in BC staging [96]. In the setting of early TNBC treated with adjuvant anthracycline-based chemotherapy with or without taxanes, a pooled data analysis [93] indicated a median TIL value of 23% at diagnosis; lower TIL levels were significantly associated with older age, larger tumor size, nodal involvement, and lower histological grade [97]. Notably, the quantity of TILs was significantly linked with improved survival outcomes in terms of OS, invasive-disease-free survival (i-DFS), and distant-disease-free survival (d-DFS), although TILs did not define the prognostic impact of different chemotherapy regimens. Moreover, node-negative TNBC with at least 30% TILs exhibited excellent survival outcomes, thus identifying 30% as an important exploratory cut-off in TIL evaluation and supporting its clinical validity in prognostication models [93]. A TIL cut-off of ≥30% has also demonstrated predictive value for excellent OS in stage I TNBC not treated with (neo)adjuvant chemotherapy [98]. The relationship between TILs and the response to PDL1-inhibitor-based immunotherapy has been variably explored as well [99,100], also in the neoadjuvant setting for TNBC [101,102,103,104,105]. Recognizing the significance of TIL scoring as a valid prognostic factor, the 2019 St Gallen Consensus [106] recommended its routine characterization and reporting, with 66% of the panelists endorsing this practice. However, 90% of the panelists were hesitant to base their treatment strategies, such as chemotherapy de-escalation, solely on TIL reports, as the clinical utility of TILs remains under scrutiny and not yet fully substantiated. Therefore, routine TIL reporting by pathologists may be suggested but not mandatory, as reiterated in the 2023 St Gallen Consensus [107]. Future developments, especially the integration of machine learning for enhanced TIL assessment, may potentially alter the current perspective [108,109].

3. Molecular Subtyping and Gene Expression Profiling

3.1. Gene Expression Profiling Assays to Inform Treatment

Gene expression profiling assays have revolutionized BC management by providing valuable prognostic and predictive information beyond traditional clinicopathological factors; they comprise the Oncotype Dx, MammaPrint, Prosigna, Endopredict, and BC index assays [48,110,111]. They enable personalized strategies, sparing patients from unnecessary chemotherapy treatments, as they aid in differentiating which patients could potentially gain advantages or disadvantages from the inclusion of chemotherapy alongside endocrine therapy, mostly in HR+/HER2- BCs [69]. Among different gene expression profiling assays, the Prosigna (PAM50) assay and 3.1.2 EndoPredict® tests have, at present, been validated only at a prognostic level, while Oncotype Dx, MammaPrint, and BCI hold both a prognostic and predictive role. The ASCO guidelines (Table 1) [73] suggest the use of Oncotype Dx, MammaPrint, or BCI in postmenopausal or >50-year-old women with HR+/HER2- early BC, which is either node-negative or with 1–3 node metastases. In premenopausal patients, Oncotype Dx is recommended in node-negative HR+/HER2- patients, while in the case of 1–3 axillary lymph node metastases, chemotherapy is advised, regardless of the genomic assay results. Currently, information regarding genomic test applications in patients with ≥4 positive nodes is lacking [73].
Similarly, the European Society for Medical Oncology (ESMO) assigns level I of evidence (i.e., evidence derived from at least one large randomized controlled trial of good methodological quality or meta-analyses of well-conducted randomized trials without heterogeneity) and grade A of recommendation (i.e., strong evidence for efficacy with a substantial clinical benefit) for both Oncotype DX and MammaPrint, as shown in Table 2.
Of note, the ESMO and ASCO guidelines do not favor one test over another, but emphasize their value in complex scenarios where selecting optimal adjuvant therapy is uncertain, as in luminal B BCs with 1–3 positive axillary nodes [3].
Beyond the conventional luminal and non-luminal molecular subtypes, additional molecular subgroups have emerged. For instance, the claudin-low subtype is associated with stem-like features and immune system modulation, and it is defined by a poorer prognosis, a triple-negative phenotype, low genomic instability, mutational burden and proliferation levels, and high levels of immune and stromal cell infiltration [112,113,114]. Another example is the molecular apocrine subtype, which is characterized by androgen receptor expression, ER negativity, and frequent HER2 positivity, paired with aggressive clinical behavior [115]. These subtypes provide further insight into BC’s heterogeneity and could potentially shape future tailored therapeutic approaches.

3.2. BRCA1/2 Mutations

BRCA1/2 mutations play a significant role in early BC development and management [116]. These mutations are autosomal-dominant inherited genetic alterations impairing the homologous recombination machinery that increases the risk of developing mainly breast and ovarian cancers. Detecting BRCA1/2 mutations in early BC patients is essential for treatment choices and risk evaluation [3]. Patients with BRCA1/2 mutations may have distinct tumor characteristics and treatment responses, as they are more likely to develop TNBCs, which are typically more aggressive and may require tailored approaches. BRCA1/2 mutations also impact therapy sensitivity; for instance, these mutations heighten the effectiveness of platinum-based chemotherapy and PARP inhibitors, targeting DNA repair deficits with synthetic lethality effects [117]. Specifically, the OlympiA trial (NCT02032823) has demonstrated that in patients with HER2-negative early BC at high risk and harboring germline BRCA1 or BRCA2 mutations, the use of the adjuvant PARP inhibitor olaparib following local treatment and neoadjuvant or adjuvant chemotherapy resulted in significantly longer overall survival and survival without invasive or metastatic spread compared with the administration of a placebo [118,119]. Olaparib [120] and other PARP inhibitors, such as niraparib [121] or talazoparib [122], have demonstrated promising results also in the neoadjuvant setting for HER2-negative BRCA-mutated early BC, although no significant difference has been observed in terms of pCR between using PARP inhibitors alone or in combination with chemotherapy [123]. Therefore, identifying BRCA1/2 mutations in early BC patients could guide treatment choices, potentially leading to improved outcomes [68]. Moreover, BRCA1/2 mutations stretch beyond initial diagnosis and treatment, affecting ongoing management and risk assessment for both patients and their families. Carriers face higher risks of contralateral BC and ovarian cancer. To mitigate these risks, proactive measures like risk-reducing surgeries (e.g., bilateral mastectomy and bilateral salpingo-oophorectomy) and intensified monitoring are recommended. The identification of BRCA mutations in BC also triggers the consideration of genetic testing and surveillance for family members at risk [124,125]. BRCA1/2 testing should be offered to high-risk groups, especially patients with strong familiarity for BRCA-related tumors, an early diagnosis (before the age of 50), a diagnosis of TNBC before the age of 60, male patients with BC, and/or patients with a personal history of ovarian cancer or second BC [3]. In essence, BRCA1/2 mutations significantly impact early BC by shaping tumor traits, treatment responses, and long-term risk management. Detecting these mutations in patients informs treatments, facilitates evaluation of future risks, and guides interventions for patients and their high-risk family members.

4. Conclusions: Challenges and Future Directions

Significant progress has been made in identifying prognostic and predictive factors for the treatment of early BC. Pathologists have played a fundamental role in BC biomarker definition and should be familiar with all the above-mentioned topics. However, challenges in biomarker testing and prognostic/predictive assays remain, and there are still opportunities for further research (Figure 1). One of the critical challenges is establishing standardized methods of assessing and interpreting biomarkers, including hormone receptors, HER2, Ki-67, TILs, as well as gene expression profiling assays. Consistency in methodologies and cut-off values is essential for reliable outcomes across various laboratories and clinical settings [68]. There are many unmet needs in the clinical management of early BC, ranging from the definition of high-risk disease to the optimal therapeutic choice in premenopausal node-positive patients, to the clinical utility of gene profiling assays in the choice of neoadjuvant treatment, to the need for the implementation of real-world data for proper biomarker validation. Gene expression profiling assays are recognized for their prognostic significance, yet their primary value lies in their potential or established predictive role in supporting chemotherapy de-escalation. Their use in early breast cancer risk assessment is currently not recommended. Further research is necessary to uncover novel biomarkers and molecular pathways that could enhance prognostic precision and explore new applications of existing tools. Many intriguing studies have explored the proteomic [126,127,128], metabolomic [129,130], and lipidomic [131,132] profiles of BC, with interesting insights that could be appliable to the early-stage disease. The integration of multi-omics data and machine-learning approaches could also provide insights into the intricate biology of BC and provide fruitful information in terms of risk assessment [133,134,135,136,137]. Collectively, these omics approaches contribute to broadening our comprehension of breast cancer, spanning from genetic susceptibility to molecular mechanisms. Nevertheless, their use in the present clinical practice is still a matter of controversy. Future clinical trials that embrace a comprehensive approach, integrating clinical, pathological, and molecular aspects, may significantly advance the risk assessment of early BC.

Author Contributions

Conceptualization, G.V. and N.F.; methodology, E.G.-R. and N.F.; software, C.P. and N.F.; writing—original draft preparation, C.P. and N.F.; writing—review and editing, E.G.-R. and G.V.; supervision, G.V. and N.F.; project administration, N.F. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by the Italian Ministry of Health with “RicercaCorrente”, and “5  ×  1000” funds. The APC was funded by the University of Milan through the APC initiative.

Informed Consent Statement

Not applicable.

Conflicts of Interest

G.V. has received consultation fees from Dako/Agilent, Roche, MSD Oncology, AstraZeneca, Daiichi Sankyo, Pfizer, and Eli Lilly. N.F. has received honoraria for consulting, advisory roles, speaker bureau, travel, and/or research grants from Merck Sharp & Dohme (MSD), Merck, Novartis, AstraZeneca, Roche, Menarini, Daiichi Sankyo, GlaxoSmithKline (GSK), Gilead, Adicet Bio, Sermonix, Reply, Veracyte Inc., Leica Biosystems, and Lilly. These companies had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; and/or in the decision to publish the results.

References

  1. Thomssen, C.; Balic, M.; Harbeck, N.; St. Gnant, M. Gallen/Vienna 2021: A Brief Summary of the Consensus Discussion on Customizing Therapies for Women with Early Breast Cancer. Breast Care 2021, 16, 135–143. [Google Scholar] [CrossRef]
  2. Schlam, I.; Tarantino, P.; Morganti, S.; Lynce, F.; Trapani, D.; Mayer, E.L.; Garrido-Castro, A.C.; Waks, A.; Tolaney, S.M. Emerging Targeted Therapies for Early Breast Cancer. Drugs 2022, 82, 1437–1451. [Google Scholar] [CrossRef]
  3. Cardoso, F.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rubio, I.T.; Zackrisson, S.; Senkus, E. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 1194–1220. [Google Scholar] [CrossRef]
  4. Thomas, A.; Parsons, H.A.; Smith, K.L. Late Recurrence Following Early Breast Cancer. J. Clin. Oncol. 2022, 40, 1400–1406. [Google Scholar] [CrossRef]
  5. Harbeck, N.; Rastogi, P.; Martin, M.; Tolaney, S.M.; Shao, Z.M.; Fasching, P.A.; Huang, C.S.; Jaliffe, G.G.; Tryakin, A.; Goetz, M.P.; et al. Adjuvant abemaciclib combined with endocrine therapy for high-risk early breast cancer: Updated efficacy and Ki-67 analysis from the monarchE study. Ann. Oncol. 2021, 32, 1571–1581. [Google Scholar] [CrossRef]
  6. Garutti, M.; Griguolo, G.; Botticelli, A.; Buzzatti, G.; De Angelis, C.; Gerratana, L.; Molinelli, C.; Adamo, V.; Bianchini, G.; Biganzoli, L.; et al. Definition of High-Risk Early Hormone-Positive HER2-Negative Breast Cancer: A Consensus Review. Cancers 2022, 14, 1898. [Google Scholar] [CrossRef]
  7. Michaels, E.; Worthington, R.O.; Rusiecki, J. Breast Cancer: Risk Assessment, Screening, and Primary Prevention. Med. Clin. N. Am. 2023, 107, 271–284. [Google Scholar] [CrossRef]
  8. Brett, J.O.; Mayer, E.L. New Developments in Systemic Management for High-Risk Early-Stage Hormone-Receptor-Positive, HER2-Negative Breast Cancer. Curr. Treat. Options Oncol. 2023, 24, 594–610. [Google Scholar] [CrossRef]
  9. Wen, H.Y.; Collins, L.C. Breast Cancer Pathology in the Era of Genomics. Hematol. Oncol. Clin. N. Am. 2023, 37, 33–50. [Google Scholar] [CrossRef]
  10. Pisapia, P.; L’Imperio, V.; Galuppini, F.; Sajjadi, E.; Russo, A.; Cerbelli, B.; Fraggetta, F.; d’Amati, G.; Troncone, G.; Fassan, M.; et al. The evolving landscape of anatomic pathology. Crit. Rev. Oncol. Hematol. 2022, 178, 103776. [Google Scholar] [CrossRef]
  11. Fusco, N.; Malapelle, U.; Criscitiello, C. Editorial: Diagnosis and Treatment of Breast Cancer in 2022: The Rise of Novel Molecular Biomarkers. Front. Mol. Biosci. 2022, 9, 1117323. [Google Scholar] [CrossRef]
  12. Hayes, D.F. Precision Medicine and Testing for Tumor Biomarkers—Are All Tests Born Equal? JAMA Oncol. 2018, 4, 773–774. [Google Scholar] [CrossRef]
  13. Angerilli, V.; Galuppini, F.; Pagni, F.; Fusco, N.; Malapelle, U.; Fassan, M. The Role of the Pathologist in the Next-Generation Era of Tumor Molecular Characterization. Diagnostics 2021, 11, 339. [Google Scholar] [CrossRef]
  14. Beňačka, R.; Szabóová, D.; Guľašová, Z.; Hertelyová, Z.; Radoňák, J. Classic and New Markers in Diagnostics and Classification of Breast Cancer. Cancers 2022, 14, 5444. [Google Scholar] [CrossRef]
  15. Curigliano, G.; Dent, R.; Llombart-Cussac, A.; Pegram, M.; Pusztai, L.; Turner, N.; Viale, G. Incorporating clinicopathological and molecular risk prediction tools to improve outcomes in early HR+/HER2- breast cancer. NPJ Breast Cancer 2023, 9, 56. [Google Scholar] [CrossRef]
  16. Liu, R.; Xiao, Z.; Hu, D.; Luo, H.; Yin, G.; Feng, Y.; Min, Y. Cancer-Specific Survival Outcome in Early-Stage Young Breast Cancer: Evidence From the SEER Database Analysis. Front. Endocrinol. 2021, 12, 811878. [Google Scholar] [CrossRef]
  17. Huang, H.; Zhou, J.; Chen, H.; Li, J.; Zhang, C.; Jiang, X.; Ni, C. The immunomodulatory effects of endocrine therapy in breast cancer. J. Exp. Clin. Cancer Res. 2021, 40, 19. [Google Scholar] [CrossRef]
  18. Tarantino, P.; Curigliano, G.; Parsons, H.A.; Lin, N.U.; Krop, I.; Mittendorf, E.A.; Waks, A.; Winer, E.P.; Tolaney, S.M. Aiming at a Tailored Cure for ERBB2-Positive Metastatic Breast Cancer: A Review. JAMA Oncol. 2022, 8, 629–635. [Google Scholar] [CrossRef]
  19. 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. [Google Scholar] [CrossRef]
  20. Russnes, H.G.; Lingjaerde, O.C.; Borresen-Dale, A.L.; Caldas, C. Breast Cancer Molecular Stratification: From Intrinsic Subtypes to Integrative Clusters. Am. J. Pathol. 2017, 187, 2152–2162. [Google Scholar] [CrossRef]
  21. Eliyatkın, N.; Yalçın, E.; Zengel, B.; Aktaş, S.; Vardar, E. Molecular Classification of Breast Carcinoma: From Traditional, Old-Fashioned Way to A New Age, and A New Way. J. Breast Health 2015, 11, 59–66. [Google Scholar] [CrossRef] [PubMed]
  22. Grizzi, G.; Ghidini, M.; Botticelli, A.; Tomasello, G.; Ghidini, A.; Grossi, F.; Fusco, N.; Cabiddu, M.; Savio, T.; Petrelli, F. Strategies for Increasing the Effectiveness of Aromatase Inhibitors in Locally Advanced Breast Cancer: An Evidence-Based Review on Current Options. Cancer Manag. Res. 2020, 12, 675–686. [Google Scholar] [CrossRef] [PubMed]
  23. McVeigh, T.P.; Kerin, M.J. Clinical use of the Oncotype DX genomic test to guide treatment decisions for patients with invasive breast cancer. Breast Cancer 2017, 9, 393–400. [Google Scholar] [CrossRef] [PubMed]
  24. Venetis, K.; Cursano, G.; Pescia, C.; D’Ercole, M.; Porta, F.M.; Blanco, M.C.; Frascarelli, C.; Ivanova, M.; Guerini Rocco, E.; Fusco, N. Liquid biopsy: Cell-free DNA based analysis in breast cancer. J. Liq. Biopsy 2023, 1, 100002. [Google Scholar] [CrossRef]
  25. Ben-Dror, J.; Shalamov, M.; Sonnenblick, A. The History of Early Breast Cancer Treatment. Genes 2022, 13, 960. [Google Scholar] [CrossRef]
  26. Kerin, E.P.; Davey, M.G.; McLaughlin, R.P.; Sweeney, K.J.; Barry, M.K.; Malone, C.M.; Elwahab, S.A.; Lowery, A.J.; Kerin, M.J. Comparison of the Nottingham Prognostic Index and OncotypeDX© recurrence score in predicting outcome in estrogen receptor positive breast cancer. Breast 2022, 66, 227–235. [Google Scholar] [CrossRef]
  27. Hajage, D.; de Rycke, Y.; Bollet, M.; Savignoni, A.; Caly, M.; Pierga, J.Y.; Horlings, H.M.; Van de Vijver, M.J.; Vincent-Salomon, A.; Sigal-Zafrani, B.; et al. External validation of Adjuvant! Online breast cancer prognosis tool. Prioritising recommendations for improvement. PLoS ONE 2011, 6, e27446. [Google Scholar] [CrossRef]
  28. Jacobson, A.; Macfarlane, C.L.; Pozniak, E. Feasibility of Integrating a Mobile Decision-Support App into a Multicomponent CME Initiative: Developing Clinician Competence at the Point of Care. J. Eur. CME 2020, 9, 1834762. [Google Scholar] [CrossRef]
  29. Lee, A.H.; Ellis, I.O. The Nottingham prognostic index for invasive carcinoma of the breast. Pathol. Oncol. Res. 2008, 14, 113–115. [Google Scholar] [CrossRef]
  30. Lambertini, M.; Pinto, A.C.; Ameye, L.; Jongen, L.; Del Mastro, L.; Puglisi, F.; Poggio, F.; Bonotto, M.; Floris, G.; Van Asten, K.; et al. The prognostic performance of Adjuvant! Online and Nottingham Prognostic Index in young breast cancer patients. Br. J. Cancer 2016, 115, 1471–1478. [Google Scholar] [CrossRef]
  31. International Agency for Research on Cancer. Breast Tumours; International Agency for Research on Cancer: Lyon, France, 2019.
  32. Fusco, N.; Sajjadi, E.; Venetis, K.; Ivanova, M.; Andaloro, S.; Guerini-Rocco, E.; Montagna, E.; Caldarella, P.; Veronesi, P.; Colleoni, M.; et al. Low-risk triple-negative breast cancers: Clinico-pathological and molecular features. Crit. Rev. Oncol. Hematol. 2022, 172, 103643. [Google Scholar] [CrossRef] [PubMed]
  33. Cserni, G.; Quinn, C.M.; Foschini, M.P.; Bianchi, S.; Callagy, G.; Chmielik, E.; Decker, T.; Fend, F.; Kovács, A.; van Diest, P.J.; et al. Triple-Negative Breast Cancer Histological Subtypes with a Favourable Prognosis. Cancers 2021, 13, 5694. [Google Scholar] [CrossRef] [PubMed]
  34. Cao, L.; Niu, Y. Triple negative breast cancer: Special histological types and emerging therapeutic methods. Cancer Biol. Med. 2020, 17, 293–306. [Google Scholar] [CrossRef]
  35. Hamza, A.; Sakhi, R.; Alrajjal, A.; Ibrar, W.; Miller, S.; Salehi, S.; Edens, J.; Ockner, D. Tumor Size in Breast Carcinoma: Gross Measurement Is Important! Int. J. Surg. Pathol. 2018, 26, 494–499. [Google Scholar] [CrossRef] [PubMed]
  36. Pritt, B.; Ashikaga, T.; Oppenheimer, R.G.; Weaver, D.L. Influence of breast cancer histology on the relationship between ultrasound and pathology tumor size measurements. Mod. Pathol. 2004, 17, 905–910. [Google Scholar] [CrossRef]
  37. Choi, B.; Jegatheeswaran, L.; Nakhoul, M.; Haria, P.; Srivastava, R.; Karki, S.; Lupi, M.; Patel, V.; Chakravorty, A.; Babu, E. Axillary staging in ductal carcinoma in situ with microinvasion: A meta-analysis. Surg. Oncol. 2021, 37, 101557. [Google Scholar] [CrossRef]
  38. Rakha, E.A.; El-Sayed, M.E.; Lee, A.H.; Elston, C.W.; Grainge, M.J.; Hodi, Z.; Blamey, R.W.; Ellis, I.O. Prognostic significance of Nottingham histologic grade in invasive breast carcinoma. J. Clin. Oncol. 2008, 26, 3153–3158. [Google Scholar] [CrossRef]
  39. Elston, C.W.; Ellis, I.O. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: Experience from a large study with long-term follow-up. Histopathology 1991, 19, 403–410. [Google Scholar] [CrossRef]
  40. Eijkelboom, A.H.; de Munck, L.; de Vries, M.; Francken, A.B.; Hendriks, M.P.; Strobbe, L.; Witteveen, A.; van Maaren, M.C.; Siesling, S. Routine and interval detection of locoregional breast cancer recurrences and risk of subsequent distant metastasis. Breast Cancer Res. Treat. 2022, 197, 123–135. [Google Scholar] [CrossRef]
  41. Caldonazzi, N.; Rizzo, P.C.; Eccher, A.; Girolami, I.; Fanelli, G.N.; Naccarato, A.G.; Bonizzi, G.; Fusco, N.; d’Amati, G.; Scarpa, A.; et al. Value of Artificial Intelligence in Evaluating Lymph Node Metastases. Cancers 2023, 15, 2491. [Google Scholar] [CrossRef]
  42. Ma, X.; Yang, X.; Yang, W.; Shui, R. Prognostic value of extranodal extension in axillary lymph node-positive breast cancer. Sci. Rep. 2021, 11, 9534. [Google Scholar] [CrossRef] [PubMed]
  43. Invernizzi, M.; Corti, C.; Lopez, G.; Michelotti, A.; Despini, L.; Gambini, D.; Lorenzini, D.; Guerini-Rocco, E.; Maggi, S.; Noale, M.; et al. Lymphovascular invasion and extranodal tumour extension are risk indicators of breast cancer related lymphoedema: An observational retrospective study with long-term follow-up. BMC Cancer 2018, 18, 935. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, S.; Zhang, D.; Yi, S.; Gong, M.; Lu, C.; Cai, Y.; Tang, X.; Zou, L. The relationship of lymphatic vessel density, lymphovascular invasion, and lymph node metastasis in breast cancer: A systematic review and meta-analysis. Oncotarget 2017, 8, 2863–2873. [Google Scholar] [CrossRef] [PubMed]
  45. Davis, B.W.; Gelber, R.; Goldhirsch, A.; Hartmann, W.H.; Hollaway, L.; Russell, I.; Rudenstam, C.M. Prognostic significance of peritumoral vessel invasion in clinical trials of adjuvant therapy for breast cancer with axillary lymph node metastasis. Hum. Pathol. 1985, 16, 1212–1218. [Google Scholar] [CrossRef] [PubMed]
  46. Torous, V.F.; Simpson, R.W.; Balani, J.P.; Baras, A.S.; Berman, M.A.; Birdsong, G.G.; Giannico, G.A.; Paner, G.P.; Pettus, J.R.; Sessions, Z. College of American pathologists cancer protocols: From optimizing cancer patient care to facilitating interoperable reporting and downstream data use. JCO Clin. Cancer Inform. 2021, 5, 47–55. [Google Scholar] [CrossRef] [PubMed]
  47. Houvenaeghel, G.; Cohen, M.; Classe, J.M.; Reyal, F.; Mazouni, C.; Chopin, N.; Martinez, A.; Daraï, E.; Coutant, C.; Colombo, P.E.; et al. Lymphovascular invasion has a significant prognostic impact in patients with early breast cancer, results from a large, national, multicenter, retrospective cohort study. ESMO Open. 2021, 6, 100316. [Google Scholar] [CrossRef]
  48. Burstein, H.J.; Curigliano, G.; Thürlimann, B.; Weber, W.P.; Poortmans, P.; Regan, M.M.; Senn, H.J.; Winer, E.P.; Gnant, M.; Aebi, S.; et al. Customizing local and systemic therapies for women with early breast cancer: The St. Gallen International Consensus Guidelines for treatment of early breast cancer 2021. Ann. Oncol. 2021, 32, 1216–1235. [Google Scholar] [CrossRef]
  49. Kuhn, E.; Gambini, D.; Despini, L.; Asnaghi, D.; Runza, L.; Ferrero, S. Updates on Lymphovascular Invasion in Breast Cancer. Biomedicines 2023, 11, 968. [Google Scholar] [CrossRef]
  50. Fusco, N.; Rizzo, A.; Costarelli, L.; Santinelli, A.; Cerbelli, B.; Scatena, C.; Macri, E.; Pietribiasi, F.; d’Amati, G.; Sapino, A.; et al. Pathological examination of breast cancer samples before and after neoadjuvant therapy: Recommendations from the Italian Group for the Study of Breast Pathology—Italian Society of Pathology (GIPaM-SIAPeC). Pathologica 2022, 114, 104–110. [Google Scholar] [CrossRef]
  51. Viale, G.; Fusco, N. Pathology after neoadjuvant treatment—How to assess residual disease. Breast 2021, 62, S25–S28. [Google Scholar] [CrossRef]
  52. Cucciniello, L.; Gerratana, L.; Del Mastro, L.; Puglisi, F. Tailoring adjuvant endocrine therapy in early breast cancer: When, how, and how long? Cancer Treat. Rev. 2022, 110, 102445. [Google Scholar] [CrossRef] [PubMed]
  53. Vasconcelos, I.; Hussainzada, A.; Berger, S.; Fietze, E.; Linke, J.; Siedentopf, F.; Schoenegg, W. The St. Gallen surrogate classification for breast cancer subtypes successfully predicts tumor presenting features, nodal involvement, recurrence patterns and disease free survival. Breast 2016, 29, 181–185. [Google Scholar] [CrossRef] [PubMed]
  54. Tang, P.; Tse, G.M. Immunohistochemical Surrogates for Molecular Classification of Breast Carcinoma: A 2015 Update. Arch. Pathol. Lab. Med. 2016, 140, 806–814. [Google Scholar] [CrossRef] [PubMed]
  55. Stuart-Harris, R.; Davis, A. Optimal adjuvant endocrine therapy for early breast cancer. Womens Health 2010, 6, 383–398. [Google Scholar] [CrossRef] [PubMed]
  56. Burstein, H.J.; Lacchetti, C.; Anderson, H.; Buchholz, T.A.; Davidson, N.E.; Gelmon, K.A.; Giordano, S.H.; Hudis, C.A.; Solky, A.J.; Stearns, V.; et al. Adjuvant Endocrine Therapy for Women With Hormone Receptor-Positive Breast Cancer: ASCO Clinical Practice Guideline Focused Update. J. Clin. Oncol. 2019, 37, 423–438. [Google Scholar] [CrossRef]
  57. Bidard, F.C.; Kaklamani, V.G.; Neven, P.; Streich, G.; Montero, A.J.; Forget, F.; Mouret-Reynier, M.A.; Sohn, J.H.; Taylor, D.; Harnden, K.K.; et al. Elacestrant (oral selective estrogen receptor degrader) Versus Standard Endocrine Therapy for Estrogen Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer: Results From the Randomized Phase III EMERALD Trial. J. Clin. Oncol. 2022, 40, 3246–3256. [Google Scholar] [CrossRef]
  58. Lopez, G.; Costanza, J.; Colleoni, M.; Fontana, L.; Ferrero, S.; Miozzo, M.; Fusco, N. Molecular Insights into the Classification of Luminal Breast Cancers: The Genomic Heterogeneity of Progesterone-Negative Tumors. Int. J. Mol. Sci. 2019, 20, 510. [Google Scholar] [CrossRef]
  59. Prat, A.; Pineda, E.; Adamo, B.; Galvan, P.; Fernandez, A.; Gaba, L.; Diez, M.; Viladot, M.; Arance, A.; Munoz, M. Clinical implications of the intrinsic molecular subtypes of breast cancer. Breast 2015, 24 (Suppl. S2), S26–S35. [Google Scholar] [CrossRef]
  60. Fusco, N.; Ragazzi, M.; Sajjadi, E.; Venetis, K.; Piciotti, R.; Morganti, S.; Santandrea, G.; Fanelli, G.N.; Despini, L.; Invernizzi, M.; et al. Assessment of estrogen receptor low positive status in breast cancer: Implications for pathologists and oncologists. Histol. Histopathol. 2021, 36, 1235–1245. [Google Scholar] [CrossRef]
  61. Xie, Y.; Yang, L.; Wu, Y.; Zheng, H.; Gou, Q. Adjuvant endocrine therapy in patients with estrogen receptor-low positive breast cancer: A prospective cohort study. Breast 2022, 66, 89–96. [Google Scholar] [CrossRef]
  62. Reinert, T.; Cascelli, F.; de Resende, C.A.A.; Gonçalves, A.C.; Godo, V.S.P.; Barrios, C.H. Clinical implication of low estrogen receptor (ER-low) expression in breast cancer. Front. Endocrinol. 2022, 13, 1015388. [Google Scholar] [CrossRef] [PubMed]
  63. Yu, K.D.; Cai, Y.W.; Wu, S.Y.; Shui, R.H.; Shao, Z.M. Estrogen receptor-low breast cancer: Biology chaos and treatment paradox. Cancer Commun. 2021, 41, 968–980. [Google Scholar] [CrossRef] [PubMed]
  64. Kim, M.C.; Park, M.H.; Choi, J.E.; Kang, S.H.; Bae, Y.K. Characteristics and Prognosis of Estrogen Receptor Low-Positive Breast Cancer. J. Breast Cancer 2022, 25, 318–326. [Google Scholar] [CrossRef] [PubMed]
  65. Nielsen, T.O.; Leung, S.C.Y.; Rimm, D.L.; Dodson, A.; Acs, B.; Badve, S.; Denkert, C.; Ellis, M.J.; Fineberg, S.; Flowers, M.; et al. Assessment of Ki67 in Breast Cancer: Updated Recommendations From the International Ki67 in Breast Cancer Working Group. JNCI J. Natl. Cancer Inst. 2020, 113, 808–819. [Google Scholar] [CrossRef] [PubMed]
  66. Dowsett, M.; Nielsen, T.O.; A’Hern, R.; Bartlett, J.; Coombes, R.C.; Cuzick, J.; Ellis, M.; Henry, N.L.; Hugh, J.C.; Lively, T.; et al. Assessment of Ki67 in breast cancer: Recommendations from the International Ki67 in Breast Cancer working group. J. Natl. Cancer Inst. 2011, 103, 1656–1664. [Google Scholar] [CrossRef]
  67. Hu, X.; Chen, W.; Li, F.; Ren, P.; Wu, H.; Zhang, C.; Gu, K. Expression changes of ER, PR, HER2, and Ki-67 in primary and metastatic breast cancer and its clinical significance. Front. Oncol. 2023, 13, 1053125. [Google Scholar] [CrossRef]
  68. Najjar, S.; Allison, K.H. Updates on breast biomarkers. Virchows Arch. 2022, 480, 163–176. [Google Scholar] [CrossRef] [PubMed]
  69. Duffy, M.J.; Harbeck, N.; Nap, M.; Molina, R.; Nicolini, A.; Senkus, E.; Cardoso, F. Clinical use of biomarkers in breast cancer: Updated guidelines from the European Group on Tumor Markers (EGTM). Eur. J. Cancer 2017, 75, 284–298. [Google Scholar] [CrossRef]
  70. Kreipe, H.; Harbeck, N.; Christgen, M. Clinical validity and clinical utility of Ki67 in early breast cancer. Ther. Adv. Med. Oncol. 2022, 14, 17588359221122725. [Google Scholar] [CrossRef]
  71. Escala Cornejo, R.A.; Muñoz García, M.; Olivares Hernández, A.; Sancho de Salas, M.; Gómez muñoz, M.A.; Claros Ampuero, J.; Figuero Pérez, L.; Escalera Martín, E.; Barrios Collado, B.; Martín García, G.; et al. 215P Identifying the best Ki67 cut-off for determining luminal breast cancer subtypes using immunohistochemical analysis and PAM50 genomic classification. Ann. Oncol. 2020, 31, S327. [Google Scholar] [CrossRef]
  72. Lombardi, A.; Lazzeroni, R.; Bersigotti, L.; Vitale, V.; Amanti, C. The Proper Ki-67 Cut-Off in Hormone Responsive Breast Cancer: A Monoinstitutional Analysis with Long-Term Follow-Up. Breast Cancer 2021, 13, 213–217. [Google Scholar] [CrossRef] [PubMed]
  73. Andre, F.; Ismaila, N.; Allison, K.H.; Barlow, W.E.; Collyar, D.E.; Damodaran, S.; Henry, N.L.; Jhaveri, K.; Kalinsky, K.; Kuderer, N.M.; et al. Biomarkers for Adjuvant Endocrine and Chemotherapy in Early-Stage Breast Cancer: ASCO Guideline Update. J. Clin. Oncol. 2022, 40, 1816–1837. [Google Scholar] [CrossRef] [PubMed]
  74. Giordano, S.H.; Freedman, R.A.; Somerfield, M.R. Abemaciclib With Endocrine Therapy in the Treatment of High-Risk Early Breast Cancer: ASCO Optimal Adjuvant Chemotherapy and Targeted Therapy Guideline Rapid Recommendation Update. J. Clin. Oncol. 2021, 40, 307–309. [Google Scholar] [CrossRef] [PubMed]
  75. de Gregorio, A.; Friedl, T.W.P.; Hering, E.; Widschwendter, P.; de Gregorio, N.; Bekes, I.; Janni, W.; Dayan, D.; Huober, J.B. Ki67 as Proliferative Marker in Patients with Early Breast Cancer and Its Association with Clinicopathological Factors. Oncology 2021, 99, 780–789. [Google Scholar] [CrossRef] [PubMed]
  76. Li, L.; Han, D.; Yu, Y.; Li, J.; Liu, Y. Artificial intelligence-assisted interpretation of Ki-67 expression and repeatability in breast cancer. Diagn. Pathol. 2022, 17, 20. [Google Scholar] [CrossRef]
  77. Wolff, A.C.; Hammond, M.E.H.; Allison, K.H.; Harvey, B.E.; Mangu, P.B.; Bartlett, J.M.S.; Bilous, M.; Ellis, I.O.; Fitzgibbons, P.; Hanna, W.; et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J. Clin. Oncol. 2018, 36, 2105–2122. [Google Scholar] [CrossRef]
  78. Pathmanathan, N.; Provan, P.J.; Mahajan, H.; Hall, G.; Byth, K.; Bilous, A.M.; Balleine, R.L. Characteristics of HER2-positive breast cancer diagnosed following the introduction of universal HER2 testing. Breast 2012, 21, 724–729. [Google Scholar] [CrossRef]
  79. O’Sullivan, C.C.; Swain, S.M. Pertuzumab: Evolving therapeutic strategies in the management of HER2-overexpressing breast cancer. Expert. Opin. Biol. Ther. 2013, 13, 779–790. [Google Scholar] [CrossRef]
  80. Perez, E.A.; Romond, E.H.; Suman, V.J.; Jeong, J.H.; Sledge, G.; Geyer, C.E., Jr.; Martino, S.; Rastogi, P.; Gralow, J.; Swain, S.M.; et al. Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: Planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831. J. Clin. Oncol. 2014, 32, 3744–3752. [Google Scholar] [CrossRef]
  81. 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: ASCO–College of American Pathologists Guideline Update. J. Clin. Oncol. 2023, 41, 3867–3872. [Google Scholar] [CrossRef]
  82. Tsang, J.Y.; Tse, G.M. Update on triple-negative breast cancers - highlighting subtyping update and treatment implication. Histopathology 2023, 82, 17–35. [Google Scholar] [CrossRef] [PubMed]
  83. 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] [PubMed]
  84. Nicolò, E.; Boscolo Bielo, L.; Curigliano, G.; Tarantino, P. The HER2-low revolution in breast oncology: Steps forward and emerging challenges. Ther. Adv. Med. Oncol. 2023, 15, 17588359231152842. [Google Scholar] [CrossRef] [PubMed]
  85. Diéras, V.; Deluche, E.; Lusque, A.; Pistilli, B.; Bachelot, T.; Pierga, J.-Y.; Viret, F.; Levy, C.; Salabert, L.; Du, F.L. Abstract PD8-02: Trastuzumab deruxtecan (T-DXd) for advanced breast cancer patients (ABC), regardless HER2 status: A phase II study with biomarkers analysis (DAISY). Cancer Res. 2022, 82, PD8-02. [Google Scholar] [CrossRef]
  86. Pruneri, G.; Vingiani, A.; Denkert, C. Tumor infiltrating lymphocytes in early breast cancer. Breast 2018, 37, 207–214. [Google Scholar] [CrossRef]
  87. Salgado, R.; Denkert, C.; Demaria, S.; Sirtaine, N.; Klauschen, F.; Pruneri, G.; Wienert, S.; Van den Eynden, G.; Baehner, F.L.; Penault-Llorca, F.; et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: Recommendations by an International TILs Working Group 2014. Ann. Oncol. 2015, 26, 259–271. [Google Scholar] [CrossRef]
  88. Kim, R.S.; Song, N.; Gavin, P.G.; Salgado, R.; Bandos, H.; Kos, Z.; Floris, G.; Eynden, G.G.G.M.V.d.; Badve, S.; Demaria, S.; et al. Stromal Tumor-infiltrating Lymphocytes in NRG Oncology/NSABP B-31 Adjuvant Trial for Early-Stage HER2-Positive Breast Cancer. J. Natl. Cancer Inst. 2019, 111, 867–871. [Google Scholar] [CrossRef]
  89. Li, S.; Zhang, Y.; Zhang, P.; Xue, S.; Chen, Y.; Sun, L.; Yang, R. Predictive and prognostic values of tumor infiltrating lymphocytes in breast cancers treated with neoadjuvant chemotherapy: A meta-analysis. Breast 2022, 66, 97–109. [Google Scholar] [CrossRef]
  90. Mao, Y.; Qu, Q.; Zhang, Y.; Liu, J.; Chen, X.; Shen, K. The Value of Tumor Infiltrating Lymphocytes (TILs) for Predicting Response to Neoadjuvant Chemotherapy in Breast Cancer: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e115103. [Google Scholar] [CrossRef]
  91. Denkert, C.; von Minckwitz, G.; Darb-Esfahani, S.; Lederer, B.; Heppner, B.I.; Weber, K.E.; Budczies, J.; Huober, J.; Klauschen, F.; Furlanetto, J.; 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. [Google Scholar] [CrossRef]
  92. Salgado, R.; Denkert, C.; Campbell, C.; Savas, P.; Nuciforo, P.; Aura, C.; de Azambuja, E.; Eidtmann, H.; Ellis, C.E.; Baselga, J.; 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, 448–454. [Google Scholar] [CrossRef] [PubMed]
  93. Loi, S.; Drubay, D.; Adams, S.; Pruneri, G.; Francis, P.A.; Lacroix-Triki, M.; Joensuu, H.; Dieci, M.V.; Badve, S.; Demaria, S.; et al. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J. Clin. Oncol. 2019, 37, 559–569. [Google Scholar] [CrossRef] [PubMed]
  94. Loi, S.; Sirtaine, N.; Piette, F.; Salgado, R.; Viale, G.; Van Eenoo, F.; Rouas, G.; Francis, P.; Crown, J.P.; Hitre, E.; et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG 02-98. J. Clin. Oncol. 2013, 31, 860–867. [Google Scholar] [CrossRef] [PubMed]
  95. Loi, S.; Michiels, S.; Salgado, R.; Sirtaine, N.; Jose, V.; Fumagalli, D.; Kellokumpu-Lehtinen, P.L.; Bono, P.; Kataja, V.; Desmedt, C.; et al. Tumor infiltrating lymphocytes are prognostic in triple negative breast cancer and predictive for trastuzumab benefit in early breast cancer: Results from the FinHER trial. Ann. Oncol. 2014, 25, 1544–1550. [Google Scholar] [CrossRef]
  96. Loi, S.; Salgado, R.; Adams, S.; Pruneri, G.; Francis, P.A.; Lacroix-Triki, M.; Joensuu, H.; Dieci, M.V.; Badve, S.; Demaria, S.; et al. Tumor infiltrating lymphocyte stratification of prognostic staging of early-stage triple negative breast cancer. NPJ Breast Cancer 2022, 8, 3. [Google Scholar] [CrossRef]
  97. Sajjadi, E.; Venetis, K.; Ivanova, M.; Noale, M.; Blundo, C.; Di Loreto, E.; Scarfone, G.; Ferrero, S.; Maggi, S.; Veronesi, P.; et al. Immune microenvironment dynamics in breast cancer during pregnancy: Impact of gestational age on tumor-infiltrating lymphocytes and prognosis. Front. Oncol. 2023, 13, 1116569. [Google Scholar] [CrossRef]
  98. Park, J.H.; Jonas, S.F.; Bataillon, G.; Criscitiello, C.; Salgado, R.; Loi, S.; Viale, G.; Lee, H.J.; Dieci, M.V.; Kim, S.B.; et al. Prognostic value of tumor-infiltrating lymphocytes in patients with early-stage triple-negative breast cancers (TNBC) who did not receive adjuvant chemotherapy. Ann. Oncol. 2019, 30, 1941–1949. [Google Scholar] [CrossRef]
  99. Loibl, S.; Untch, M.; Burchardi, N.; Huober, J.; Sinn, B.V.; Blohmer, J.U.; Grischke, E.M.; Furlanetto, J.; Tesch, H.; Hanusch, C.; et al. A randomised phase II study investigating durvalumab in addition to an anthracycline taxane-based neoadjuvant therapy in early triple-negative breast cancer: Clinical results and biomarker analysis of GeparNuevo study. Ann. Oncol. 2019, 30, 1279–1288. [Google Scholar] [CrossRef]
  100. El Bairi, K.; Haynes, H.R.; Blackley, E.; Fineberg, S.; Shear, J.; Turner, S.; de Freitas, J.R.; Sur, D.; Amendola, L.C.; Gharib, M.; et al. The tale of TILs in breast cancer: A report from The International Immuno-Oncology Biomarker Working Group. npj Breast Cancer 2021, 7, 150. [Google Scholar] [CrossRef]
  101. Karn, T.; Denkert, C.; Weber, K.E.; Holtrich, U.; Hanusch, C.; Sinn, B.V.; Higgs, B.W.; Jank, P.; Sinn, H.P.; Huober, J.; et al. Tumor mutational burden and immune infiltration as independent predictors of response to neoadjuvant immune checkpoint inhibition in early TNBC in GeparNuevo. Ann. Oncol. 2020, 31, 1216–1222. [Google Scholar] [CrossRef]
  102. Sajjadi, E.; Venetis, K.; Noale, M.; Azim, H.A., Jr.; Blundo, C.; Bonizzi, G.; Di Loreto, E.; Scarfone, G.; Ferrero, S.; Maggi, S.; et al. Breast Cancer during Pregnancy as a Special Type of Early-Onset Breast Cancer: Analysis of the Tumor Immune Microenvironment and Risk Profiles. Cells 2022, 11, 2286. [Google Scholar] [CrossRef] [PubMed]
  103. Mittendorf, E.A.; Zhang, H.; Barrios, C.H.; Saji, S.; Jung, K.H.; Hegg, R.; Koehler, A.; Sohn, J.; Iwata, H.; Telli, M.L.; et al. Neoadjuvant atezolizumab in combination with sequential nab-paclitaxel and anthracycline-based chemotherapy versus placebo and chemotherapy in patients with early-stage triple-negative breast cancer (IMpassion031): A randomised, double-blind, phase 3 trial. Lancet 2020, 396, 1090–1100. [Google Scholar] [CrossRef] [PubMed]
  104. Schmid, P.; Cortes, J.; Pusztai, L.; McArthur, H.; Kümmel, S.; Bergh, J.; Denkert, C.; Park, Y.H.; Hui, R.; Harbeck, N.; et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N. Engl. J. Med. 2020, 382, 810–821. [Google Scholar] [CrossRef] [PubMed]
  105. Nanda, R.; Liu, M.C.; Yau, C.; Shatsky, R.; Pusztai, L.; Wallace, A.; Chien, A.J.; Forero-Torres, A.; Ellis, E.; Han, H.; et al. Effect of Pembrolizumab Plus Neoadjuvant Chemotherapy on Pathologic Complete Response in Women With Early-Stage Breast Cancer: An Analysis of the Ongoing Phase 2 Adaptively Randomized I-SPY2 Trial. JAMA Oncol. 2020, 6, 676–684. [Google Scholar] [CrossRef] [PubMed]
  106. Balic, M.; Thomssen, C.; Würstlein, R.; Gnant, M.; St. Harbeck, N. Gallen/Vienna 2019: A Brief Summary of the Consensus Discussion on the Optimal Primary Breast Cancer Treatment. Breast Care 2019, 14, 103–110. [Google Scholar] [CrossRef]
  107. Balic, M.; Thomssen, C.; Gnant, M.; St. Harbeck, N. Gallen/Vienna 2023: Optimization of Treatment for Patients with Primary Breast Cancer—A Brief Summary of the Consensus Discussion. Breast Care 2023, 18, 213–222. [Google Scholar] [CrossRef]
  108. Le, H.; Gupta, R.; Hou, L.; Abousamra, S.; Fassler, D.; Torre-Healy, L.; Moffitt, R.A.; Kurc, T.; Samaras, D.; Batiste, R.; et al. Utilizing Automated Breast Cancer Detection to Identify Spatial Distributions of Tumor-Infiltrating Lymphocytes in Invasive Breast Cancer. Am. J. Pathol. 2020, 190, 1491–1504. [Google Scholar] [CrossRef] [PubMed]
  109. Porta, F.M.; Sajjadi, E.; Venetis, K.; Frascarelli, C.; Cursano, G.; Guerini-Rocco, E.; Fusco, N.; Ivanova, M. Immune Biomarkers in Triple-Negative Breast Cancer: Improving the Predictivity of Current Testing Methods. J. Pers. Med. 2023, 13, 1176. [Google Scholar] [CrossRef]
  110. Oliveira, L.J.C.; Amorim, L.C.; Megid, T.B.C.; de Resende, C.A.A.; Mano, M.S. Gene expression signatures in early breast cancer: Better together with clinicopathological features. Crit. Rev. Oncol. Hematol. 2022, 175, 103708. [Google Scholar] [CrossRef]
  111. Bou Zerdan, M.; Ibrahim, M.; El Nakib, C.; Hajjar, R.; Assi, H.I. Genomic Assays in Node Positive Breast Cancer Patients: A Review. Front. Oncol. 2021, 10, 609100. [Google Scholar] [CrossRef]
  112. Dias, K.; Dvorkin-Gheva, A.; Hallett, R.M.; Wu, Y.; Hassell, J.; Pond, G.R.; Levine, M.; Whelan, T.; Bane, A.L. Claudin-Low Breast Cancer; Clinical & Pathological Characteristics. PLoS ONE 2017, 12, e0168669. [Google Scholar] [CrossRef]
  113. Fougner, C.; Bergholtz, H.; Norum, J.H.; Sørlie, T. Re-definition of claudin-low as a breast cancer phenotype. Nat. Commun. 2020, 11, 1787. [Google Scholar] [CrossRef] [PubMed]
  114. Prat, A.; Parker, J.S.; Karginova, O.; Fan, C.; Livasy, C.; Herschkowitz, J.I.; He, X.; Perou, C.M. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010, 12, 1–18. [Google Scholar] [CrossRef] [PubMed]
  115. Lehmann-Che, J.; Hamy, A.S.; Porcher, R.; Barritault, M.; Bouhidel, F.; Habuellelah, H.; Leman-Detours, S.; de Roquancourt, A.; Cahen-Doidy, L.; Bourstyn, E.; et al. Molecular apocrine breast cancers are aggressive estrogen receptor negative tumors overexpressing either HER2 or GCDFP15. Breast Cancer Res. 2013, 15, R37. [Google Scholar] [CrossRef]
  116. Wan, A.; Zhang, G.; Ma, D.; Zhang, Y.; Qi, X. An overview of the research progress of BRCA gene mutations in breast cancer. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188907. [Google Scholar] [CrossRef]
  117. Cortesi, L.; Rugo, H.S.; Jackisch, C. An Overview of PARP Inhibitors for the Treatment of Breast Cancer. Target. Oncol. 2021, 16, 255–282. [Google Scholar] [CrossRef]
  118. Tutt, A.N.J.; Garber, J.E.; Kaufman, B.; Viale, G.; Fumagalli, D.; Rastogi, P.; Gelber, R.D.; de Azambuja, E.; Fielding, A.; Balmaña, J.; et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef]
  119. Geyer, C.E., Jr.; Garber, J.E.; Gelber, R.D.; Yothers, G.; Taboada, M.; Ross, L.; Rastogi, P.; Cui, K.; Arahmani, A.; Aktan, G.; et al. Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer. Ann. Oncol. 2022, 33, 1250–1268. [Google Scholar] [CrossRef]
  120. Eikesdal, H.P.; Yndestad, S.; Blix, E.S.; Lundgren, S.; Vagstad, G.; Espelid, H.; Gilje, B.; Janssen, E.A.; Geisler, J.; Aas, T.; et al. Neoadjuvant olaparib monotherapy in primary triple negative breast cancer. Ann. Oncol. 2019, 30, v60. [Google Scholar] [CrossRef]
  121. Spring, L.M.; Han, H.; Liu, M.C.; Hamilton, E.; Irie, H.; Santa-Maria, C.A.; Reeves, J.; Pan, P.; Shan, M.; Tang, Y.; et al. Neoadjuvant study of niraparib in patients with HER2-negative, BRCA-mutated, resectable breast cancer. Nat. Cancer 2022, 3, 927–931. [Google Scholar] [CrossRef]
  122. Litton, J.K.; Scoggins, M.E.; Hess, K.R.; Adrada, B.E.; Murthy, R.K.; Damodaran, S.; DeSnyder, S.M.; Brewster, A.M.; Barcenas, C.H.; Valero, V.; et al. Neoadjuvant Talazoparib for Patients With Operable Breast Cancer With a Germline BRCA Pathogenic Variant. J. Clin. Oncol. 2020, 38, 388–394. [Google Scholar] [CrossRef]
  123. Caramelo, O.; Silva, C.; Caramelo, F.; Frutuoso, C.; Pinto, L.; Almeida-Santos, T. Efficacy of different neoadjuvant treatment regimens in BRCA-mutated triple negative breast cancer: A systematic review and meta-analysis. Hered. Cancer Clin. Pract. 2022, 20, 34. [Google Scholar] [CrossRef] [PubMed]
  124. Yoshida, R. Hereditary breast and ovarian cancer (HBOC): Review of its molecular characteristics, screening, treatment, and prognosis. Breast Cancer 2021, 28, 1167–1180. [Google Scholar] [CrossRef] [PubMed]
  125. t’Kint de Roodenbeke, M.D.; Pondé, N.; Buisseret, L.; Piccart, M. Management of early breast cancer in patients bearing germline BRCA mutations. Semin. Oncol. 2020, 47, 243–248. [Google Scholar] [CrossRef] [PubMed]
  126. Sinha, I.; Fogle, R.L.; Gulfidan, G.; Stanley, A.E.; Walter, V.; Hollenbeak, C.S.; Arga, K.Y.; Sinha, R. Potential Early Markers for Breast Cancer: A Proteomic Approach Comparing Saliva and Serum Samples in a Pilot Study. Int. J. Mol. Sci. 2023, 24, 4164. [Google Scholar] [CrossRef] [PubMed]
  127. Pathania, S.; Khan, M.I.; Kumar, A.; Gupta, A.K.; Rani, K.; Ramesh Parashar, T.; Jayaram, J.; Ranjan Mishra, P.; Srivastava, A.; Mathur, S.; et al. Proteomics of Sentinel Lymph Nodes in Early Breast Cancer for Identification of Thymidylate Synthase as a Potential Biomarker to Flag Metastasis: A Preliminary Study. Cancer Manag. Res. 2020, 12, 4841–4854. [Google Scholar] [CrossRef]
  128. Asleh, K.; Negri, G.L.; Spencer Miko, S.E.; Colborne, S.; Hughes, C.S.; Wang, X.Q.; Gao, D.; Gilks, C.B.; Chia, S.K.L.; Nielsen, T.O.; et al. Proteomic analysis of archival breast cancer clinical specimens identifies biological subtypes with distinct survival outcomes. Nat. Commun. 2022, 13, 896. [Google Scholar] [CrossRef]
  129. Da Cunha, P.A.; Nitusca, D.; Canto, L.M.D.; Varghese, R.S.; Ressom, H.W.; Willey, S.; Marian, C.; Haddad, B.R. Metabolomic Analysis of Plasma from Breast Cancer Patients Using Ultra-High-Performance Liquid Chromatography Coupled with Mass Spectrometry: An Untargeted Study. Metabolites 2022, 12, 447. [Google Scholar] [CrossRef]
  130. Wei, Y.; Jasbi, P.; Shi, X.; Turner, C.; Hrovat, J.; Liu, L.; Rabena, Y.; Porter, P.; Gu, H. Early Breast Cancer Detection Using Untargeted and Targeted Metabolomics. J. Proteome Res. 2021, 20, 3124–3133. [Google Scholar] [CrossRef]
  131. Eghlimi, R.; Shi, X.; Hrovat, J.; Xi, B.; Gu, H. Triple Negative Breast Cancer Detection Using LC-MS/MS Lipidomic Profiling. J. Proteome Res. 2020, 19, 2367–2378. [Google Scholar] [CrossRef]
  132. Chistyakov, D.V.; Guryleva, M.V.; Stepanova, E.S.; Makarenkova, L.M.; Ptitsyna, E.V.; Goriainov, S.V.; Nikolskaya, A.I.; Astakhova, A.A.; Klimenko, A.S.; Bezborodova, O.A.; et al. Multi-Omics Approach Points to the Importance of Oxylipins Metabolism in Early-Stage Breast Cancer. Cancers 2022, 14, 2041. [Google Scholar] [CrossRef] [PubMed]
  133. Sammut, S.-J.; Crispin-Ortuzar, M.; Chin, S.-F.; Provenzano, E.; Bardwell, H.A.; Ma, W.; Cope, W.; Dariush, A.; Dawson, S.-J.; Abraham, J.E.; et al. Multi-omic machine learning predictor of breast cancer therapy response. Nature 2022, 601, 623–629. [Google Scholar] [CrossRef] [PubMed]
  134. Choi, J.M.; Chae, H. moBRCA-net: A breast cancer subtype classification framework based on multi-omics attention neural networks. BMC Bioinform. 2023, 24, 169. [Google Scholar] [CrossRef] [PubMed]
  135. Ortiz, M.M.O.; Andrechek, E.R. Molecular Characterization and Landscape of Breast cancer Models from a multi-omics Perspective. J. Mammary Gland. Biol. Neoplasia 2023, 28, 12. [Google Scholar] [CrossRef]
  136. Garcia-Recio, S.; Hinoue, T.; Wheeler, G.L.; Kelly, B.J.; Garrido-Castro, A.C.; Pascual, T.; De Cubas, A.A.; Xia, Y.; Felsheim, B.M.; McClure, M.B.; et al. Multiomics in primary and metastatic breast tumors from the AURORA US network finds microenvironment and epigenetic drivers of metastasis. Nat. Cancer 2023, 4, 128–147. [Google Scholar] [CrossRef]
  137. Sajjadi, E.; Frascarelli, C.; Venetis, K.; Bonizzi, G.; Ivanova, M.; Vago, G.; Guerini-Rocco, E.; Fusco, N. Computational pathology to improve biomarker testing in breast cancer: How close are we? Eur. J. Cancer Prev. 2023, 32, 460–467. [Google Scholar] [CrossRef]
Figure 1. The risk assessment of early breast cancer (BC) involves analyzing various biomarkers and clinicopathologic features. Traditional histopathological characteristics like tumor size, lymph node involvement, histological grade, and lymphovascular invasion form the core of the pathology report, offering essential prognostic information. Some pathologists may report TILs, especially in TNBC and HER2+ BC, although current recommendations do not suggest basing therapeutic strategies on TILs due to their lack of clinical utility. Immunohistochemical assessment of hormone receptor status, HER2 status, and Ki67 is equally vital, reflecting the luminal/non-luminal molecular classification and guiding treatment choices with both prognostic and predictive implications. Integrating gene profiling assays into early BC evaluation optimizes adjuvant treatment decisions, especially for post-menopausal luminal patients with or without 1–3 node metastases and pre-menopausal luminal patients without lymph node involvement. In cases suggestive of hereditary BC syndrome, BRCA1/2 testing is recommended. Abbreviations: BC, breast cancer; TILs, tumor-infiltrating lymphocytes; ER, estrogen receptor; PgR, progesterone receptor; TNBC, triple-negative BC.
Figure 1. The risk assessment of early breast cancer (BC) involves analyzing various biomarkers and clinicopathologic features. Traditional histopathological characteristics like tumor size, lymph node involvement, histological grade, and lymphovascular invasion form the core of the pathology report, offering essential prognostic information. Some pathologists may report TILs, especially in TNBC and HER2+ BC, although current recommendations do not suggest basing therapeutic strategies on TILs due to their lack of clinical utility. Immunohistochemical assessment of hormone receptor status, HER2 status, and Ki67 is equally vital, reflecting the luminal/non-luminal molecular classification and guiding treatment choices with both prognostic and predictive implications. Integrating gene profiling assays into early BC evaluation optimizes adjuvant treatment decisions, especially for post-menopausal luminal patients with or without 1–3 node metastases and pre-menopausal luminal patients without lymph node involvement. In cases suggestive of hereditary BC syndrome, BRCA1/2 testing is recommended. Abbreviations: BC, breast cancer; TILs, tumor-infiltrating lymphocytes; ER, estrogen receptor; PgR, progesterone receptor; TNBC, triple-negative BC.
Cancers 15 05430 g001
Table 1. Recommended genomic tests in HR+/HER2- early breast cancer based on menopausal status, patient’s age, and number of lymph node metastases, according to the latest ASCO recommendations on gene profile assays, adapted from Andre F. et al. [73]. (***) high evidence quality and strong recommendation; (**) intermediate evidence quality and strong recommendation; (*) intermediate evidence quality and moderate recommendation.
Table 1. Recommended genomic tests in HR+/HER2- early breast cancer based on menopausal status, patient’s age, and number of lymph node metastases, according to the latest ASCO recommendations on gene profile assays, adapted from Andre F. et al. [73]. (***) high evidence quality and strong recommendation; (**) intermediate evidence quality and strong recommendation; (*) intermediate evidence quality and moderate recommendation.
NPremenopausal or ≤50 Years OldPostmenopausal or >50 Years Old
pN0Oncotype Dx (***)Oncotype Dx (***)
MammaPrint (**)
EndoPredict (*)
Prosigna (*)
BCI (*)
pN1a-cNot recommendedOncotype Dx (***)
MammaPrint (**)
EndoPredict (*)
BCI (*)
pN2Not recommendedNot recommended
Table 2. ESMO recommendations on gene profiling assays in HR+/HER2- early breast cancer, adapted from Cardoso et al. [3]. LoE, level of evidence; GoR, grade of recommendation.
Table 2. ESMO recommendations on gene profiling assays in HR+/HER2- early breast cancer, adapted from Cardoso et al. [3]. LoE, level of evidence; GoR, grade of recommendation.
Genomic SignaturesMethodLoEGoR
Oncotype DxqRT-PCRIA
MammaPrintDNA microarrayIA
ProsignanCounterIB
EndoPredictqRT-PCRIB
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

Pescia, C.; Guerini-Rocco, E.; Viale, G.; Fusco, N. Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies. Cancers 2023, 15, 5430. https://doi.org/10.3390/cancers15225430

AMA Style

Pescia C, Guerini-Rocco E, Viale G, Fusco N. Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies. Cancers. 2023; 15(22):5430. https://doi.org/10.3390/cancers15225430

Chicago/Turabian Style

Pescia, Carlo, Elena Guerini-Rocco, Giuseppe Viale, and Nicola Fusco. 2023. "Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies" Cancers 15, no. 22: 5430. https://doi.org/10.3390/cancers15225430

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