Next Article in Journal
The Ethical Complexity of Medical Decision Making in the Adolescent Oncology Patient
Previous Article in Journal
MRI-Targeted Prostate Fusion Biopsy: What Are We Missing outside the Target? Implications for Treatment Planning
Previous Article in Special Issue
Multidisciplinary and Tailored Treatment of Locally Advanced Breast Cancer in Progression during Neoadjuvant Chemotherapy: Case Report
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Insight for Axillary De-Escalation in Breast Cancer Surgery: “SoFT Study” Retrospective Analysis

by
Gianluca Vanni
1,
Marco Materazzo
1,2,*,
Floriana Paduano
1,
Marco Pellicciaro
1,2,
Giordana Di Mauro
3,
Enrica Toscano
3,
Federico Tacconi
4,
Benedetto Longo
1,5,
Valerio Cervelli
5,
Massimiliano Berretta
6,† and
Oreste Claudio Buonomo
1,7,†
1
Breast Unit, Department of Surgical Science, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy
2
Ph.D. Program in Applied Medical-Surgical Sciences, Department of Surgical Science, University of Rome “Tor Vergata”, Viale Oxford 81, 00133 Rome, Italy
3
Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
4
Department of Surgical Sciences, Unit of Thoracic Surgery, Tor Vergata University, 00133 Rome, Italy
5
Plastic and Reconstructive Surgery at Department of Surgical Science, Tor Vergata University of Rome, Via Montpellier 1, 00133 Rome, Italy
6
Department of Clinical and Experimental Medicine, University of Messina, 98100 Messina, Italy
7
General Surgery Program, Department of Health Science, UNIBAS, University of Basilicata, Via dell’Ateneo Lucano, 10, 85100 Potenza, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work and are co-last authors.
Curr. Oncol. 2024, 31(8), 4141-4157; https://doi.org/10.3390/curroncol31080309 (registering DOI)
Submission received: 21 June 2024 / Revised: 20 July 2024 / Accepted: 21 July 2024 / Published: 23 July 2024
(This article belongs to the Collection New Insights into Breast Cancer Diagnosis and Treatment)

Abstract

:
Background: The SOUND study demonstrated that an axillary de-escalation may be sufficient in locoregional and distant disease control in selected early breast cancer (EBC) patients. To establish any preoperative variables that may drive sentinel lymph node biopsy (SLNB) omission, a study named sentinel omission risk factor (SOFT) 1.23 was planned. Methods: A single-center retrospective study from a prospectively maintained database was designed, aiming at underlying preoperative prognostic factors involved in sentinel lymph node (SLN) metastasis (lymph node involvement (LN+) vs. negative lymph node (LN−) group). Secondary outcomes included surgical room occupancy analysis for SLNB in patients fulfilling the SOUND study inclusion criteria. The institutional ethical committee Area Territoriale Lazio 2 approved the study (n° 122/23). Results: Between 1 January 2022 and 30 June 2023, 160 patients were included in the study and 26 (%) were included in the LN+ group. Multifocality, higher cT stage, and larger tumor diameter were reported in the LN+ group (p = 0.020, p = 0.014, and 0.016, respectively). Tumor biology, including estrogen and progesterone receptors, and molecular subtypes showed association with the LN+ group (p < 0.001; p = 0.001; and p = 0.001, respectively). A total of 117 (73.6%) patients were eligible for the SOUND study and the potential operating room time saved was 2696.81 min. Conclusions: De-escalating strategies may rationalize healthcare activities. Multifactorial risk stratification may further refine the selection of patients who could benefit from SLNB omission.

1. Introduction

In the past century, sentinel lymph node biopsy (SLNB) emerged as the standard of care in patients with early breast cancer (EBC). In milestone papers, Veronesi et al. and Karg et al. aimed to perform axillary surgical de-escalation in EBC, given the equivalent oncological outcome of SLNB compared with axillary lymph node dissection (ALND) [1,2]. While previously ALND was considered the gold standard, in both clinical trials, ALND with curative intent in EBC was preserved only for cases of SLN involvement.
In 2017, the results of the 10-year American College of Surgeons Oncology Group Z0011 (ACOSOGZ0011) represented another milestone in the de-escalation of axillary surgical management. The ACOSOGZ0011 study demonstrated that even in cases of limited axillary disease in patients undergoing breast-conserving surgery (BCS), and medical and radiation adjuvant treatment, the omission of ALND did not lead to detrimental effects on locoregional and distant outcomes [3]. Once confirmed by other randomized clinical trials, the safe omission of ALND required new paradigms and strategies in locoregional axillary treatment [4,5,6,7].
In the modern era of breast surgery, while evidence from the ACOSOGZ0011 trial demonstrated reduced morbidity for a significant cohort of EBC patients, these results led to a reevaluation of the clinical significance of SLNB in the era of genomic assessment and consequently considerations of alternatives to surgical axillary staging [8]. From this perspective, the recent SOUND study aimed to demonstrate that a safe complete de-escalation with axillary lymph node (ALN) ultrasound may be sufficient to control local and distant disease without affecting adjuvant treatment [9]. While the cutting-edge evidence from the SOUND study will be practice-changing for a significant proportion of patients, many authors have started to explore the limitations of the study and determine which will be the real population that will eventually benefit from this study [10,11]. In order to establish whether other patients may safely omit SLNB, a study named sentinel omission risk factor (SOFT) 1.23 was planned in our department. Therefore, the aim of the present paper is to identify any preoperative variables that may be applied to the selection of a wider population where axillary invasive assessment may be safely omitted, and evaluate the potential beneficial effect of de-escalating strategies on surgical room occupancy.

2. Materials and Methods

A single-center retrospective study from a prospectively maintained database, named SOFT 1.23, was planned. The primary endpoint of the study was to underline the preoperative prognostic factors related to lymph node (LN) and/or SLN metastasis, aiming at surgical de-escalation. The institutional ethical committee Area Territoriale Lazio 2 approved the study (n° 122/23). Therefore, all consecutive patients fulfilling the inclusion criteria were enrolled between 1 January 2022 and 30 June 2023. Secondary outcomes included the analysis of surgical room occupancy for SLNB in patients fulfilling the inclusion criteria for the SOUND study population.

2.1. Patients’ Characteristics

All consecutive invasive EBC patients (cT1a-2 cN0 cM0) scheduled for BCS and SLNB were included in the study. Additional inclusion criteria were age ≥ 18 years, undergoing an upfront surgical procedure without prior primary medical treatment. All patients enrolled prior to surgery required a clinical examination, bilateral mammogram, breast and axillary ultrasound, and histological breast cancer (BC) diagnosis (triple assessment). For patients under the age of 40 years, a mammogram was not considered mandatory, and core needle biopsy (CNB) or vacuum-assisted biopsy (VAB) were considered suitable for preoperative BC diagnosis. Patients who underwent diagnostic lumpectomy were excluded from the study. Bilateral contrast enhancement magnetic resonance or bilateral contrast enhancement mammogram were not considered mandatory for preoperative assessment. Moreover, patients with distant metastasis, a medical history of thoracic radiotherapy, ipsilateral breast or axillary surgical treatment that could interfere with ALN appearance, and pregnancy were excluded from the analysis. Before admission to our facility, all patients signed a general study consent for enrollment in our database. Moreover, once included in the study, all patients provided a specific written consent for the study.

2.2. Data Collection

A retrospective analysis of our prospectively maintained database was performed. Demographic data analyzed in the study included age, age at menarche, age at diagnosis, and body mass index (BMI). BC diameter was retrieved from the radiological reports of ultrasound and mammogram. The clinical stage was assessed according to the AJCC 2018 eighth edition of the TNM classification with clinical examination, bilateral mammogram, breast and axillary ultrasound, and histological BC diagnosis [12]. BC preoperative characteristics regarding tumor dimensions and biomolecular features were included in the analysis, including histotype, estrogen receptor (ER), progesterone receptor (PR), Ki67, and epidermal growth factor receptor-2 (HER2) score according to the ASCO HER-2 2018 guideline update [13]. Further analysis included tumor grade according to the Nottingham histologic score system (the Elston–Ellis modification of Scarff–Bloom–Richardson grading system), and biomolecular classification according to the 2013 San Gallen criteria [14]. Intraoperative data were retrieved from surgical notes and single surgical procedures were calculated separately (breast-conserving surgery and SLNB). Due to internal policy, SLNB frozen sections were performed in the case of clinical suspicion, when SLN were enlarged >1 cm, or when clinical involvement was suspected during surgery. Intraoperative staining and definitive staining were assessed for pN.

2.3. Statistical Analysis

All data were collected in a prospectively maintained database (Excel 2016 ver 2406, Microsoft 365, Washington, DC, USA). Statistical analysis was conducted using the statistical package for the social sciences (SPSS v.15.0; SPSS, Inc., Chicago, IL, USA). Patients were grouped based on ALN involvement according to the definitive pNx(sn) stage (pN0(sn) for LN− and pN1(sn) for LN+). Continuous variables were expressed as medians and interquartile ranges (IQR), while categorical variables were presented as frequencies and percentages. Continuous variables between groups were compared using the Student’s t-test or Mann–Whitney U test, based on the Kolmogorov–Smirnov test results. Categorical and dichotomous variables were compared using the chi-square test or Fisher’s exact test, depending on the sample size. For multiple categorical variables, Monte Carlo correction was applied to both tests (e.g., T stage, biomolecular classification). Variables with p < 0.05 in the univariate analysis were considered statistically significant.

3. Results

Between January 2022 and June 2023, 451 invasive BC patients underwent breast surgery in our department. A total of 291 BC patients were excluded from the study for the following reasons: 153 received neoadjuvant chemotherapy, 101 underwent mastectomy, 18 underwent upfront ALND, and 19 did not undergo any axillary surgical exploration after multidisciplinary assessment due to age or low-performance status. Therefore, 160 patients were included in the study, as shown in Figure 1. The median age of the population was 61 (50;74) years. Among these, 26 patients (16.25%) had axillary macrometastatic disease identified during pathological assessment (LN+), while 134 patients (83.75%) were included in the LN− group.
The univariate analysis comparing LN+ and LN− groups is reported in Table 1. Preoperative data such as age (p = 0.880), BMI (p = 0.427), localization (quadrant) (p = 0.771), laterality (right vs. left) (p = 0.433), and BI-RADS breast density (p = 0.237) did not show a statistically significant association with LN status. Additionally, dividing the population by age, over 70 showed no statistically significant result (p = 0.768). Multifocality/multicentricity was associated with a higher rate of SLN involvement (20.90% vs. 42.31%; p = 0.020). Higher clinical stages were also associated with SLN involvement (p = 0.016). However, the histological assessment revealed that the preoperative tumor histology classification or tumor grade did not demonstrate a statistically significant difference between groups, with similar distribution even across rare variants (p = 0.103; p = 0.296, respectively).
In terms of tumor biology, a significantly higher rate of ER expression was observed in the LN+ group compared to the LN− group (90% vs. 74%; p < 0.001). Similar results were reported for PR expression (90% vs. 70%; p = 0.001). Conversely, Ki67, whether calculated as a continuous variable or as a dichotomous variable with cut-offs of 7.5% and 14%, did not show a statistically significant difference between the two groups (p = 0.756; p = 0.287; p = 0.687, respectively). Her2 expression patterns also showed no significant difference between the two groups (p = 0.865). Finally, both groups were analyzed according to the 2013 St. Gallen biomolecular classification. A higher rate of luminal B tumors was documented in the LN+ group compared to the LN− group, with fewer non-luminal tumors (3.85% vs. 13.44%) and luminal A tumors (30.77% vs. 55.22%) (p = 0.011).
Operating room occupancy analysis showed a median time per surgical procedure of 92.64 (58.39–141.34) minutes. Among the study population, 27 patients (16.86%) underwent intraoperative histological examination. In 3 out of these 27 patients (11.11%), the frozen section predicted LN involvement, and ALND was performed. The SLNB frozen section evaluation exhibited a 75% sensitivity and a 93% specificity (Table 2). The median axillary procedure time was 23.69 (16.87–27.57) minutes, while for conservative surgery without axillary surgery, the median duration was 70.60 (35.66–117.50) minutes.
Finally, our sample was stratified based on the SOUND study criteria to evaluate the number of SLNB procedures that could have been avoided. A sub-analysis was conducted on the population with lesions < 2 cm in preoperative assessment. Among them, a total of 117 (73.6%) patients were deemed eligible for the SOUND study. Within this subgroup, 11 (9.48%) BC patients showed an axillary positivity. The potential operating room time spared was 2696.81 min, which could have accommodated an additional 38.20 conservative surgery procedures without SLNB.

4. Discussion

In the last 30 years, the pursuit of safe axillary surgical de-escalation has been a pivotal research focus in modern breast surgery, aiming to improve the quality of life for patients with EBC. In this regard, the SOUND study has demonstrated for the first time that a complete and safe de-escalation is possible in a specific patient subgroup [9]. However, concerns have been raised by several authors regarding the lack of criteria related to age, menopausal status, or tumor biology for enrollment, which may limit its broad applicability [10]. Our real-world retrospective analysis demonstrated how factors such as ER and PR expression, and molecular subtype could be implemented in the de-escalation decision-making process.
Traditionally, the determination of pathologic nodal status obtained through surgical exploration has been the main factor driving recommendations for adjuvant systemic therapy and radiation therapy. Without a deeper understanding of the biological pathways underlying BC progression, pathological nodal status was considered the strongest predictor of distant disease [15]. In this context, the SLNB framework in the 1990s represented the first successful attempt toward axillary de-escalation. It effectively discriminated between SLNB node-negative and node-positive patients, with the latter eventually undergoing ALND [16]. However, further evidence from the ACOSOG Z0011 and AMAROS trials demonstrated that even in cases of a low nodal disease burden, completion of ALND could be avoided in the context of multidisciplinary treatment [3,17]. In recent years, as the therapeutic significance of axillary surgery has diminished, molecular genomic assessment has become a standard of care in the breast cancer decision-making process. This approach can determine the benefit of chemotherapy even in node-positive postmenopausal women [18,19,20,21]. Moreover, in addition to genomic assessment, innovative adjuvant treatments, and advanced breast imaging strategies, the concerns have been shifted from oncological safety to identifying which patient subsets would benefit the most from axillary surgical de-escalation [22,23,24,25].
Currently, supported by the growing body of evidence supporting de-escalating protocols, only a few routine indications are maintained for ALND. However, while safe, a large number of clinical strategies still partially rely on ALN status [26]. For instance, according to the RxPONDER trial, premenopausal luminal patients with one to three positive SLNs should undergo chemotherapy regardless of their oncotype DX recurrence score [21]. Moreover, findings of the monarchE trial restricted the use of innovative treatments such as abemaciclib in patients with ≥4 positive ALNs or 1–3 positive ALNs plus other high-risk features [27].
Combining these results with the increasing evidence supporting surgical de-escalation in different clinical settings, a mere dimensional criterion outlined in the SOUND study may not include some patients who could potentially benefit from a non-invasive axillary assessment [26]. Table 3 provides similar studies published in the last 5 years.
Tumor size has been classically associated with ALN involvement in BC patients. Historical series demonstrate how tumor diameter represents one of the strongest factors associated with SLN metastasis [51,52]. As stated before, tumor dimension was considered as the first inclusion criterion for the SOUND study [9]. Our experiments corroborate previous results in which tumor dimension has been highly associated with SLN metastasis. Moreover, similar results were obtained in pN1(sn) between our series and the SLNB group from the SOUND study (9.58% vs. 8.6%) [9]. As expected, the higher rate observed in our series may be justified by multicentric/multifocal tumors that were included in our analysis but then excluded from the SOUND study. Our series confirmed the association between multicentricity/multifocality and an increased rate of SLNB metastasis.
Additionally, our preliminary analysis demonstrated how SLNB may be predicted from well-known preoperative factors. Besides the SOUND results, age has been classically investigated in order to select a population for a safe de-escalation. Unlike the findings of the study conducted by Abdulla et al., our data did not demonstrate a difference in age distribution between groups [33]. Robust evidence demonstrated how older age (>70 years) may affect changes in hormonal receptor status and HER2 status. Moreover, older BC populations tend to present with smaller tumors with a low ki67 index, which could eventually affect the SLNB rate [33,53]. In line with our results, we believe that age should not be considered a discriminating factor a priori to avoid surgical axillary staging. Instead, it should be integrated into a multidisciplinary approach to assess whether axillary staging could influence adjuvant treatment and patients’ outcomes [54].
Besides tumor dimension and age group, modern multidisciplinary treatment encompasses tumor biological characteristics in order to tailor a multidisciplinary treatment for each patient [55]. In our analysis, we demonstrated how biological characteristics may predict SLNB results. Specifically, in the LN+ group a higher rate of ER, PR expression, high nuclear grade, and luminal B neoplasms was observed. PR and ER are key prognostic biomarkers, defining hormone-positive breast cancer and its response toward systemic hormonal therapy and/or innovative target therapy as cyclin-dependent kinase 4/6 inhibitors [56,57,58]. In clinical practice, PR-positive breast cancers show a higher endocrine response than PR-negative ones. While the predictive efficacy of endocrine therapy response is limited, PR remains a valuable indicator. Its role in regulating genes in a cell cycle-dependent manner suggests that some key PR target genes, especially those in the S phase, might have been missed in studies using unsynchronized cell lines [56,57,58]. Moreover, ER and PR status are routinely incorporated into the classification of intrinsic breast cancer subtypes [14].
Intrinsic BC subtypes are used to drive multidisciplinary treatment in association with performance status and clinical stage. Besides their clinical application in multidisciplinary treatment, some authors have started to explore their clinical application in SLNB status prediction in order to reduce the potential harm of disease underestimation or overtreatment [59]. As is widely known, due to the novel neoadjuvant strategies and preferential hematogenous spread of triple negative and HER2 type BC, LN involvement rates were lower compared to luminal tumors. Therefore, it supports potential biological-driven safe axillary de-escalation strategies in clinical settings different from those of the SOUND study [59,60,61,62,63]. As expected, luminal B tumors in our series were associated with higher rates of SLN metastasis, which shares with luminal A its indolent counterpart, a specific lymphotropic spread.
We are aware that our study may have some limitations. Due to its exploratory nature, no power analysis and multivariate analysis were performed. Moreover, retrospective analysis may have altered our results. Despite this potential limitation, the monocentric design provides a greater homogeneity among populations. Despite the mentioned limitations, this preliminary analysis has allowed us to identify factors associated with SLNB involvement such as tumor size ultrasound and biomolecular characteristics such as grade, luminal subtype, and ER/PR expression. Further, larger studies are needed to assess the role of each single risk factor to predict SLNB involvement in a multidisciplinary setting and to promote a rationalized approach useful to provide a safe de-escalating approach in different settings where clinicopathological factors, biological behavior, and genomic assessment may reduce the impact on patient quality of life and healthcare costs [64,65,66]. In our series, 73.6% of BC patients were eligible for the SOUND study, allowing a further 38.20 conservative surgery procedures to be performed without SLNB. In this clinical setting, without a clear benefit for the patients, the lack of implementation of de-escalation protocols could lead to multiple adverse effects such as detrimental effects on quality of life, increased surgical complications, escalating healthcare costs, and even a rise in healthcare-related environmental pollution [65,66,67,68].
In conclusion, our study demonstrated that applying the results of the SOUND study can effectively rationalize healthcare activities, potentially improving patient quality of life and reducing the detrimental effects of invasive ALN staging. While preliminary results from a large database demonstrated how short-term outcomes may be affected by invasive ALN staging, a comparative study to assess patients’ reported outcome measures (PROM) or quality of life is lacking, and further studies are needed to explore PROM in axillary EBC staging.
Regardless of these limitations, in the future, we believe that risk stratification of the population, beyond mere size criteria, through known preoperative prognostic and predictive factors, could further refine the selection of patients who could benefit from de-escalating strategies.

Author Contributions

Conceptualization, M.M. and F.P.; methodology, G.V. and F.T.; formal analysis, M.M., F.P. and G.V.; investigation, B.L. and V.C.; data curation, F.P. and M.P.; writing—original draft preparation, M.M.; writing—review and editing, G.V.; visualization, G.D.M. and E.T.; supervision, M.B. and O.C.B. project administration, M.M. and G.V. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by Area Territoriale Lazio 2 (n° 122/23).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors upon request.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Veronesi, U.; Viale, G.; Paganelli, G.; Zurrida, S.; Luini, A.; Galimberti, V.; Veronesi, P.; Intra, M.; Maisonneuve, P.; Zucca, F.; et al. Sentinel Lymph Node Biopsy in Breast Cancer: Ten-Year Results: Of a Randomized Controlled Study. Ann. Surg. 2010, 251, 595–600. [Google Scholar] [CrossRef] [PubMed]
  2. Krag, D.N.; Anderson, S.J.; Julian, T.B.; Brown, A.M.; Harlow, S.P.; Ashikaga, T.; Weaver, D.L.; Miller, B.J.; Jalovec, L.M.; Frazier, T.G.; et al. Technical Outcomes of Sentinel-Lymph-Node Resection and Conventional Axillary-Lymph-Node Dissection in Patients with Clinically Node-Negative Breast Cancer: Results from the NSABP B-32 Randomised Phase III Trial. Lancet Oncol. 2007, 8, 881–888. [Google Scholar] [CrossRef] [PubMed]
  3. Giuliano, A.E.; Ballman, K.V.; McCall, L.; Beitsch, P.D.; Brennan, M.B.; Kelemen, P.R.; Ollila, D.W.; Hansen, N.M.; Whitworth, P.W.; Blumencranz, P.W.; et al. Effect of Axillary Dissection vs No Axillary Dissection on 10-Year Overall Survival among Women with Invasive Breast Cancer and Sentinel Node Metastasis: The ACOSOG Z0011 (Alliance) Randomized Clinical Trial. JAMA—J. Am. Med. Assoc. 2017, 318, 918–926. [Google Scholar] [CrossRef] [PubMed]
  4. Tinterri, C.; Gentile, D.; Gatzemeier, W.; Sagona, A.; Barbieri, E.; Testori, A.; Errico, V.; Bottini, A.; Marrazzo, E.; Dani, C.; et al. Preservation of Axillary Lymph Nodes Compared with Complete Dissection in T1-2 Breast Cancer Patients Presenting One or Two Metastatic Sentinel Lymph Nodes: The SINODAR-ONE Multicenter Randomized Clinical Trial. Ann. Surg. Oncol. 2022, 29, 5732–5744. [Google Scholar] [CrossRef]
  5. Galimberti, V.; Cole, B.F.; Viale, G.; Veronesi, P.; Vicini, E.; Intra, M.; Mazzarol, G.; Massarut, S.; Zgajnar, J.; Taffurelli, M.; et al. Axillary Dissection versus No Axillary Dissection in Patients with Breast Cancer and Sentinel-Node Micrometastases (IBCSG 23-01): 10-Year Follow-up of a Randomised, Controlled Phase 3 Trial. Lancet Oncol. 2018, 19, 1385–1393. [Google Scholar] [CrossRef]
  6. Brackstone, M.; Baldassarre, F.G.; Perera, F.E.; Cil, T.; Chavez Mac Gregor, M.; Dayes, I.S.; Engel, J.; Horton, J.K.; King, T.A.; Kornecki, A.; et al. Management of the Axilla in Early-Stage Breast Cancer: Ontario Health (Cancer Care Ontario) and ASCO Guideline. J. Clin. Oncol. 2021, 39, 3056–3082. [Google Scholar] [CrossRef] [PubMed]
  7. Rashmi Kumar, N.; Schonfeld, R.; Gradishar, W.J.; Lurie, R.H.; Moran, M.S.; Abraham, J.; Abramson, V.; Aft, R.; Agnese, D.; Allison, K.H.; et al. NCCN Guidelines Version 1.2024 Breast Cancer; NCCN: Plymouth Meeting, PA, USA, 2024. [Google Scholar]
  8. Gentilini, O.; Veronesi, U. Abandoning Sentinel Lymph Node Biopsy in Early Breast Cancer? A New Trial in Progress at the European Institute of Oncology of Milan (SOUND: Sentinel Node vs Observation after Axillary UltraSouND). Breast 2012, 21, 678–681. [Google Scholar] [CrossRef]
  9. Gentilini, O.D.; Botteri, E.; Sangalli, C.; Galimberti, V.; Porpiglia, M.; Agresti, R.; Luini, A.; Viale, G.; Cassano, E.; Peradze, N.; et al. Sentinel Lymph Node Biopsy vs No Axillary Surgery in Patients With Small Breast Cancer and Negative Results on Ultrasonography of Axillary Lymph Nodes: The SOUND Randomized Clinical Trial. JAMA Oncol. 2023, 9, 1557–1564. [Google Scholar] [CrossRef] [PubMed]
  10. Hersh, E.H.; King, T.A. De-Escalating Axillary Surgery in Early-Stage Breast Cancer. Breast 2022, 62, S43–S49. [Google Scholar] [CrossRef]
  11. Schwartz, T. At the Speed of SOUND: The Pace of Change for Axillary Management in Breast Cancer. Ann. Surg. Oncol. 2024, 31, 2801–2803. [Google Scholar] [CrossRef]
  12. Amin, M.B. AJCC Cancer Staging System, 8th ed.; American Joint Committee on Cancer: Chicago, IL, USA, 2017. [Google Scholar]
  13. Wolff, A.C.; McShane, L.M.; Hammond, M.E.H.; Allison, K.H.; Fitzgibbons, P.; Press, M.F.; Harvey, B.E.; Mangu, P.B.; Bartlett, J.M.S.; 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. Arch. Pathol. Lab. Med. 2018, 142, 1364–1382. [Google Scholar] [CrossRef] [PubMed]
  14. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.J.; Albain, K.S.; André, F.; Bergh, J.; et al. Personalizing the Treatment of Women with Early Breast Cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef] [PubMed]
  15. Buonomo, O.C.; Caredda, E.; Portarena, I.; Vanni, G.; Orlandi, A.; Bagni, C.; Petrella, G.; Palombi, L.; Orsaria, P. New Insights into the Metastatic Behavior after Breast Cancer Surgery, According to Well-Established Clinicopathological Variables and Molecular Subtypes. PLoS ONE 2017, 12, e0184680. [Google Scholar] [CrossRef] [PubMed]
  16. Veronesi, U.; Paganelli, G.; Viale, G.; Galimberti, V.; Luini, A.; Zurrida, S.; Robertson, C.; Sacchini, V.; Veronesi, P.; Orvieto, E.; et al. Sentinel Lymph Node Biopsy and Axillary Dissection in Breast Cancer: Results in a Large Series. J. Natl. Cancer Inst. 1999, 91, 368–373. [Google Scholar] [CrossRef] [PubMed]
  17. Donker, M.; van Tienhoven, G.; Straver, M.E.; Meijnen, P.; van de Velde, C.J.H.; Mansel, R.E.; Cataliotti, L.; Westenberg, A.H.; Klinkenbijl, J.H.G.; Orzalesi, L.; et al. Radiotherapy or Surgery of the Axilla after a Positive Sentinel Node in Breast Cancer (EORTC 10981-22023 AMAROS): A Randomised, Multicentre, Open-Label, Phase 3 Non-Inferiority Trial. Lancet Oncol. 2014, 15, 1303–1310. [Google Scholar] [CrossRef] [PubMed]
  18. Sparano, J.A.; Paik, S. Development of the 21-Gene Assay and Its Application in Clinical Practice and Clinical Trials. J. Clin. Oncol. 2008, 26, 721–728. [Google Scholar] [CrossRef] [PubMed]
  19. Sparano, J.A.; Gray, R.J.; Makower, D.F.; Pritchard, K.I.; Albain, K.S.; Hayes, D.F.; Geyer, C.E.; Dees, E.C.; Goetz, M.P.; Olson, J.A.; et al. Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N. Engl. J. Med. 2018, 379, 111–121. [Google Scholar] [CrossRef] [PubMed]
  20. Lee, M.K.; Montagna, G.; Pilewskie, M.L.; Sevilimedu, V.; Morrow, M. Axillary Staging Is Not Justified in Postmenopausal Clinically Node-Negative Women Based on Nodal Disease Burden. Ann. Surg. Oncol. 2023, 30, 92–97. [Google Scholar] [CrossRef] [PubMed]
  21. Kalinsky, K.; Barlow, W.E.; Gralow, J.R.; Meric-Bernstam, F.; Albain, K.S.; Hayes, D.F.; Lin, N.U.; Perez, E.A.; Goldstein, L.J.; Chia, S.K.L.; et al. 21-Gene Assay to Inform Chemotherapy Benefit in Node-Positive Breast Cancer. N. Engl. J. Med. 2021, 385, 2336–2347. [Google Scholar] [CrossRef]
  22. Chopra, S.; Khosla, M.; Vidya, R. Innovations and Challenges in Breast Cancer Care: A Review. Medicina 2023, 59, 957. [Google Scholar] [CrossRef]
  23. Vanni, G.; Materazzo, M.; Di Lorenzo, N.; Tacconi, F.; Pellicciaro, M.; Berretta, M.; Di Mauro, G.; Pistolese, C.A.; Noce, A.; Longo, B.; et al. Contrast-Enhanced Ultrasound Using Intradermal Microbubble Sulfur Hexafluoride in Non-Invasive Axillary Staging in Breast Cancer: Are We Missing a Chance? Anticancer Res. 2024, 44, 2021–2030. [Google Scholar] [CrossRef] [PubMed]
  24. Vanni, G.; Tazzioli, G.; Pellicciaro, M.; Materazzo, M.; Paolo, O.; Cattadori, F.; Combi, F.; Papi, S.; Pistolese, C.A.; Cotesta, M.; et al. Delay in Breast Cancer Treatments During the First COVID-19 Lockdown. A Multicentric Analysis of 432 Patients. Anticancer Res. 2020, 40, 7119–7125. [Google Scholar] [CrossRef] [PubMed]
  25. Buonomo, O.C.; Materazzo, M.; Pellicciaro, M.; Iafrate, G.; Ielpo, B.; Rizza, S.; Pistolese, C.A.; Perretta, T.; Meucci, R.; Longo, B.; et al. Contrast-Enhanced Ultrasound Using Intradermal Microbubble Sulfur Hexafluoride for Identification of Sentinel Lymph Nodes During Breast Cancer Surgery: A Clinical Trial. Anticancer Res. 2023, 43, 557–567. [Google Scholar] [CrossRef]
  26. Tinterri, C.; Canavese, G.; Gentile, D. To Dissect or Not to Dissect? The Surgeon’s Perspective on the Prediction of Greater Than or Equal to 4 Axillary Lymph Node Metastasis in Early-Stage Breast Cancer: A Comparative Analysis of the Per-Protocol Population of the SINODAR-ONE Clinical Trial. Ann. Surg. Open 2024, 5, e405. [Google Scholar] [CrossRef]
  27. Johnston, S.R.D.; Harbeck, N.; Hegg, R.; Toi, M.; Martin, M.; Shao, Z.M.; Zhang, Q.Y.; Martinez Rodriguez, J.L.; Campone, M.; Hamilton, E.; et al. Abemaciclib Combined With Endocrine Therapy for the Adjuvant Treatment of HR+, HER2-, Node-Positive, High-Risk, Early Breast Cancer (MonarchE). J. Clin. Oncol. 2020, 38, 3987–3998. [Google Scholar] [CrossRef]
  28. Akbari, M.E.; Akbari, A.; Ebrahimian, S. Pathological Characteristics Predicting Sentinel Lymph Node Metastasis in Early Breast Cancer Patients. Casp. J. Intern. Med. 2024, 15, 472–477. [Google Scholar] [CrossRef]
  29. Lee, S.Y.; Yoo, T.K.; Kim, J.; Chung, I.Y.; Ko, B.S.; Kim, H.J.; Lee, J.W.; Son, B.H.; Lee, S.B. Characteristics and Risk Factors of Axillary Lymph Node Metastasis of Microinvasive Breast Cancer. Breast Cancer Res. Treat. 2024, 206, 495–507. [Google Scholar] [CrossRef]
  30. Liu, R.; Chen, J.; Cao, W.; Li, T.; Liao, Y.; Li, Y. Risk Factors and Prognosis of Sentinel Lymph Node Metastasis in Breast-Conserving Breast Cancer: A Retrospective Study Based on the SEER Database. Medicine 2024, 103, E37263. [Google Scholar] [CrossRef]
  31. Pang, W.; Wang, Y.; Zhu, Y.; Jia, Y.; Nie, F. Predictive Value for Axillary Lymph Node Metastases in Early Breast Cancer: Based on Contrast-Enhanced Ultrasound Characteristics of the Primary Lesion and Sentinel Lymph Node. Clin. Hemorheol. Microcirc. 2024, 86, 357–367. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, Z.; Jiang, Q.; Wang, J.; Yang, X. A Nomogram Model for Predicting the Risk of Axillary Lymph Node Metastasis in Patients with Early Breast Cancer and CN0 Status. Oncol. Lett. 2024, 28, 345. [Google Scholar] [CrossRef]
  33. Abdulla, H.A.; Salman, A.Z.; Alaraibi, S.J.; Nazzal, K.; Ahmed, S.A.; Almahari, S.A.; Dhaif, A. Risk Factors Associated With Sentinel Lymph Node Metastasis in Clinically Node-Negative Breast Cancer. Eur. J. Breast Health 2023, 19, 229–234. [Google Scholar] [CrossRef] [PubMed]
  34. Jin, J.; Liu, P.F.; Ye, J.D.; Wu, Y.Y. Analysis of Risk Factors of Axillary Lymph-Node Metastasis in Triple-Negative Breast Cancer. Asian J. Surg. 2023, 46, 2265–2267. [Google Scholar] [CrossRef] [PubMed]
  35. Fu, F.; Zhang, Y.; Sun, J.; Zhang, C.; Zhang, D.; Xie, L.; Chu, F.; Yu, X.; Xie, Y. Predictors of Sentinel Lymph Node Metastasis in Chinese Women with Clinical T1-T2 N0 Breast Cancer and a Normal Axillary Ultrasound. Acta Radiol. 2022, 63, 1463–1468. [Google Scholar] [CrossRef] [PubMed]
  36. Gao, X.; Luo, W.; He, L.; Yang, L. Nomogram Models for Stratified Prediction of Axillary Lymph Node Metastasis in Breast Cancer Patients (CN0). Front. Endocrinol. 2022, 13, 967062. [Google Scholar] [CrossRef] [PubMed]
  37. Wu, S.L.; Da Gai, J.; Yu, X.M.; Mao, X.; Jin, F. A Novel Nomogram and Risk Classification System for Predicting Lymph Node Metastasis of Breast Mucinous Carcinoma: A SEER-Based Study. Cancer Med. 2022, 11, 4767–4783. [Google Scholar] [CrossRef] [PubMed]
  38. Xiong, J.; Zuo, W.; Wu, Y.; Wang, X.; Li, W.; Wang, Q.; Zhou, H.; Xie, M.; Qin, X. Ultrasonography and Clinicopathological Features of Breast Cancer in Predicting Axillary Lymph Node Metastases. BMC Cancer 2022, 22, 1155. [Google Scholar] [CrossRef] [PubMed]
  39. Yiming, A.; Wubulikasimu, M.; Yusuying, N. Analysis on Factors behind Sentinel Lymph Node Metastasis in Breast Cancer by Color Ultrasonography, Molybdenum Target, and Pathological Detection. World J. Surg. Oncol. 2022, 20, 72. [Google Scholar] [CrossRef] [PubMed]
  40. Zhu, Y.; Lv, W.; Wu, H.; Yang, D.; Nie, F. A Preoperative Nomogram for Predicting the Risk of Sentinel Lymph Node Metastasis in Patients with T1-2N0 Breast Cancer. Jpn. J. Radiol. 2022, 40, 595–606. [Google Scholar] [CrossRef] [PubMed]
  41. Hu, X.; Xue, J.; Peng, S.; Yang, P.; Yang, Z.; Yang, L.; Dong, Y.; Yuan, L.; Wang, T.; Bao, G. Preoperative Nomogram for Predicting Sentinel Lymph Node Metastasis Risk in Breast Cancer: A Potential Application on Omitting Sentinel Lymph Node Biopsy. Front. Oncol. 2021, 11, 665240. [Google Scholar] [CrossRef]
  42. Minami, S.; Sakimura, C.; Irie, J.; Tokai, Y.; Okubo, H.; Ohno, T. Predictive Factors Among Clinicopathological Characteristics for Sentinel Lymph Node Metastasis in T1-T2 Breast Cancer. Cancer Manag. Res. 2021, 13, 215–223. [Google Scholar] [CrossRef]
  43. Wang, L.; Li, J.; Qiao, J.; Guo, X.; Bian, X.; Guo, L.; Liu, Z.; Lu, Z. Establishment of a Model for Predicting Sentinel Lymph Node Metastasis in Early Breast Cancer Based on Contrast-Enhanced Ultrasound and Clinicopathological Features. Gland Surg. 2021, 10, 1701–1712. [Google Scholar] [CrossRef] [PubMed]
  44. Yang, L.; Wang, H.; Ma, J.; Hao, J.; Zhang, C.; Ma, Q.; Wang, B. Association between the Platelet to Lymphocyte Ratio, Neutrophil to Lymphocyte Ratio and Axillary Lymph Node Metastasis in CT1N0 Breast Cancer Patients. Am. J. Transl. Res. 2021, 13, 1854–1861. [Google Scholar]
  45. Catteau, X.; Koopmansch, C.; Maris, C.; Colart, P.; Noël, J.C. Predictive Factors of Lymph Node Metastasis and Effectiveness of Intraoperative Examination of Sentinel Lymph Node in Breast Carcinoma: A Retrospective Belgian Study. Ann. Diagn. Pathol. 2020, 49, 151607. [Google Scholar] [CrossRef]
  46. Fan, Y.; Chen, X.; Li, H. Clinical Value of Serum Biomarkers CA153, CEA, and White Blood Cells in Predicting Sentinel Lymph Node Metastasis of Breast Cancer. Int. J. Clin. Exp. Pathol. 2020, 13, 2889–2894. [Google Scholar]
  47. Fan, B.; Pardo, J.A.; Serres, S.; Alapati, A.C.; Szewczyk, J.; Mele, A.; James, T.A. Role of Sentinel Lymph Node Biopsy in Microinvasive Breast Cancer. Ann. Surg. Oncol. 2020, 27, 4468–4473. [Google Scholar] [CrossRef] [PubMed]
  48. He, Z.; Lan, X.; Tan, Y.; Lin, X.; Wen, G.; Wang, X.; Huang, X.; Yang, F. Identification of Risk Factors Associated with Axillary Lymph Node Metastasis for Sentinel Lymph Node-Positive Breast Cancer Patients. J. Oncol. 2020, 2020, 8884337. [Google Scholar] [CrossRef]
  49. Takada, K.; Kashiwagi, S.; Asano, Y.; Goto, W.; Kouhashi, R.; Yabumoto, A.; Morisaki, T.; Shibutani, M.; Takashima, T.; Fujita, H.; et al. Prediction of Lymph Node Metastasis by Tumor-Infiltrating Lymphocytes in T1 Breast Cancer. BMC Cancer 2020, 20, 598. [Google Scholar] [CrossRef] [PubMed]
  50. Zhang, Y.; Li, J.; Fan, Y.; Li, X.; Qiu, J.; Zhu, M.; Li, H. Risk Factors for Axillary Lymph Node Metastases in Clinical Stage T1-2N0M0 Breast Cancer Patients. Medicine 2019, 98, e17481. [Google Scholar] [CrossRef]
  51. Rivadeneira, D.E.; Simmons, R.M.; Christos, P.J.; Hanna, K.; Daly, J.M.; Osborne, M.P. Predictive Factors Associated with Axillary Lymph Node Metastases in T1a and T1b Breast Carcinomas: Analysis in More than 900 Patients. J. Am. Coll. Surg. 2000, 191, 1–6. [Google Scholar] [CrossRef]
  52. Aitken, E.; Osman, M. Factors Affecting Nodal Status in Invasive Breast Cancer: A Retrospective Analysis of 623 Patients. Breast J. 2010, 16, 271–278. [Google Scholar] [CrossRef]
  53. Vanni, G.; Materazzo, M.; Pellicciaro, M.; Morando, L.; Portarena, I.; Anemona, L.; D’Angelillo, R.M.; Barbarino, R.; Chiaravalloti, A.; Meucci, R.; et al. Does Age Matter? Estimating Risks of Locoregional Recurrence After Breast-Conservative Surgery. In Vivo 2020, 34, 1125–1132. [Google Scholar] [CrossRef] [PubMed]
  54. Shah, H.; Surujballi, J.; Awan, A.A.; Hutton, B.; Arnaout, A.; Shorr, R.; Vandermeer, L.; Alzahrani, M.J.; Clemons, M. A Scoping Review Characterizing “Choosing Wisely®” Recommendations for Breast Cancer Management. Breast Cancer Res. Treat. 2021, 185, 533–547. [Google Scholar] [CrossRef] [PubMed]
  55. Dimitrov, G.; Atanasova, M.; Popova, Y.; Vasileva, K.; Milusheva, Y.; Troianova, P. Molecular and Genetic Subtyping of Breast Cancer: The Era of Precision Oncology. World Cancer Res. J. 2022, 9, e2367. [Google Scholar] [CrossRef]
  56. Dressing, G.E.; Lange, C.A. Integrated Actions of Progesterone Receptor and Cell Cycle Machinery Regulate Breast Cancer Cell Proliferation. Steroids 2009, 74, 573–576. [Google Scholar] [CrossRef] [PubMed]
  57. Li, Z.; Wei, H.; Li, S.; Wu, P.; Mao, X. The Role of Progesterone Receptors in Breast Cancer. Drug Des. Dev. Ther. 2022, 16, 305–314. [Google Scholar] [CrossRef] [PubMed]
  58. Reinius, M.A.V.; Smyth, E. Anti-Cancer Therapy with Cyclin-Dependent Kinase Inhibitors: Impact and Challenges. Expert Rev. Mol. Med. 2021, 23, e6. [Google Scholar] [CrossRef]
  59. Kędzierawski, P.; Bocian, A.; Radowicz-Chil, A.; Huruk-Kuchinka, A.; Mężyk, R. Subtype of Breast Cancer Influences Sentinel Lymph Node Positivity. Arch. Med. Sci. 2023, 19, 618–625. [Google Scholar] [CrossRef] [PubMed]
  60. Gangi, A.; Mirocha, J.; Leong, T.; Giuliano, A.E. Triple-Negative Breast Cancer Is Not Associated with Increased Likelihood of Nodal Metastases. Ann. Surg. Oncol. 2014, 21, 4098–4103. [Google Scholar] [CrossRef] [PubMed]
  61. Pilewskie, M.; Morrow, M. Axillary Nodal Management Following Neoadjuvant Chemotherapy. JAMA Oncol. 2017, 3, 549–555. [Google Scholar] [CrossRef]
  62. Vici, P.; Pizzuti, L.; Natoli, C.; Gamucci, T.; Di Lauro, L.; Barba, M.; Sergi, D.; Botti, C.; Michelotti, A.; Moscetti, L.; et al. Triple Positive Breast Cancer: A Distinct Subtype? Cancer Treat. Rev. 2015, 41, 69–76. [Google Scholar] [CrossRef]
  63. Baker, J.L.; Muhsen, S.; Zabor, E.C.; Stempel, M.; Gemignani, M.L. Does Lymph Node Status Prior to Neoadjuvant Chemotherapy Influence the Number of Sentinel Nodes Removed? Ann. Surg. Oncol. 2019, 26, 336–342. [Google Scholar] [CrossRef]
  64. Gentilini, O.; Botteri, E.; Dadda, P.; Sangalli, C.; Boccardo, C.; Peradze, N.; Ghisini, R.; Galimberti, V.; Veronesi, P.; Luini, A.; et al. Physical Function of the Upper Limb after Breast Cancer Surgery. Results from the SOUND (Sentinel Node vs. Observation after Axillary Ultra-SouND) Trial. Eur. J. Surg. Oncol. 2016, 42, 685–689. [Google Scholar] [CrossRef] [PubMed]
  65. Shubeck, S.P.; Morrow, M.; Dossett, L.A. De-Escalation in Breast Cancer Surgery. NPJ Breast Cancer 2022, 8, 25. [Google Scholar] [CrossRef] [PubMed]
  66. Vanni, G.; Materazzo, M.; Pellicciaro, M.; Tacconi, F.; Faccini, B.; Longo, B.; Cervelli, V.; Buonomo, O. Breast Cancer Treatment De-Escalation: Breaking the SOUND Barrier. World Cancer Res. J. 2023, 10, e2682. [Google Scholar] [CrossRef]
  67. Materazzo, M.; Facchini, A.; Garozzo, D.; Buonomo, C.; Pellicciaro, M.; Vanni, G. Maintaining Good Practice in Breast Cancer Management and Reducing the Carbon Footprint of Care: Study Protocol and Preliminary Results. World Cancer Res. J. 2022, 9, e2438. [Google Scholar] [CrossRef]
  68. Vanni, G.; Materazzo, M.; Portarena, I.; Pellicciaro, M.; Meacci, A.; Pizzimenti, A.R.; Buonomo, O.C. Socioeconomic Impact of OncotypeDX on Breast Cancer Treatment: Preliminary Results. In Vivo 2023, 37, 2510–2516. [Google Scholar] [CrossRef]
Figure 1. Study flowchart. BC: breast cancer; ALND: axillary lymph node dissection; SLNB: sentinel lymph node biopsy.
Figure 1. Study flowchart. BC: breast cancer; ALND: axillary lymph node dissection; SLNB: sentinel lymph node biopsy.
Curroncol 31 00309 g001
Table 1. Demographic and preoperative data.
Table 1. Demographic and preoperative data.
LN− Group
(n = 134)
LN+ Group
(n = 26)
p-Value
Age (IQR) years60 (50; 71)67 (50; 74)0.880
Age > 70 years yes, n (%)36 (26.87%) 8 (30.77%)0.768
BMI (IQR) kg/m224.22 (22.06; 27.31)26,04 (23.38; 28.52)0.427
Localization (quadrant), n (%)
Upper-outer (UOQ)57 (42.55%) 11 (42.31%) 0.771
Upper-inner (UIQ)15 (11.19%) 4 (15.38%)
Lower-outer (LOQ)18 (13.43%) 4 (15.38%)
Lower-inner (LIQ)21 (15.67%) 5 (19.23%)
Other localization23 (17.16%)2 (7.69%)
Laterality (right vs. left), n (%)
Right69 (51.49%)11 (42.31%) 0.433
Left65 (48.51%)15 (57.69%)
Focality (unifocal vs. multifocal), n (%)
Unifocal106 (79.10%)15 (57.69%)0.103
Multifocal14 (10.45%)6 (23.08%)
Multicentric 14 (10.45%)5 (19.23%)
Unifocal106 (79.10%)15 (57.69%)0.020 *
Multifocal/multicentric28 (20.90%)11 (42.31%)
Clinical Stage, n (%)
cT1a9 (6.72%)1 (3.85%)0.014 *
cT1b44 (32.84%)5 (19.23%)
cT1c58 (43.28%)8 (30.77%)
cT223 (17.16%)12 (46.15%)
Breast Density, n (%)
A6 (4.48%)7 (26.92%)0.237
B99 (73.88%)14 (53.85%)
C23 (17.16%)4 (15.38%)
D6 (4.48%)1 (3.85%)
Tumor Diameter, mm (IQR)12 (9.0; 18.5)19 (13.0; 23.5)0.016 *
Tumor size > 20 mm yes, n (%) 23 (17.16%)12 (46.15%)0.001 *
Microcalcification yes, n (%)31 (23.13%)9 (34.62%)0.184
Tumor Histology, n (%)
Invasive ductal110 (82.09%)19 (73.08%)0.103
Invasive lobular17 (12.69%)4 (15.38%)
Other7 (5.22%)3 (11.54%)
Tumor Grade, n (%)
117 (12.69%)4 (15.38%)0.296
285 (63.43%)17 (65.39%)
330 (22.39%)2 (7.69%)
N/A2 (1.49%)3 (11.54%)
Immunohistochemistry
ER % (IQR)74 (72.5; 95)90 (80; 95)<0.001 *
PR % (IQR)70 (1.25; 90)90 (40; 95)0.001 *
Ki67 % (IQR)18 (10; 30)25 (16.75; 30)0.756
Ki67 > 7.5% yes, n (%)110 (82.09%)19 (73.08%)0.287
Ki67 > 14% yes, n (%)82 (61.19%)17 (65.38%)0.687
HER2 Score, n (%)
031 (23.14%)4 (15.38%)0.865
183 (61.94%)15 (57.69%)
27 (5.22%)3 (11.55%)
313 (9.70%)4 (15.38%)
Molecular Subtype, n (%)
Luminal A74 (55.22%)8 (30.77%)0.011 *
Luminal B42 (31.34%)17 (65.38%)
HER2 type4 (2.99%)1 (3.85%)
Triple negative14 (10.45%)0 (0%)
All continuous variables are expressed as median and interquartile range (IQR), while categorical variables are indicated as frequencies and percentages. Continuous variables between groups were compared with a Student t-test or Mann–Whitney U test, according to the Kolmogorov–Smirnov test. Categorical and dichotomous variables were compared between groups with a chi-square test or Fisher’s exact test according to the sample size. p-values < 0.05 are highlighted with * and considered statistically significant. BMI: body mass index; ER: estrogen receptor; LN: lymph node; PR: progesterone receptor.
Table 2. SLNB Frozen section performance according to the SLNB definitive staining.
Table 2. SLNB Frozen section performance according to the SLNB definitive staining.
SLNB Definitive Staining Macrometastasis
YesNoTotal
SLNB Frozen Section
Macrometastasis
Yes3 (11.54%)0 (%)3 (11.54%)
No1 (3.84%)22 (84.62%) 23 (88.46%)
Total4 (15.38%)22 (84.62%)26 (100%)
SLNB: sentinel lymph node biopsy.
Table 3. Trial investigating SLNB positive predictive value in EBC.
Table 3. Trial investigating SLNB positive predictive value in EBC.
Author (Year)CountryDesignClinical Stagen=ALN+
n=
Variables ExaminedUnivariate Preoperative Prognostic FactorMultivariate Preoperative Prognostic Factor
Akbari et al. [28]
(2024)
IranRetrospectivecT1-3 cN07333Clinical
Pathological
LVI
Histology
Ki67
N.A.
Lee et al. [29]
(2024)
South KoreaRetrospectivepT1(mi) cN0168870Clinical
Pathological
Age
Breast surgery
Axillary surgery
SLNs number
Tumor size
Grade
LVI
Foci of microinvasion number
ER
PR
Molecular subtype
Radiotherapy
Endocrine therapy
Age
Axillary surgery
SLNs number
LVI
Foci of microinvasion number
ER
PR
Liu et al. [30]
(2024)
ChinaSEER databaseBCS cT1-3 cN0169832338Clinical
Pathological
Age
Race
Tumor site
Tumor size grade
ER
PR
SLN number
Radiation
Chemotherapy
Age
Race
Grade
Radiation
ER
PR
Pang et al. [31] (2024)ChinaRetrospectivecT1-2 cN0118N.A.US
CEUS
Clinical
Pathological
Age
HER2
Nutrient vessel
BC CEUS enhancement pattern
SLN CEUS patterns

CEUS pattern of enhancement lesion
CEUS patterns of SLN
Zhang et al. [32] (2024)ChinaRetrospectivecT1-2 cN0998228US
Clinical
Pathological
LVI
Tumor location
ALN US
Tumor size
Histological grade
Lymphovascular invasion
ALN US
Maximum diameter
Abdulla et al. [33]
(2023)
BahrainRetrospectivecT1-3 cN016035Clinical
Pathological
Age
Grade
ER
LVI
Tumor size
Tumor grade
ER
LVI
Tumor size
Jin et al. [34] (2023)ChinaSEER research plus dataTNBC cT1-3 cN017554N.A.Clinical
Pathological
Age
Race
Histology
Grade
Tumor size
Marital status
Sex
Age
Race
Tumor size
Grade
Fu et al. [35]
(2022)
ChinaRetrospectivecT1-2 cN014126Clinical
Pathological
Tumor location
ER
PR
LVI
Tumor location
PR
LVI
Gao et al. [36]
(2022)
ChinaSEER databasecT1-3 cN0

ALN− vs. ALN+(1–2) vs. ALN+ (>2)
4753ALN+(1-2) = 1961)
ALN + (>2) = 371
Clinical
Pathological
ALN− vs. ALN+(1–2) vs. ALN+ (>2):

Age
Race
Tumor size
Tumor location
Molecular subtype
ER
PR
HER2
Grade
Histology
ALN− vs. ALN+:
Tumor size
Tumor location
Molecular subtype
Grade
Histology
Wu et al. [37]
(2022)
ChinaSEER databaseMBC cT1-3 cN066551Clinical
Pathological
Age
Tumor size
Grade
HER2
Age ≥ 70
Tumor size
Grade II/IV
Xiong et al. [38]
(2022)
ChinaRetrospectivecT1-2 cN01076437US
Clinical
Pathological
Age
Grade
Multifocality
Molecular subtype
Tumor location
Tumor size
US margin
Skin distance
Age
Grade
Molecular subtype
Tumor location
Tumor size
US margin
Skin distance
Yiming et al. [39]
(2022)
ChinaRetrospectivecT1-3 cN09949US
Clinical
Pathological
Age
Tumor size
CK5/6
HER2
TP53 mutation
BRCA1 mutation
BRCA2 mutation
N.A.
Zhu et al. [40]
(2022)
ChinaprospectivecT1-2 cN011459US
CEUS
Clinical
Pathological
Multifocal
HER2
Tumor size
Resistance index
CEUS extended range
Tumor diameter
HER2
Tumor size
Resistance index
Hu et al. [41]
(2021)
ChinaRetrospectivec T1-3 cN0.624147US
Clinical
Pathological
Age
BMI
Her2 type vs. LumA
Ki67
Tumor size
Inner echo
Calcification
Color Doppler flow
Aspect ratio
Age
BMI
Ki67
Tumor size
Tumor margin
Calcification
Aspect ratio
Minami et al. [42]
(2021)
JapanRetrospectivec T1-2 cN0.31354Clinical
Serum
Pathological
IGT
Tumor size
Nipple distance
Tumor location
Tumor stage
IGT
Tumor size
Nipple distance
Wang et al. [43]
(2021)
ChinaRetrospectivec T1-3 cN0.29774US
CEUS
MAM
Clinical
Pathological
BMI
Tumor resection biopsy
Tumor size
LVI
ER
PR
CK5/6
HER2
ALN US aspect ratio
US lymphatic structure
Cortical medulla US
SLN CEUS patterns
SLN CEUS aspect ratio
ALN MAM aspect ratio
BMI
SLN US aspect ratio
SLN CEUS patterns
SLN CEUS aspect ratio
ALN MAM aspect ratio
CK5/6
Yang et al. [44]
(2021)
ChinaRetrospectivecT1
cN0
15432Clinical
Serum
Pathological
Tumor size
LVI
PLR
NLR
N.A.
Catteau et al. [45]
(2020)
BelgiumRetrospectiveTis-T4212 Clinical
Pathological
Age
Tumor size
T stage
T grade
LVI
Molecular classification
Ki67 20%
N.A.
Fan et al. [46]
(2020)
ChinaRetrospectivecT1-2 cN012156Clinical
Serum
Pathological
CA153
CEA
WBC
Tumor size
ER
PR
CA153
CEA
WBC
Fan et al. [47] (2020)USANational cancer databasecT1mi cN0260976Clinical
Serum
Pathological
N.A.Grade
Age
Molecular subtype
He et al. [48]
(2020)
ChinaRetrospectivecT1-2 cN0556235Clinical
Pathological
SLN positive absolute number
SLN metastasis rate
LVI
SLN positive absolute number
SLN metastasis rate
LVI
Takada et al. [49]
(2020)
JapanRetrospectivecT1 cN033216Clinical
Pathological
Tumor size
LVI
TILS yes/no
Tumor Size
LVI
TILS yes/no
Zhang et al. [50]
(2019)
ChinaRetrospectivecT1-2 cN01671541US
Clinical
Pathological
Tumor size
Palpable
Nipple distance
Tumor location
Grade
ISAT
IIAT
Tumor size
Nipple distance
ISAT
IIAT
ALN: Axillary lymph nodes, BC: breast cancer; BCS: breast-conserving surgery; BMI: body mass index; BRCA: breast cancer gene; CA153: cancer antigen 15-3; CEA: carcino-embryonic antigen; CEUS: contrast enhancement ultrasound; CK5/6: cytokeratin 5/6; EBC: early breast cancer; ER: estrogen receptor; IGT: impaired glucose tolerance; ISAT: infiltration of subcutaneous adipose tissue; IIAT: infiltration of the interstitial adipose tissue; LVI: lymphovascular invasion; HER2: human epidermal growth factor receptor 2; MAM: mammogram; MBC: mucinous breast cancer; PR: progesterone receptor; SLN: sentinel lymph node; SLNB: sentinel lymph node biopsy; TNBC: triple negative breast cancer; TP53: tumor protein 53.
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

Vanni, G.; Materazzo, M.; Paduano, F.; Pellicciaro, M.; Di Mauro, G.; Toscano, E.; Tacconi, F.; Longo, B.; Cervelli, V.; Berretta, M.; et al. New Insight for Axillary De-Escalation in Breast Cancer Surgery: “SoFT Study” Retrospective Analysis. Curr. Oncol. 2024, 31, 4141-4157. https://doi.org/10.3390/curroncol31080309

AMA Style

Vanni G, Materazzo M, Paduano F, Pellicciaro M, Di Mauro G, Toscano E, Tacconi F, Longo B, Cervelli V, Berretta M, et al. New Insight for Axillary De-Escalation in Breast Cancer Surgery: “SoFT Study” Retrospective Analysis. Current Oncology. 2024; 31(8):4141-4157. https://doi.org/10.3390/curroncol31080309

Chicago/Turabian Style

Vanni, Gianluca, Marco Materazzo, Floriana Paduano, Marco Pellicciaro, Giordana Di Mauro, Enrica Toscano, Federico Tacconi, Benedetto Longo, Valerio Cervelli, Massimiliano Berretta, and et al. 2024. "New Insight for Axillary De-Escalation in Breast Cancer Surgery: “SoFT Study” Retrospective Analysis" Current Oncology 31, no. 8: 4141-4157. https://doi.org/10.3390/curroncol31080309

Article Metrics

Back to TopTop