Original Article

Sentinel Lymph Node Biopsy After Neoadjuvant Chemotherapy in cN0 Breast Cancer: Impact of HER2-Positive Status on Survival

10.4274/ejbh.galenos.2024.2023-11-3

  • Juan Alors-Ruiz
  • Salomé Sanz-Viedma
  • Francisco Javier Fernández-Garcia
  • Francisco Sendra-Portero

Received Date: 30.11.2023 Accepted Date: 02.01.2024 Eur J Breast Health 2024;20(2):94-101

Objective:

High rates of negative sentinel lymph node biopsy (SLNB) in clinically node-negative (cN0) breast cancer (BC) after neoadjuvant chemotherapy (NAC) have been described. These results are associated with triple-negative (TNBC) and human epidermal growth factor receptor 2 (HER2+) subtypes achieving pathologic complete response (pCR). This study evaluates predictive variables and survival in order to assess the possible omission of SLNB after NAC.

Materials and Methods:

Prospective study of women with cN0 BC treated with NAC and subsequent surgery, between April 2010 and May 2021. SLNB technique included, performing axillary lymphadenectomy in the absence of detection or SLNB-positivity. Multivariable logistic regression was used for analysis of NAC-response and SLNB-results in molecular subtypes: HR-/HER2+, TNBC, HR+/HER2- and HR+/HER2+. Kaplan-Meyer and log-rank were used for survival analysis.

Results:

A total of 179 patients (50.5±10.1 years) were included. Of these, 39.7% achieved pCR (ypT0/Tis). HR-negative subtypes had higher pCR rates (HR-/HER2+: 59.4%; TNBC: 53.4%), with no cases of SLNB-positive. With residual disease, HR-/HER2+ and TNBC showed low rates of SLNB-positivity (6.7% and 10.3%) versus HR+ (HR+/HER2+: 20%; HR+/HER2-: 44%; p<0.001). Multivariable analysis identified independent predictors of SLNB-negativity (p<0.0001) to be: HR- [odds ratio (OR)=0.15; 95% confidence interval (CI): 0.06–0.37; p = 0.0001], HER2+ (OR=0.34; 95% CI: 0.14–0.81; p = 0.015) and high-grade Nottingham (OR=0.42; 95% CI: 0.18–0.99; p = 0.048). Disease-free survival showed worse outcomes with SLNB-positivity (p<0.0001), HR+/HER2- (p = 0.0277), larger tumor size (p = 0.002) and residual disease after NAC (p<0.0001).

Conclusion:

Patient selection based on NAC response, molecular subtype, and survival outcomes is a priority for establishing individualized therapeutic strategies after NAC. Molecular subtypes with higher pCR rates and lower rates of SLNB-positivity could benefit from non-invasive strategies that include omission of SLNB.

Keywords: HER-2/neu, neoadjuvant chemotherapy, sentinel lymph node biopsy, survival, triple negative breast cancer

Key Points

• SLNB after NAC safe and effective treatment for cN0.

• Molecular subtype tumor size predictors pCR.

• NAC response strongest prognosis predictor.

• SLNB-negative pCR achieved better prognosis.

• HER2+ benefit omission SLNB technique.


Introduction

Sentinel lymph node biopsy (SLNB) in breast cancer (BC) is a validated tool for axillary staging after neoadjuvant chemotherapy (NAC) in patients with clinically negative nodes (cN0) (1). Tumor size and BC molecular subtype are important predictors of NAC-response (2). cN0 patients with triple-negative (TNBC) and human epidermal growth factor receptor 2 (HER2+) BC show high rates of SLNB-negativity (ypN0) (3, 4, 5). Patients with a pathological complete response (pCR) show higher disease-free survival (DFS) and overall survival (OS) (6). SLNB after NAC allows better assessment of response to NAC (7, 8, 9). Molecular subtypes are important for predicting SLNB-negativity with high probability of pCR. There is no standard that recommends omitting axillary surgery in cN0 patients undergoing NAC (10). There are currently several ongoing trials (11), including two prospective trials that aim to assess axillary recurrence-free survival (ARFS) when omitting SLNB after NAC in patients initially diagnosed as cN0 (12, 13). This study presents the survival outcome of a cohort of patients who received NAC, with the aim of providing data for the omission of axillary surgery in selected cases.


Materials and Methods

Between April 2010 and May 2021, 179 women were retrospectively and consecutively included in the study. All patients and their associated data originate from a single healthcare institution: the ‘Hospital Clínico Virgen de la Victoria’ in the city of Malaga, Spain. It is a first-level hospital, a reference center in BC treatment that provides care to a population of 500,000 inhabitants. The following inclusion criteria were established: Age between 18 and 80 years, newly diagnosed invasive breast carcinoma, clinically negative axilla and/or confirmed through Fine-Needle Aspiration Biopsy (FNAB), undergoing complete SLNB technique with a dual tracer, receiving NAC consisting of Anthracyclines + Taxanes or Cyclophosphamide, receiving adjuvant chemotherapy after surgery, receiving local and axillary radiotherapy after surgery, and receiving Trastuzumab and/or Pertuzumab in HER2-positive patients, as well as hormonal therapy in hormone receptor-positive patients.

Exclusion criteria comprised; age >80 years, as international guidelines did not clarify the use of SLNB in this age group at the beginning of the study; history of previous neoplasia, either BC or any other origin; development of a new neoplasia of a different origin than breast; positive metastasis in the biopsy of a suspicious lymph node by FNAB; any other chemotherapy regimen not mentioned in the inclusion criteria; absence of radiotherapy treatment; absence of hormonal treatment if required; and absence of anti-HER2 treatment if required (Figure 1).

The initial anatomopathological diagnosis of the tumor was performed on samples obtained by core needle biopsy. The material was immediately fixed in buffered neutral formalin and embedded in paraffin. Three-millimeter sections were stained with hematoxalin and eosin (H&E) and macroscopically analyzed for tumor type and histological grade, which adhered to the Nottingham (Scarff-Bloom-Richardson) system. Subsequently, an immunohistochemical analysis was performed to define the molecular subtype.

The criteria for NAC indication in cN0 BC patients have been based on the presence of HER2+ or TNBC subtypes and/or the accepted indication for reducing tumor volume to enable more conservative surgery. These NAC indications have been determined by a multidisciplinary team and have been crucial in evaluating the chemotherapy response in these specific cases, thus contributing to establishing a well-defined patient cohort.

SLNB technique was performed by intradermal periareolar injection with 37 MBq of 99mTc-nanocolloid of human serum albumin (Nanocoll®) for lymphoscintigraphy. Intraoperative localization of SLN was performed with gamma probe by an experienced Nuclear Medicine specialist.

During the intraoperative examination, both the tumor and sentinel lymph nodes (SLNs) were promptly sent to the Pathology Department for further analysis. A skilled pathologist conducted a macroscopic evaluation of the lymph node and subsequently sectioned it longitudinally/vertically based on its morphology, creating sections that were 2 mm thick. The most suspicious section, identified macroscopically, was frozen at -20 °C and later cut into 5–10 micrometer-thick sections, which were stained with H&E to assess malignancy. This procedure took approximately 15–25 minutes.

Following the intraoperative assessment of the SLNs, the definitive histopathological study of the tumor and SLN was performed. The tumor was processed with 3-millimeter sections in blocks, and an immunohistochemical study was conducted in separate blocks. Each lymph node was individually fixed in formalin and embedded in separate paraffin blocks. From each block, two 3-micrometer sections were obtained, with an interval between them of 3–5 micrometers, and subsequently stained with H&E. Tumor and lymph node involvement were defined according to the American Joint Committee on Cancer (AJCC - 8th edition) Breast Cancer Staging standard (14) and the Residual Cancer Burden (RCB; MD Anderson Cancer Center, Houston, Texas, USA) (15). This comprehensive approach allowed for accurate assessment and characterization of the NAC response, contributing to the robustness of the study’s findings. Isolated tumor cells, micrometastases, and macrometastases were considered as tumor presence at the lymph node level. The cases from our series evaluated through the Miller and Payne system, before the development of Symmans’ RCB system, were reevaluated and assigned an RCB index and class and yp stage for a correct evaluation of the series. Axillary lymph node dissection (ALD) was performed with intraoperative SLNB-positive and with definitive positive results.

Clinical follow-up after surgery was scheduled every 6–12 months for a period of at least five years.


Statistical Analyses

Clinical variables were prospectively recorded and evaluated with parametrical or non-parametrical test according to appropriateness. Our hypotheses included assessing survival outcomes (DFS, OS, and ARFS) after NAC and identifying predictive factors for negative SLNB results in patients achieving a pCR. The primary outcome was OS, with secondary outcomes including DFS and ARFS. The study’s variables encompassed patients’ demographics, clinical characteristics, tumor subtypes, NAC response, and corresponding outcomes, which were analyzed. For OS and DFS Kaplan-Meier analysis and the log-rank test were used. For all analyses, SPSS, version 22 for Windows was used (SPSS, Inc., Chicago, IL, USA).


Results

The clinical characteristics of the patients are shown in Table 1. Most frequent NAC protocol was anthracyclines and taxanes (n = 156; 87.2%), including docetaxel and cyclophosphamide/carboplatin, palbociclib or T-DM1. HER2 therapy and hormone therapy were used, if indicated. Breast surgery was performed six months after NAC. Median (range) time between diagnosis and NAC was 36 (14–67) days and mean NAC was 5.9±1 months. SLN-negatives (n = 140) not submitted to ALD were followed from diagnosis for a mean of 51±29 months with no case of axillary involvement. There were 36 cases which were SLNB-positive [HR+: 28 (77.8%), TNBC: 6 (16.7%) and HER2+/HR-: 2 (5.5%)] and in three cases ALD was performed due to SLNB non-detection.

In total 71 patients (38.5%) had breast pCR (Table 2) and higher rates was obtained in HER2+ (p = 0.046) and HR- (p<0.0001). HR+/HER2- was associated with breast pCR in 6.8%, compared to 59.4% in HR-/HER2+ and 53.4% TNBC patients (p<0.001). Significant predictors of pCR were HR- (p<0.0001), Nottingham score (p = 0.0013), HER2+ (p = 0.05), and cT/tumour size (p = 0.04/p = 0.0018). HR- (p = 0.0006) and HER2+ (p = 0.0087) were independent predictors of pCR (Table 3).

The most frequent molecular subtype in the 36 patients with ypN+ status (20.5%) was HR+ (77.8%). Breast pCR was a significant predictor of SLNB-negativity (97.2%; p<0.001). The strongest predictors of ypN0 before surgery were molecular subtype (p<0.001), tumour size (p = 0.005), and Nottingham score (p = 0.003) (Table 4).

Disease progression occurred in 21 (11.7%), subdivided into local recurrence (n = 11; 6.15%), and disseminated disease (n = 16; 8.93%). Mean time from surgery to local recurrence was 25±17 months, disseminated disease 26±17 months and exitus 38.7±18 months. Death occurred in 10 cases so that OS was 90.6%. In pCR, the OS at 5 years was 100% (non-pCR 84.2%; p = 0.007). DFS showed significant differences regarding SLNB (p<0.0001), HER2 expression (p = 0.0277), tumour size (p = 0.002), and NAC-response (p<0.0001) (Figure 2).


Discussion and Conclusion

SLN identification reached the recommended value of at least 95% (16, 17). Periareolar intradermal injection can obtain better radiotracer drainage compared to intra- or peritumoral injections. It is important to highlight the absence of axillary recurrence (AR) in the cases of negative SLNB, in line with previous publications (18, 19, 20) suggesting that SLNB performs better than ALD. We do not attribute this to the average length of follow-up, which was longer than in other published studies (34 months) (21), nor to the interval of time until AR. Another reason for the absence of AR was the use of adjuvant systemic treatment, which lowers the risk for local and regional recurrence (22, 23). In the present study, patient selection was based on the chemotherapy course, in which all patients received hormone and anti-HER2 therapy depending on the molecular subtype, NAC was based on anthracyclines and taxanes in 91.6% of cases, of which only 5% did not complete treatment. The increase in the rate of local recurrence in these cases of high fibrosis due to good response to NAC, which translates into pCR, is a matter of concern for surgeons. In our study, we had no recurrence is any patient achieving pCR.

Tumor size and molecular subtypes are independent predictors of pCR (18, 19, 20, 21). Some authors claim that achieving pCR does not completely rule out long-term recurrence. Thus, for the design of our study we took into account the limitation of previous studies (retrospective nature, lack of knowledge of NAC courses, Nottingham scoreing, and pathological data) to evaluate the survival results. We found an OS and a DFS at five years of 100% in the group that achieved pCR, independently of the tumour size at diagnosis and the molecular subtype. The strengths of these results lie in the well-selected patient sample, with a high homogeneity of chemotherapy scheme and an exhaustive registry of the administered cycles and the causes for treatment interruption. The presence of HR+ could negatively influence the pCR rate of the HER2+ group, whereas HR-/HER2+ achieves higher pCR rates, with impact on NAC response and OS/DFS. OS and DFS were 100% in the pCR group, probably due to well-selected patients, with homogeneous NAC protocols and anti-HER2 therapy. Furthermore, and according to literature, there could be a slight difference in prognosis with respect to the in situ presence of tumour after NAC (24, 25). Based on this evidence, another strength of our study is the registry of all variables of the pathological examination of the samples, which provided exact data on staging of the AJCC and RCB of Symmans after NAC. We obtained an OS and a DFS at five years of 100% in the group of women who had an in situ component in the samples that corresponded to the ypTis stage and the pCR category. Therefore, in our study, these women showed the same excellent results regarding OS and DFS at five and eight years as those achieving a complete pCR, categorized as ypT0.

It is worth highlighting that in our study the DFS at five years for our TNBC group, considered as a good response to NAC (pCR rate of 51.7%) was 84.7%, whereas the DFS at five and eight years for the HR+/HER2- group was 73.3% and 54.3%, with a pCR rate of only 6.8%. We explain this notable prognosis difference between groups, compared to other studies (20) by homogeneity in the NAC courses, with subsequent adjuvant chemotherapy.

Current evidence suggests that molecular criteria should be prioritized over anatomical criteria, especially in higher probability of recurrence in patients with HR+ tumours (26, 27, 28). Our OS and DFS results in the HR+/HER2- subtype suggest considering an initial surgery and a later adjuvant treatment, omitting NAC, with the objective of removing the biggest amount of tumour tissue with low probability of response to chemotherapy as soon as possible.

Predictive factors of SLNB would permit the patient selection for omission of SLNB after NAC.

A limitation of this study is that magnetic resonance imaging was not performed (29), without radiologic complete response assessment. Nonetheless, results regarding the association of RCB index and molecular subtype show its value as a predictive tool for breast pCR and negative-SLNB rate. Significant rates of ypN0 in HR-/HER2+ and TNBC, compared to HR+ show molecular subtype as an initial criterion to select patients for omission of SLNB after NAC. Tumour subtype and breast pCR were the strongest predictive characteristics in SLNB-negativity after NAC. Omitting SLNB could be an option in TNBC and HR-/HER2+ who achieve breast pCR, with the support of correct assessment with imaging techniques (30).

The findings of this study affirm that SLNB after NAC is an appropriate, safe and effective treatment for cN0. The most important predictors of pCR were molecular subtype and tumor size. Response to NAC is the strongest predictor with better prognosis if SLNB-negativity and pCR are achieved. A categorization of molecular subtypes based on response to NAC, SLNB and survival is a priority to establish individualized therapeutic strategies after NAC. Molecular subtypes with higher pCR rates and lower SLNB-positivity rates could benefit from non-invasive axillary evaluation strategies that include omission of SLNB.


Acknowledgment

We would like to extend our gratitude to software engineer/UX specialist Jose A. García-Guijarro for his invaluable help and dedicated contribution of his technological expertise to the authors.

Ethics Committee Approval: This study received ethical approval from Institution. Hospital Universitario Virgen de la Victoria and Medicine Faculty of Malaga University.

Informed Consent: Retrospective study.

Authorship Contributions

Surgical and Medical Practices: J.A.-R., S.S.-V., F.J.F.-G.; Concept: J.A.-R., S.S.-V., F.S.-P.; Design: J.A.-R., S.S.-V., F.S.-P.; Data Collection and/or Processing: J.A.-R., S.S.-V., F.J.F.-G.; Analysis and/or Interpretation: J.A.-R., S.S.-V., F.J.F.-G., F.S.-P.; Literature Search: J.A.-R., S.S.-V.; Writing: J.A.-R., S.S.-V., F.J.F.-G., F.S.-P.

Conflict of Interest: The authors have no conflicts of interest to declare.

Financial Disclosure: The authors declared that this study has received no financial support.

  1. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol 2018; 19: 27-39. (PMID: 29242041)
  2. Gajdos C, Tartter PI, Estabrook A, Gistrak MA, Jaffer S, Bleiweiss IJ. Relationship of clinical and pathologic response to neoadjuvant chemotherapy and outcome of locally advanced breast cancer. J Surg Oncol 2002; 80: 4-11. (PMID: 11967899)
  3. Tadros AB, Yang WT, Krishnamurthy S, Rauch GM, Smith BD, Valero V, et al. Identification of Patients With Documented Pathologic Complete Response in the Breast After Neoadjuvant Chemotherapy for Omission of Axillary Surgery. JAMA Surg 2017; 152: 665-670. Erratum in: JAMA Surg 2017; 152: 70. (PMID: 28423171)
  4. Barron AU, Hoskin TL, Day CN, Hwang ES, Kuerer HM, Boughey JC. Association of Low Nodal Positivity Rate Among Patients With ERBB2-Positive or Triple-Negative Breast Cancer and Breast Pathologic Complete Response to Neoadjuvant Chemotherapy. JAMA Surg 2018; 153: 1120-1126. (PMID: 30193375)
  5. Murphy BL, L Hoskin T, Heins CDN, Habermann EB, Boughey JC. Preoperative Prediction of Node-Negative Disease After Neoadjuvant Chemotherapy in Patients Presenting with Node-Negative or Node-Positive Breast Cancer. Ann Surg Oncol 2017; 24: 2518-2525. (PMID: 8484921)
  6. Goorts B, van Nijnatten TJ, de Munck L, Moossdorff M, Heuts EM, de Boer M, et al. Clinical tumor stage is the most important predictor of pathological complete response rate after neoadjuvant chemotherapy in breast cancer patients. Breast Cancer Res Treat 2017; 163: 83-91. (PMID: 28205044)
  7. Kuehn T, Bauerfeind I, Fehm T, Fleige B, Hausschild M, Helms G, et al. Sentinel-lymph-node biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA): a prospective, multicentre cohort study. Lancet Oncol 2013; 14: 609-618. (PMID: 23683750)
  8. Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, et al. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast cancer: the ACOSOG Z1071 (Alliance) clinical trial. JAMA 2013; 310: 1455-1461. (PMID: 24101169)
  9. Fontein DB, van de Water W, Mieog JS, Liefers GJ, van de Velde CJ. Timing of the sentinel lymph node biopsy in breast cancer patients receiving neoadjuvant therapy - recommendations for clinical guidance. Eur J Surg Oncol 2013; 39: 417-424. (PMID: 3473972)
  10. Hersh EH, King TA. De-escalating axillary surgery in early-stage breast cancer. Breast 2022; 62(Suppl 1): 43-49. (PMID: 34949533)
  11. Kuru B. The Adventure of Axillary Treatment in Early Stage Breast Cancer. Eur J Breast Health 2020; 16: 1-15. (PMID: 31912008)
  12. Vrancken Peeters MJ, van Leeuwenhoek A, The Netherlands Cancer Institute: Avoiding Sentinel Lymph Node Biopsy in Breast Cancer Patients After Neoadjuvant Chemotherapy (ASICS). Clinicaltrials.gov 2020. https://beta.clinicaltrials.gov/study/NCT04225858
  13. Reimer T, Glass A, Botteri E, Loibl S, D Gentilini O. Avoiding Axillary Sentinel Lymph Node Biopsy after Neoadjuvant Systemic Therapy in Breast Cancer: Rationale for the Prospective, Multicentric EUBREAST-01 Trial. Cancers (Basel) 2020; 12: 3698. (PMID: 33317077)
  14. No authors listed. Erratum: Breast Cancer-Major changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin 2017; 67: 345. (PMID: 28689371)
  15. Symmans WF, Peintinger F, Hatzis C, Rajan R, Kuerer H, Valero V, et al. Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol 2007; 25: 4414-4422. (PMID: 17785706)
  16. Mocellin S, Goldin E, Marchet A, Nitti D. Sentinel node biopsy performance after neoadjuvant chemotherapy in locally advanced breast cancer: A systematic review and meta-analysis. Int J Cancer 2016; 138: 472-480. (PMID: 26084763)
  17. Canavese G, Bruzzi P, Catturich A, Tomei D, Carli F, Garrone E, et al. Sentinel Lymph Node Biopsy Versus Axillary Dissection in Node-Negative Early-Stage Breast Cancer: 15-Year Follow-Up Update of a Randomized Clinical Trial. Ann Surg Oncol 2016; 23: 2494-2500. (PMID: 26975739)
  18. Domènech A, Benitez A, Bajén MT, Pla MJ, Gil M, Martín-Comín J. Patients with breast cancer and negative sentinel lymph node biopsy without additional axillary lymph node dissection: a follow-up study of up to 5 years. Oncology 2007; 72: 27-32. (PMID: 17998787)
  19. Bañuelos Andrío L, Rodríguez Caravaca G, Argüelles Pintos M, Mitjavilla Casanova M. Biopsia selectiva del ganglio centinela en cáncer de mama: sin recurrencias axilares tras un seguimiento medio de 4,5 años [Selective biopsy of the sentinel lymph node in breast cancer: without axillary recurrences after a mean follow-up of 4.5 years]. Rev Esp Med Nucl Imagen Mol 2015; 34: 81. (PMID: 24560598)
  20. Martelli G, Miceli R, Folli S, Guzzetti E, Chifu C, Maugeri I, et al. Sentinel node biopsy after primary chemotherapy in cT2 N0/1 breast cancer patients: Long-term results of a retrospective study. Eur J Surg Oncol 2017; 43: 2012-2020. (PMID: 28912071)
  21. van der Ploeg IM, Nieweg OE, van Rijk MC, Valdés Olmos RA, Kroon BB. Axillary recurrence after a tumour-negative sentinel node biopsy in breast cancer patients: A systematic review and meta-analysis of the literature. Eur J Surg Oncol 2008; 34: 1277-1284. (PMID: 18406100)
  22. Guenther JM, Hansen NM, DiFronzo LA, Giuliano AE, Collins JC, Grube BL, et al. Axillary dissection is not required for all patients with breast cancer and positive sentinel nodes. Arch Surg 2003; 138: 52-56. (PMID: 12511150)
  23. Fant JS, Grant MD, Knox SM, Livingston SA, Ridl K, Jones RC, et al. Preliminary outcome analysis in patients with breast cancer and a positive sentinel lymph node who declined axillary dissection. Ann Surg Oncol 2003; 10: 126-130. (PMID: 12620906)
  24. von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, et al. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. J Clin Oncol 2012; 30: 1796-1804. (PMID: 22508812)
  25. van der Noordaa MEM, van Duijnhoven FH, Cuijpers FNE, van Werkhoven E, Wiersma TG, Elkhuizen PHM, et al. Toward omitting sentinel lymph node biopsy after neoadjuvant chemotherapy in patients with clinically node-negative breast cancer. Br J Surg 2021; 108: 667-674. (PMID: 34157085)
  26. Ruano R, Ramos M, García-Talavera JR, Ramos T, Rosero AS, González-Orus JM, et al. Sentinel node biopsy after neoadjuvant chemotherapy in breast cancer. Its relation with molecular subtypes. Rev Esp Med Nucl Imagen Mol 2014; 33: 340-345. (PMID: 24856234)
  27. Metzger-Filho O, Sun Z, Viale G, Price KN, Crivellari D, Snyder RD, et al. Patterns of Recurrence and outcome according to breast cancer subtypes in lymph node-negative disease: results from international breast cancer study group trials VIII and IX. J Clin Oncol 2013; 31: 3083-3090. (PMID: 23897954)
  28. van Nijnatten TJ, Simons JM, Moossdorff M, de Munck L, Lobbes MB, van der Pol CC, et al. Prognosis of residual axillary disease after neoadjuvant chemotherapy in clinically node-positive breast cancer patients: isolated tumor cells and micrometastases carry a better prognosis than macrometastases. 2027; 163: 159-166. (PMID: 28213782)
  29. Tasoulis MK, Lee HB, Yang W, Pope R, Krishnamurthy S, Kim SY, et al. Accuracy of Post-Neoadjuvant Chemotherapy Image-Guided Breast Biopsy to Predict Residual Cancer. JAMA Surg 2020; 155: e204103. (PMID: 33026457)
  30. Garcia-Tejedor A, Falo C, Quetglas C, Soler T, Marqueta B, Ortega R, et al. Feasibility, accuracy and prognosis of sentinel lymph node biopsy before neoadjuvant therapy in breast cancer. A prospective study. Int J Surg 2017; 39: 141-147. (PMID: 28153783)