Sentinel lymph node biopsy following neoadjuvant chemotherapy: an evidence-based review and recommendations for current practice
Review Article

Sentinel lymph node biopsy following neoadjuvant chemotherapy: an evidence-based review and recommendations for current practice

Francisco Pimentel Cavalcante1^, Eduardo Camargo Millen2^, Guilherme Garcia Novita3^, Felipe Pereira Zerwes4^, André Mattar5^, Rafael Henrique Szymanski Machado6^, Antônio Luiz Frasson4,7^

1Department of Breast Surgery, Hospital Geral de Fortaleza (HGF), Fortaleza, Ceará, Brazil; 2Department of Research, Instituto Oncoclinicas, Rio de Janeiro, RJ, Brazil; 3Department of Breast Surgery, Instituto Oncoclinicas, São Paulo, SP, Brazil; 4Department of Breast Surgery, Pontificia Universidade Católica do Rio Grande do Sul (PUCRS), RS, Brazil; 5Department of Breast Surgery, Hospital Pérola Byington, São Paulo, SP, Brazil; 6Department of Gynecology and Breast Surgery, Hospital Federal da Lagoa, Rio de Janeiro, RJ, Brazil; 7Department of Oncology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil

Contributions: (I) Conception and design: All authors; (II) Administrative support: All authors; (III) Provision of study materials or patients: All authors; (IV) Collection and assembly of data: All authors; (V) Data analysis and interpretation: All authors; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

^ORCID: Francisco Pimentel Cavalcante, 0000-0002-7156-2890; Eduardo Camargo Millen, 0000-0002-2113-6324; Guilherme Garcia Novita, 0000-0003-2983-3199; Felipe Pereira Zerwes, 0000-0002-1643-727X; André Mattar, 0000-0001-5973-623X; Rafael Henrique Szymanski Machado, 0000-0003-1848-7630; Antônio Luiz Frasson, 0000-0003-1860-6898.

Correspondence to: Francisco Pimentel Cavalcante. Rua Avila Goulart 900, Papicu, 60150-160 Fortaleza, Ceará, Brazi. Email: fpimentelcavalcante@gmail.com.

Abstract: Sentinel lymph node biopsy (SLNB) at upfront surgery is the gold-standard surgical method for axillary lymph node staging in early stage breast cancer: the technique provides adequate information regarding axillary status, with similar oncological safety and lower morbidity compared to axillary dissection, despite the false negative rates. Neoadjuvant chemotherapy (NACT), traditionally used for locally advanced breast cancer, plays an important role in the treatment of early stage breast cancer, making downstaging possible in axillary lymph node and breast cancer, thus minimizing the impact of surgery and reducing morbidity, as well as enabling patients with residual disease to be selected for adjuvant treatment. In this respect, the role of SLNB has proved controversial, particularly in view of the lack of data from randomized clinical trials on this subject. Currently, the de-escalation of axillary surgery after NACT is mainly based on retrospectives studies and false negative rates. This paper reviews current evidence on the management of axillary surgery following NACT under different circumstances, with suggested recommendations in each scenario: clinically negative nodes at diagnosis and SLNB after NACT, clinically positive nodes at diagnosis and SLNB after NACT, positive SLNB following NACT and finally the possibility of omitting axillary surgery in good responders.

Keywords: Breast cancer; sentinel node biopsy; neoadjuvant chemotherapy (NACT); axillary dissection


Submitted Nov 08, 2022. Accepted for publication Feb 09, 2023. Published online Feb 16, 2023.

doi: 10.21037/cco-22-110


Introduction

Sentinel lymph node biopsy (SLNB) at upfront surgery is the gold-standard surgical method for axillary lymph node staging in early stage breast cancer. Survival and oncological outcomes are pivotal when validating a new treatment or procedure; however, with respect to the sentinel lymph node (SLN), early studies overlooked these parameters and considered the rate of false-negative findings as the principal endpoint. In those series, patients with early-stage tumors were submitted to SLNB followed by conventional axillary dissection. The ideal false-negative rate (FNR) was defined as ≤5%, a number arbitrarily established in consensuses of specialists (1-3). Subsequent randomized clinical trials assessing oncological outcomes such as overall survival, disease-free survival and local control showed oncological safety similar to that achieved with axillary dissection. The FNR in some analyses, however, exceeded the value considered ideal in previous studies (4.6–9.8%) (4-6). Nevertheless, the technique has the advantages of reducing surgical morbidity with no negative effect on prognosis (4-7).

Neoadjuvant chemotherapy (NACT) was developed to convert originally inoperable tumors into operable tumors and to evaluate response to treatment. The National Surgical Adjuvant Breast and Bowel Project studies, NSABP B-18 and NSABP B-27, showed that initiating treatment with chemotherapy or providing adjuvant chemotherapy did not change prognosis (8,9). In the NSABP B-18 study, the group that began treatment with NACT had a higher rate of breast preservation (69.8% vs. 59.8%) and a similar rate of local recurrence (HR =0.98; 95% CI: 0.83–1.15; P=0.78) (8). In addition, neoadjuvant therapy showed a non-significant trend towards a better prognosis in women under 50 years of age (9). Prognosis was better in the women who experienced pathologic complete response (10% of those in the doxorubicin + cyclophosphamide arm and 17% of those in the doxorubicin + cyclophosphamide + docetaxel arm) (9). This came to be seen as a means of identifying those patients who would respond well to the drugs, but resulted in no changes in treatment (8). Based on those studies, the indications for NACT became highly objective: locally advanced, inoperable tumors (T3, T4, N1-3) or tumors for which breast-conserving surgery was not possible. In cases of tumors that could be treated with breast-conserving surgery (T1/T2, N0), surgery was generally the first treatment option. However, as knowledge increased regarding tumor biology and new treatments were implemented, the indications for NACT changed to include cases of tumors that can be treated with breast-conserving surgery. In this new scenario, axillary downstaging in responding tumors leads us to consider SLNB after NACT to avoid morbidity from complete axillary dissection.

Progress made in SLNB after NACT has followed the same pathway as upfront SLNB in that no randomized studies on oncological safety have been conducted up to the present time, irrespective of initial axillary node status. In patients with initially negative axilla (cN0) who receive NACT, FNRs are generally acceptable (≤10%) (10-12), and, indeed, data from non-randomized studies have shown low rates of axillary recurrence (12-14). Conversely, patients with clinically positive axilla (cN1/2) prior to treatment and who experience clinical complete response represent a more challenging group. In addition to the lack of randomized studies assessing clinical outcomes, overall FNRs are considered high, possibly impacting on local control and important prognostic information (15-23). More recently, there has been an increasing tendency to omit axillary dissection in patients with a positive SLNB (ypN+) following NACT, as in cases of upfront surgery in circumstances similar to those of the Z11 study, even when the residual burden is high (24-29). Another line of investigation has evaluated the consequences of omitting any axillary surgery in patients who respond well to NACT (30-37). The objective of the present paper is to review current findings on SLNB following NACT in different clinical circumstances, based on the best available evidence.

Clinically negative nodes at diagnosis and SLNB after NACT

In patients with initially negative axilla (cN0), SLNB following NACT has been performed over the years, principally on the basis of FNRs similar to those found with upfront surgery. Some meta-analyses have reported overall FNRs of around 10% irrespective of the use of dual tracers or the number of lymph nodes removed (10-12). These rates are similar to that found in the NSABP B-32 study and are considered safe (Table 1). One of those reviews included 2,148 patients from studies conducted between 1993 and 2009. The analysis resulted in an SLN identification rate of 90.9% (88–93.1%), FNR of 10.5% (8.1–13.6%), accuracy of 94.4%, and negative predictive value of 89% (12). In another meta-analysis involving 24 studies conducted between 2000 and 2007 with a total of 1,799 patients, the SLN identification rate was 89.6% (86–92.3%), with a FNR of 8.4% (6.4–10.9%) (10). On the other hand, studies on clinical outcome are limited to data from non-randomized trials. A retrospective study conducted at the MD Anderson Cancer Center evaluated cN0, T1–T3 patients undergoing SLNB following NACT (n=575) or upfront surgery (n=3,171). Lymph node recurrence rate was 1.2% in the NACT group and there was no difference in terms of disease-free survival or overall survival (13). The GANEA-2 study was a prospective multi-institutional French cohort study aimed at assessing the accuracy and safety of SNLB after NACT in initially cN0 and pN1 patients. Of the 419 initially cN0 patients treated with SNLB alone, only one patient had lymph node recurrence after a mean follow-up time of 36 months, while patients allocated to the pN1 group underwent SLN dissection (SLND) and axillary dissection (FNR: 11.9%) (14).

Table 1

Studies on false-negative rates in initially negative axilla (cN0) patients submitted to sentinel lymph node biopsy following neoadjuvant chemotherapy

Author Number of patients False-negative rate
Shirzadi et al. (11) 1,521 7% (5–9%)
Kelly et al. (10) 1,799 8.4% (6.4–10.9%)
van Deurzen et al. (12) 2,148 10.5% (8.1–13.6%)

Initially positive axilla (cN1/2) and negative SLNB following NACT

Traditionally, the FNRs for SLNB in patients with initially clinically positive axilla who achieved clinical/imaging complete response following NACT were considered unacceptable (>20%); therefore, all these women were submitted to axillary dissection despite the fact that a considerable number of patients had axillary pathologic complete response (11). The SENTINA, Z1071 and SN-FNAC studies changed this concept. The overall FNRs reported by those studies for patients submitted to NACT were 14.2%, 12.6% and 13.3%, respectively, rates that were higher than that of 10% previously specified as being safe but lower than other previously reported rates (15-17). Furthermore, analysis of subgroups revealed that the identification of more than three lymph nodes; SLN mapping using dual tracer imaging with the patent blue dye and radioisotope combination technique; clipping the affected lymph node prior to NACT; or even the use of immunohistochemistry reduced the FNR to <10% (15-17). A recent meta-analysis that included 1,921 patients with biopsy-proven node-positive breast cancer reported an identification rate of 90% and a FNR of 14%; however, when three or more lymph nodes were identified, the FNR fell to 4% (19).

Clipping the metastatic lymph node before neoadjuvant treatment and removing the clipped node at the time of surgery have been suggested as a means of reducing FNRs (18). In the Z1071 study, a FNR of 7.2% was found in a subgroup of patients in whom the metastatic lymph node was marked with a clip prior to NACT and then resected (16). The findings varied with the technique used to identify the clipped lymph node: the SLN was indeed the clipped lymph node in 78% of cases (101/130) when dual tracers were used and 50% when a single tracer was used (16). In a retrospective study conducted at the MD Anderson Cancer Center, of the 134 patients who underwent SLND, the clipped node was not identified as an SLN in 23% (31 of 134) of patients. The overall FNR was 10.1% for SLNB, 4.2% for clipped lymph nodes and 2% when the SLN was marked and located using iodine-125 seeds (I-125 seed), a procedure referred to as targeted axillary dissection (TAD). In TAD, patients with a clipped node (biopsy-confirmed nodal metastases) are marked with an I-125 seed. This is performed after undergoing NACT and receiving an injection of mapping agents (radioisotope and/or blue dye) prior to surgery. A gamma probe is then used to identify the seed and the radioisotope-containing nodes removed during surgery, including nodes containing blue dye alone or those found to be palpable (18). In another study, the identification rate was 77.8% (329/423) for cases of clipped lymph node and 86.9% (199/229) when an I-125 seed was used. In 35.2% of cases, the clipped lymph node proved not to be the sentinel lymph node. The authors reported a FNR of 7.2% for the clipped lymph node technique and 4.2% for the I-125 seed technique (38) (Table 2).

Table 2

Identification rates and false-negative rates with clipped lymph node and targeted axillary dissection

Author Clipped lymph node not identified False-negative rate of clipped lymph node False-negative rate with targeted axillary dissection
Boughey et al. (16) 37% 7.2% 1.4%
Caudle et al. (18) 23% 4.2% 2.0%
Kuemmel et al. (38) 22% 7.2% 4.2%

The efforts made to reduce the FNR in these circumstances reflect the absence of data from randomized clinical trials on oncological safety. Nevertheless, data from single centers suggest that axillary recurrence could be very low (39-42). In a consecutive cohort of 688 patients submitted to NACT at the European Institute of Oncology and followed up for ten years, axillary recurrence was 1.6% in the initially cN1/2 group (n=123) (40). The higher rate of false-negative results was considered to pose no clinical risk and the SLNB technique was recommended to be used in these circumstances irrespective of the number of lymph nodes resected or whether dual-tracer mapping or clipping of the axillary lymph node is used (in 50% of cases in that series, only one SLN was resected) (40). In another analysis conducted at the McGill University Medical School, including 58 cN1/2 patients who obtained clinical complete response and in whom SLNB was used, there was no axillary lymph node recurrence during follow up (39). In a series of 769 consecutive patients with positive lymph nodes submitted to NACT at the Memorial Sloan Kettering Cancer Center, 555 had clinical complete response and in 234 cases (42%) axillary dissection was avoided through the use of dual-tracer mapping and the identification of at least three lymph nodes (41). After 4 years of follow-up, there was only one case (0.4%) of axillary recurrence synchronous with local recurrence in the entire cohort and that case consisted of a patient who had refused radiotherapy (41). In another study conducted at the Mayo Clinic with 159 initially cN1/2 patients submitted to SLNB without axillary dissection, one patient had axillary lymph node recurrence over a short follow-up time (42) (Table 3).

Table 3

Studies of patients submitted to sentinel lymph node biopsy without axillary dissection who had initially positive axillary lymph nodes prior to neoadjuvant chemotherapy

Author Initial axilla Number of patients Regional recurrence
Piltin et al. (42) cN1/2 159 <1.0%
Kahler-Ribeiro-Fontana et al. (40) cN1/2 123 1.6%
Barrio et al. (41) cN1 234 <1.0%
Wong et al. (39) cN1/2 58 0%

The RTOG 1304/ NSABP B51 study will provide definitive data on this subject (43). The search for a lower FNR remains most relevant, since, even if there is no locoregional effect of a higher FNR, identifying a residual lesion could be crucial when deciding on whether to use systemic adjuvant therapy with capecitabine for triple-negative tumors or olaparib for patients with the BRCA mutations and trastuzumab emtansine (T-DM1) in HER2 disease. Studies with these drugs have shown important benefits in women with residual disease following NACT (20-22).

Positive SLN following NACT

In recent years, following publication of the American College of Surgeons Oncology Group (Alliance) ACOSOG Z0011 (Z11) study (44), there has been an increasing tendency to omit axillary dissection, even in patients with SLN metastases after NACT (23,24). Indeed, the Z11 study showed excellent locoregional control with the omission of additional axillary surgery in patients with positive SLN. However, as in other studies that also evaluated the omission of axillary dissection at upfront surgery, patients submitted to NACT were not included in those analyses (45-49). Furthermore, residual disease following NACT means resistance to systemic treatment, hence the possibility of a high residual burden of disease. Single center analyses have shown high residual cancer burden in around 60% of cases after NACT irrespective of the extent of metastasis (micro- or macro-metastasis) or of the subtype of the disease (26-28). This rate compares with the rate of 27% found in the Z11 trial and 13% for micro-metastasis alone (IBCSG 23-01) at upfront surgery (44,49). It is reasonable to assume that the results found at upfront surgery may not have the same outcome following NACT.

The NSABP B-18 and B-27 studies evaluated the role of anthracyclines or of anthracyclines associated with taxanes as neoadjuvant therapy for patients submitted either to breast-conserving surgery with radiotherapy or to mastectomy without radiotherapy associated with axillary dissection. A combined analysis of these studies (50), which evaluated 335 locoregional recurrences (12.6% in mastectomized patients and 10.3% following breast-conserving surgery), found high accumulated 10-year regional recurrence rates in patients with initially positive axillary lymph nodes that remained positive following NACT and axillary dissection (7.5% in women ≥50 years of age and 8.7% in those <50 years of age at breast-conserving surgery with radiotherapy). More recent studies have produced controversial results. A study conducted in the Netherlands (51) using initial axillary ultrasonography reported four cases of regional recurrence (4/118 or 3.4%) after three years of follow-up in patients with residual disease in the SLN following NACT who had fewer than four affected lymph nodes at axillary ultrasonography and who were given only radiotherapy as additional axillary treatment, without axillary dissection. Another two retrospective studies involving a small number of patients found no differences in terms of oncological outcome (52,53). On the other hand, a study conducted using the National Cancer Database (54) included 1,617 women initially cN1 submitted to NACT and compared axillary dissection with SLNB associated with axillary radiotherapy in a design similar to that of the ongoing prospective randomized study conducted by the ALLIANCE group (A11202). Better survival rates were associated with axillary dissection. In an exploratory analysis, the authors reported similar survival rates in hormone receptor (HR)-positive tumors and metastasis in a single lymph node (Table 4).

Table 4

Oncological outcomes in studies that evaluated sentinel lymph node biopsy alone compared to axillary lymph node dissection when residual axillary disease is present.

Author Patients Duration of follow-up Oncological outcome
Ling et al. (52) 161 3 years 92.6% with SLNB vs. 96.4% with AD; P=0.616
Chun et al. (53) 324 71 months 91.2% with SLNB vs. 91.4% with AD; P=0.594
Almahariq et al. (54) 1,617 5 years 71% with SLNB vs. 77% with AD; P=0.01§

Oncological outcome evaluated: , regional control; , axillary recurrence; §, overall survival. SLNB, sentinel lymph node biopsy; AD, axillary dissection.

Omitting axillary surgery in good responders

SLNB allowed similar prognostic information to be obtained; however, although morbidity is less than with axillary dissection, the procedure is not without complications. The largest study on SLNB (NSABP-B32) found numbness in 7.5% of cases, paresthesia in 6% and lymphedema of the ipsilateral upper limb in 8% in an evaluation performed 36 months after the procedure (4-7). This led to an ongoing debate on whether surgical procedures on the axilla should be avoided under specific circumstances such as when information on axillary status would not affect the decision regarding whether to use systemic or regional therapy or when prior evaluation with imaging tests would enable the negative predictive value of the lymph nodes to be established with a high degree of certainty. The problem of omitting axillary surgery under these conditions with excellent responders is that evaluating the presence of residual disease is very important when making a decision regarding adjuvant therapy. Indeed, in patients with initially positive axilla who achieve clinical complete response to NACT, SLNB will identify residual disease in more than 50% of cases (16).

In HER2-positive breast cancer, the results of the KATHERINE randomized clinical trial (21) showed that the use of T-DM1 in patients with any residual lesion increases disease-free survival. In triple-negative breast cancer, the CREATE-X randomized clinical trial evaluated the addition of capecitabine for patients with residual disease and confirmed that its use increased disease-free and overall survival (20). More recently, the adjuvant use of olaparib in patients with the BRCA gene mutations and residual disease after NACT was associated with better invasive disease-free survival in the OlympiA study (22). Therefore, ignoring pathologic lymph node status in such cases could result in the omission of treatments capable of changing relevant clinical outcomes. On the other hand, the KEYNOTE-522 study (55), which used pembrolizumab associated with chemotherapy as neoadjuvant therapy in triple-negative tumors, showed that the addition of immunotherapy increased pathologic complete response and disease-free survival. It can also be used as adjuvant therapy irrespective of the presence of a residual lesion. In this case, knowledge of pathologic lymph node status would not affect the indication of pembrolizumab. However, a combination of pembrolizumab and capecitabine in patients who used the immunotherapy drug as neoadjuvant therapy and did not achieve pathologic complete response was not evaluated. Another relevant factor in relation to lymph node status is the indication of regional radiotherapy for those patients with positive axillary lymph nodes following NACT. The current tendency is to recommend it in cases of axillary lymph node metastasis following neoadjuvant treatment (56,57). Nevertheless, with the previously presented data on the extremely low likelihood of positive axillary lymph nodes, we believe that the number needed to treat (NNT) to prevent regional recurrence in cases in which radiotherapy is omitted could be very high.

The advances made in these NACT regimes, with the addition of new drugs and a more appropriate selection of patients has, on the other hand, resulted in a high rate of pathologic complete response, leading some investigators to question whether surgery could be avoided under these circumstances (30-37). In 2017, a study conducted in the MD Anderson Cancer Center with 572 cT1/2 cN0/1 patients with triple-negative and HER2-positive breast cancer who underwent NACT showed that of the 290 cN0 patients included, 116 obtained complete pathologic response in the breast (32). When pathologic response in the axillary lymph nodes was evaluated, it was found that none of the patients had axillary metastasis, showing that the likelihood of residual axillary disease following NACT and complete pathologic response in the breast in patients initially presenting with clinically negative axilla in these subtypes is very low. Another study, conducted using data from the United States National Cancer Database, included 30,281 patients and encompassed all the immunohistochemical profiles. The probability of finding a positive lymph node in cT1/2 cN0 patients with clinically negative lymph nodes following NACT was evaluated in patients who achieved pathologic complete response in the breast. Residual disease in axillary lymph nodes was found in 1.0%, 1.6%, 2.1% and 4% of cases of HER2-positive/HR-negative, triple-negative (HER2-negative/HR-negative), HER2-positive/HR-positive and HR-positive/HER2-negative tumors, respectively (30). Similar data were reported from another two studies published later (Table 5) (31,33).

Table 5

The probability of disease in the lymph node in patients with cT1/2cN0 tumors submitted to neoadjuvant chemotherapy and with pathologic complete response in the breast

Author Number of patients HER2 Triple-negative
Barron et al. (30) 5,377 HR-positive: 2.1%; HR-negative: 1% 1.6%
Samiei et al. (31) 986 HR-positive: 1.6%; HR-negative: 0% 1.5%
Tadros et al. (32) 116 0% 0%
van der Noordaa et al. (33) 89 0% 0%

HER2, human epidermal growth factor receptor 2; HR, hormone receptor.

With the routine use of axillary ultrasonography in patients with an indication for NACT, these numbers should currently be even lower in series with larger sample sizes (51,58,59). Indeed, some studies are evaluating the role of axillary ultrasonography in omitting axillary surgery, either during upfront surgery or even after NACT (34-37) (Table 6). The EUBREAST-01 trial (37) was conducted under the concept that the probability of axillary disease in patients with triple-negative and HER2-positive breast cancer with pathologic complete response in the breast after NACT and normal findings at axillary ultrasonography is extremely low and is evaluating omitting SLNB in such cases. The primary endpoint of the study is 3-year axillary lymph node recurrence-free survival, with the acceptable rate being ≥98.5%. The patients who did not achieve pathologic complete response in the breast will undergo further surgery for axillary management. A result of less than 96% in that study would be considered negative.

Table 6

Ongoing studies to evaluate the role of axillary ultrasonography in omitting sentinel lymph node biopsy at upfront surgery or after neoadjuvant chemotherapy

Study Upfront surgery Neoadjuvant chemotherapy
SOUND (34) Yes No
INSEMA (35) Yes No
VENUS (36) Yes Yes
EUBREAST-01 (37) No Yes

Conclusions and recommendations

Initially negative axilla (cN0) and SLNB

SLNB can be performed in initially cN0 patients following NACT without further concern with respect to the number of lymph nodes removed or the use of dual-tracer mapping, since the overall FNR is similar to that with upfront surgery and non-randomized studies have shown a low recurrence rate.

Initially positive axilla (cN1/2) and clinical complete response following NACT

In these circumstances, based on a higher FNR, the absence of randomized clinical trials for clinical outcomes and with the objective of selecting patients for adjuvant therapy when there is residual disease, tactics to reduce the FNR should be encouraged until the results of randomized clinical trials become available.

Positive SLN following NACT

Axillary dissection should be the current standard until the results of randomized clinical trials become available, since the residual axillary burden is high irrespective of the extent of the lymph node metastasis. In addition, data from non-randomized trials are debatable.

Omission of axillary surgery in good responders

Axillary surgery should be performed routinely in all cases irrespective of clinical response or findings at imaging tests following NACT, since there are no data from randomized clinical trials on oncological safety and there is a considerable risk of missing candidates for adjuvant therapy. Specific circumstances are being evaluated in clinical trials.


Acknowledgments

Funding: None.


Footnote

Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-22-110/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-22-110/coif). FPC received honoraria for lectures from Roche, AstraZeneca, MSD, Pfizer and Libbs and participated in a Data Safety Monitoring Board or Advisory Board for Roche, MSD and Pfizer. FPZ received honoraria for lectures from AstraZeneca, MSD and Novartis. AM received consulting fees from MAPE Solutions, Daiichi Sankyo, AstraZeneca and Roche; honoraria for lectures from Daiichi Sankyo, AstraZeneca and Roche; support for attending meetings/travel from Daiichi Sankyo; and participated in a Data Safety Monitoring Board or Advisory Board for MAPE Solutions, Daiichi Sankyo and AstraZeneca. RHSM received honoraria for lectures from Roche, AstraZeneca, Pfizer, and Merck Sharp and Dohme. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Veronesi U, Paganelli G, Galimberti V, et al. Sentinel-node biopsy to avoid axillary dissection in breast cancer with clinically negative lymph-nodes. Lancet 1997;349:1864-7. [Crossref] [PubMed]
  2. Cody HS 3rd, Borgen PI. State-of-the-art approaches to sentinel node biopsy for breast cancer: study design, patient selection, technique, and quality control at Memorial Sloan-Kettering Cancer Center. Surg Oncol 1999;8:85-91. [Crossref] [PubMed]
  3. Veronesi U, Paganelli G, Viale G, et al. Sentinel lymph node biopsy and axillary dissection in breast cancer: results in a large series. J Natl Cancer Inst 1999;91:368-73. [Crossref] [PubMed]
  4. Krag DN, Anderson SJ, Julian TB, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol 2010;11:927-33. [Crossref] [PubMed]
  5. Veronesi U, Paganelli G, Viale G, et al. A randomized comparison of sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med 2003;349:546-53. [Crossref] [PubMed]
  6. Ashikaga T, Krag DN, Land SR, et al. Morbidity results from the NSABP B-32 trial comparing sentinel lymph node dissection versus axillary dissection. J Surg Oncol 2010;102:111-8. [Crossref] [PubMed]
  7. Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: the ALMANAC Trial. J Natl Cancer Inst. 2006;98:599-609. Erratum in: J Natl Cancer Inst 2006;98:876. [Crossref] [PubMed]
  8. Fisher B, Bryant J, Wolmark N, et al. Effect of preoperative chemotherapy on the outcome of women with operable breast cancer. J Clin Oncol 1998;16:2672-85. [Crossref] [PubMed]
  9. Rastogi P, Anderson SJ, Bear HD, et al. Preoperative chemotherapy: updates of National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27. J Clin Oncol 2008;26:778-85. [Crossref] [PubMed]
  10. Kelly AM, Dwamena B, Cronin P, et al. Breast cancer sentinel node identification and classification after neoadjuvant chemotherapy-systematic review and meta analysis. Acad Radiol 2009;16:551-63. [Crossref] [PubMed]
  11. Shirzadi A, Mahmoodzadeh H, Qorbani M. Assessment of sentinel lymph node biopsy after neoadjuvant chemotherapy for breast cancer in two subgroups: Initially node negative and node positive converted to node negative - A systemic review and meta-analysis. J Res Med Sci 2019;24:18. [Crossref] [PubMed]
  12. van Deurzen CH, Vriens BE, Tjan-Heijnen VC, et al. Accuracy of sentinel node biopsy after neoadjuvant chemotherapy in breast cancer patients: a systematic review. Eur J Cancer 2009;45:3124-30. [Crossref] [PubMed]
  13. Hunt KK, Yi M, Mittendorf EA, et al. Sentinel lymph node surgery after neoadjuvant chemotherapy is accurate and reduces the need for axillary dissection in breast cancer patients. Ann Surg 2009;250:558-66. [Crossref] [PubMed]
  14. Classe JM, Loaec C, Gimbergues P, et al. Sentinel lymph node biopsy without axillary lymphadenectomy after neoadjuvant chemotherapy is accurate and safe for selected patients: the GANEA 2 study. Breast Cancer Res Treat 2019;173:343-52. [Crossref] [PubMed]
  15. Kuehn T, Bauerfeind I, Fehm T, 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;14:609-18. [Crossref] [PubMed]
  16. Boughey JC, Suman VJ, Mittendorf EA, et al. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast cancer: the ACOSOG Z1071 (Alliance) clinical trial. JAMA 2013;310:1455-61. [Crossref] [PubMed]
  17. Boileau JF, Poirier B, Basik M, et al. Sentinel node biopsy after neoadjuvant chemotherapy in biopsy-proven node-positive breast cancer: the SN FNAC study. J Clin Oncol 2015;33:258-64. [Crossref] [PubMed]
  18. Caudle AS, Yang WT, Krishnamurthy S, et al. Improved Axillary Evaluation Following Neoadjuvant Therapy for Patients With Node-Positive Breast Cancer Using Selective Evaluation of Clipped Nodes: Implementation of Targeted Axillary Dissection. J Clin Oncol 2016;34:1072-8. [Crossref] [PubMed]
  19. Tee SR, Devane LA, Evoy D, et al. Meta-analysis of sentinel lymph node biopsy after neoadjuvant chemotherapy in patients with initial biopsy-proven node-positive breast cancer. Br J Surg 2018;105:1541-52. [Crossref] [PubMed]
  20. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med 2017;376:2147-59. [Crossref] [PubMed]
  21. von Minckwitz G, Huang CS, Mano MS, et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N Engl J Med 2019;380:617-28. [Crossref] [PubMed]
  22. Tutt ANJ, Garber JE, Kaufman B, et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N Engl J Med 2021;384:2394-405. [Crossref] [PubMed]
  23. Cavalcante FP, Millen EC, Zerwes FP, et al. Role of Axillary Surgery After Neoadjuvant Chemotherapy. JCO Glob Oncol 2020;6:238-41. [Crossref] [PubMed]
  24. Kantor O, Pesce C, Liederbach E, et al. Are the ACOSOG Z0011 Trial Findings Being Applied to Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy? Breast J 2017;23:554-62. [Crossref] [PubMed]
  25. Cavalcante FP, Zerwes F, Millen EC, et al. Management of the positive sentinel lymph node following neoadjuvant chemotherapy: results of a survey conducted with breast surgeons. Ecancermedicalscience 2022;16:1357. [Crossref] [PubMed]
  26. Moo TA, Edelweiss M, Hajiyeva S, et al. Is Low-Volume Disease in the Sentinel Node After Neoadjuvant Chemotherapy an Indication for Axillary Dissection? Ann Surg Oncol 2018;25:1488-94. [Crossref] [PubMed]
  27. Barron AU, Hoskin TL, Boughey JC. Predicting Non-sentinel Lymph Node Metastases in Patients with a Positive Sentinel Lymph Node After Neoadjuvant Chemotherapy. Ann Surg Oncol 2018;25:2867-74. [Crossref] [PubMed]
  28. Moo TA, Pawloski KR, Flynn J, et al. Is Residual Nodal Disease at Axillary Dissection Associated with Tumor Subtype in Patients with Low Volume Sentinel Node Metastasis After Neoadjuvant Chemotherapy? Ann Surg Oncol 2021;28:6044-50. [Crossref] [PubMed]
  29. Millen EC, Cavalcante FP, Zerwes F, et al. The Attitudes of Brazilian Breast Surgeons on Axillary Management in Early Breast Cancer-10 Years after the ACOSOG Z0011 Trial First Publication. Ann Surg Oncol 2022;29:1087-95. [Crossref] [PubMed]
  30. Barron AU, Hoskin TL, Day CN, et al. 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;153:1120-6. [Crossref] [PubMed]
  31. Samiei S, van Nijnatten TJA, de Munck L, et al. Correlation Between Pathologic Complete Response in the Breast and Absence of Axillary Lymph Node Metastases After Neoadjuvant Systemic Therapy. Ann Surg 2020;271:574-80. [Crossref] [PubMed]
  32. Tadros AB, Yang WT, Krishnamurthy S, et al. Identification of Patients With Documented Pathologic Complete Response in the Breast After Neoadjuvant Chemotherapy for Omission of Axillary Surgery. JAMA Surg 2017;152:665-70. [Crossref] [PubMed]
  33. van der Noordaa MEM, van Duijnhoven FH, Cuijpers FNE, et al. Toward omitting sentinel lymph node biopsy after neoadjuvant chemotherapy in patients with clinically node-negative breast cancer. Br J Surg 2021;108:667-74. [Crossref] [PubMed]
  34. 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;21:678-81. [Crossref] [PubMed]
  35. Reimer T, Stachs A, Nekljudova V, et al. Restricted Axillary Staging in Clinically and Sonographically Node-Negative Early Invasive Breast Cancer (c/iT1-2) in the Context of Breast Conserving Therapy: First Results Following Commencement of the Intergroup-Sentinel-Mamma (INSEMA) Trial. Geburtshilfe Frauenheilkd 2017;77:149-57. [Crossref] [PubMed]
  36. Araújo DCM, Duarte GM, Jales RM, et al. Sentinel lymph node biopsy vs no axillary surgery in early breast cancer clinically and ultrasonographically node negative: A prospective randomized controlled trial-VENUS trial. Breast J 2020;26:2087-9. [Crossref] [PubMed]
  37. Reimer T, Glass A, Botteri E, et al. 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. [Crossref] [PubMed]
  38. Kuemmel S, Heil J, Rueland A, et al. A Prospective, Multicenter Registry Study to Evaluate the Clinical Feasibility of Targeted Axillary Dissection (TAD) in Node-positive Breast Cancer Patients. Ann Surg 2022;276:e553-62. [Crossref] [PubMed]
  39. Wong SM, Basik M, Florianova L, et al. Oncologic Safety of Sentinel Lymph Node Biopsy Alone After Neoadjuvant Chemotherapy for Breast Cancer. Ann Surg Oncol 2021;28:2621-9. [Crossref] [PubMed]
  40. Kahler-Ribeiro-Fontana S, Pagan E, Magnoni F, et al. Long-term standard sentinel node biopsy after neoadjuvant treatment in breast cancer: a single institution ten-year follow-up. Eur J Surg Oncol 2021;47:804-12. [Crossref] [PubMed]
  41. Barrio AV, Montagna G, Mamtani A, et al. Nodal Recurrence in Patients With Node-Positive Breast Cancer Treated With Sentinel Node Biopsy Alone After Neoadjuvant Chemotherapy-A Rare Event. JAMA Oncol 2021;7:1851-5. [Crossref] [PubMed]
  42. Piltin MA, Hoskin TL, Day CN, et al. Oncologic Outcomes of Sentinel Lymph Node Surgery After Neoadjuvant Chemotherapy for Node-Positive Breast Cancer. Ann Surg Oncol 2020;27:4795-801. [Crossref] [PubMed]
  43. Garg AK, Buchholz TA. Influence of neoadjuvant chemotherapy on radiotherapy for breast cancer. Ann Surg Oncol 2015;22:1434-40. [Crossref] [PubMed]
  44. Giuliano AE, Ballman K, McCall L, et al. Locoregional Recurrence After Sentinel Lymph Node Dissection With or Without Axillary Dissection in Patients With Sentinel Lymph Node Metastases: Long-term Follow-up From the American College of Surgeons Oncology Group (Alliance) ACOSOG Z0011 Randomized Trial. Ann Surg 2016;264:413-20. [Crossref] [PubMed]
  45. Tinterri C, Gentile D, Gatzemeier W, 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;29:5732-44. [Crossref] [PubMed]
  46. Donker M, van Tienhoven G, Straver ME, 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;15:1303-10. [Crossref] [PubMed]
  47. Sávolt Á, Péley G, Polgár C, et al. Eight-year follow up result of the OTOASOR trial: The Optimal Treatment Of the Axilla - Surgery Or Radiotherapy after positive sentinel lymph node biopsy in early-stage breast cancer: A randomized, single centre, phase III, non-inferiority trial. Eur J Surg Oncol 2017;43:672-9. [Crossref] [PubMed]
  48. Solá M, Alberro JA, Fraile M, et al. Complete axillary lymph node dissection versus clinical follow-up in breast cancer patients with sentinel node micrometastasis: final results from the multicenter clinical trial AATRM 048/13/2000. Ann Surg Oncol 2013;20:120-7. [Crossref] [PubMed]
  49. Galimberti V, Cole BF, Zurrida S, et al. Axillary dissection versus no axillary dissection in patients with sentinel-node micrometastases (IBCSG 23-01): a phase 3 randomised controlled trial. Lancet Oncol 2013;14:297-305. Erratum in: Lancet Oncol 2013;14:e254. [Crossref] [PubMed]
  50. Mamounas EP, Anderson SJ, Dignam JJ, et al. Predictors of locoregional recurrence after neoadjuvant chemotherapy: results from combined analysis of National Surgical Adjuvant Breast and Bowel Project B-18 and B-27. J Clin Oncol 2012;30:3960-6. [Crossref] [PubMed]
  51. van Loevezijn AA, van der Noordaa MEM, Stokkel MPM, et al. Three-year follow-up of de-escalated axillary treatment after neoadjuvant systemic therapy in clinically node-positive breast cancer: the MARI-protocol. Breast Cancer Res Treat 2022;193:37-48. [Crossref] [PubMed]
  52. Ling DC, Iarrobino NA, Champ CE, et al. Regional Recurrence Rates With or Without Complete Axillary Dissection for Breast Cancer Patients with Node-Positive Disease on Sentinel Lymph Node Biopsy after Neoadjuvant Chemotherapy. Adv Radiat Oncol 2019;5:163-70. [Crossref] [PubMed]
  53. Chun JW, Kim J, Chung IY, et al. Sentinel node biopsy alone for breast cancer patients with residual nodal disease after neoadjuvant chemotherapy. Sci Rep 2021;11:9056. [Crossref] [PubMed]
  54. Almahariq MF, Levitin R, Quinn TJ, et al. Omission of Axillary Lymph Node Dissection is Associated with Inferior Survival in Breast Cancer Patients with Residual N1 Nodal Disease Following Neoadjuvant Chemotherapy. Ann Surg Oncol 2021;28:930-40. [Crossref] [PubMed]
  55. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N Engl J Med 2020;382:810-21. [Crossref] [PubMed]
  56. Loibl S, Poortmans P, Morrow M, et al. Breast cancer. Lancet 2021;397:1750-69. [Crossref] [PubMed]
  57. Burstein HJ, Curigliano G, Thürlimann B, 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;32:1216-35. [Crossref] [PubMed]
  58. Kwak HY, Chae BJ, Bae JS, et al. Feasibility of sentinel lymph node biopsy in breast cancer patients clinically suspected of axillary lymph node metastasis on preoperative imaging. World J Surg Oncol 2013;11:104. [Crossref] [PubMed]
  59. Abe H, Schacht D, Sennett CA, et al. Utility of preoperative ultrasound for predicting pN2 or higher stage axillary lymph node involvement in patients with newly diagnosed breast cancer. AJR Am J Roentgenol 2013;200:696-702. [Crossref] [PubMed]
Cite this article as: Cavalcante FP, Millen EC, Novita GG, Zerwes FP, Mattar A, Machado RHS, Frasson AL. Sentinel lymph node biopsy following neoadjuvant chemotherapy: an evidence-based review and recommendations for current practice. Chin Clin Oncol 2023;12(1):6. doi: 10.21037/cco-22-110

Download Citation