Você está na página 1de 29

Accepted Manuscript

Controversies and recommendations regarding sentinel lymph node biopsy in primary


breast cancer: a comprehensive review of current data

Petros Charalampoudis, MD PhD FRCS FEBS, Christos Markopoulos, MD MPhil PhD


FEBS, Tibor Kovacs, PhD FRCS FEBS

PII: S0748-7983(17)30955-1
DOI: 10.1016/j.ejso.2017.10.215
Reference: YEJSO 4774

To appear in: European Journal of Surgical Oncology

Received Date: 1 September 2017


Revised Date: 21 September 2017
Accepted Date: 10 October 2017

Please cite this article as: Charalampoudis P, Markopoulos C, Kovacs T, Controversies and
recommendations regarding sentinel lymph node biopsy in primary breast cancer: a comprehensive
review of current data, European Journal of Surgical Oncology (2017), doi: 10.1016/j.ejso.2017.10.215.

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to
our customers we are providing this early version of the manuscript. The manuscript will undergo
copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please
note that during the production process errors may be discovered which could affect the content, and all
legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT

Controversies and recommendations regarding sentinel lymph node biopsy in primary

breast cancer: a comprehensive review of current data.

PT
Petros Charalampoudis MD PhD FRCS FEBS1,2, Christos Markopoulos MD MPhil PhD FEBS3

and Tibor Kovacs PhD FRCS FEBS1,2

RI
SC
1
Breast Unit, Guy’s and Saint Thomas’ NHS Foundation Trust, London, UK

U
2
Division of Cancer Studies, King’s College London, UK
AN
3
Breast Unit, Athens University School of Medicine, Greece
M

For all correspondence: Petros Charalampoudis MD PhD FRCS FEBS, Consultant Oncoplastic
D

Breast Surgeon and Clinical Lecturer


TE

Breast Unit, Guy’s and Saint Thomas’ NHS Foundation Trust, Great Maze Pond, London SE1
EP

9RT, United Kingdom

petros.charalampoudis@gmail.com
C
AC

Conflicts of interest: none

1
ACCEPTED MANUSCRIPT

Abstract

In primary breast cancer, sentinel lymph node biopsy has been established as the gold standard

for regional axillary staging. A robust body of randomized data support its accuracy and safety in

PT
patients with early, clinically node negative disease. However, the role of SLNB remains

debatable in various patient subgroups, and recent advances in histopathology, dedicated axillary

RI
ultrasound imaging and chemotherapy regimens, put its role under a new perspective. Herein, we

review the current literature data on the indications for SLNB and discuss the challenges in

SC
management germane to special patient subgroups and patterns of disease. We also present

U
emerging data on the optimal management of the SLN+ patient, in light of recent trials
AN
challenging the dogma of completion axillary dissection after a positive sentinel node biopsy.
M

Introduction
D

Axillary nodal status in breast cancer patients is a paramount prognosticator, next to


TE

primary tumor size and grade. Hence, axillary staging remains the standard of care for all breast

cancers amenable to curative treatment(1,2). Sentinel lymph node biopsy (SLNB) has been
EP

established as the gold standard for axillary staging and has supplanted axillary lymph node

dissection (ALND) as a means of regional nodal staging in clinically and radiologically node-
C

negative breast cancer(3–7). SLNB provides adequate axillary nodal staging information to guide
AC

further regional and systemic treatment, whilst it spares the patient the increased risk of

complications associated with ALND, most importantly lymphedema, motor deficit and

dysesthesia of the operated arm(6,8–10). However, the appropriateness of SLNB in specific

patient subgroups, as well as the optimal further management of the positive sentinel lymph node

2
ACCEPTED MANUSCRIPT

(SLN) remain highly debatable. In fact, axillary nodal management in breast cancer still

represents one of the hottest topics of controversy across breast cancer multidisciplinary teams.

Herein, we assort the controversies and recommendations regarding SLNB in three

PT
groups. Firstly, we look at whether SLNB is always necessary in early, clinically node-negative

breast cancer and the role of preoperative axillary ultrasound during triple assessment (Group 1).

RI
Secondly, we review the issues related to the technique, feasibility and indications of SLNB in

modern breast cancer care (Group 2). Finally, we look at the current evidence and

SC
recommendations with regards to the appropriate management of the axilla following a positive

U
SLNB finding (Group 3). AN
Group 1
M

Is SLNB always necessary in early invasive breast cancer? The role of preoperative axillary
D

ultrasound.
TE

Since its conception, sentinel lymph node biopsy (SLNB) has achieved a ground-

breaking shift of practice in primary breast cancer management and has been established as the
EP

gold standard of care for upfront axillary staging in early, clinically node-negative disease. The
C

clinical value, feasibility and safety of the SLNB have been validated in a robust body of
AC

randomized controlled trials, which enrolled patients with early breast cancers and no clinically

palpable lymph nodes(3,5,7,9–11). Currently, SLNB has supplanted axillary lymph node

dissection (ALND) as the modality for primary axillary staging in early, clinically node negative

breast cancer(11). Following a negative SLNB, completion of ALND is unnecessary as it does

not add any clinical benefit (3,12,13). SLNB provides non-inferior prognostic information, is less

3
ACCEPTED MANUSCRIPT

invasive and confers less complications to the affected arm (lymphedema, motor deficit,

dysesthesia), as compared to ALND(14,15). Importantly, omission of ALND in SLNB-negative

patients does not increase the risk of axillary recurrence, nor it impacts negatively on

survival(4,6,16–18).

PT
Next to clinical palpation of the axilla, regional nodal evaluation with axillary ultrasound

RI
(AUS) has recently emerged as a useful adjunct modality to stage the axilla during triple

assessment of patients with symptoms and signs suspicious for breast cancer(19). AUS is

SC
implemented in the clinical practice guidelines across dedicated breast units as a tool of

U
significant clinical utility(20). Patients who have abnormal or suspicious nodes on AUS, i.e.
AN
nodes demonstrating loss of their fatty hilum, notable enlargement, cortical thickening or a

combination thereof, undergo AUS-guided fine needle aspiration cytology (FNAC) or core
M

biopsy (ultrasound needle biopsy - UNB). Following an AUS-guided biopsy-proven regional

spread by the primary breast cancer, the axilla is considered clinically positive and the patient is
D

routinely committed to ALND, without the need for SLNB as a staging procedure. It has been
TE

suggested that AUS and UNB represent an efficient, feasible and cost-effective staging strategy

in preoperative axillary staging(21). In an updated meta-analysis, preoperative AUS with UNB in


EP

early breast cancer had a positive predictive value (PPV) of 100%, a sensitivity of 50% and a
C

false negative rate (FNR) of 25%(22). Notably, AUS findings may be less accurate in the setting
AC

of lymphovascular invasion (LVI), and it has been suggested that a negative AUS in patients

with LVI should be interpreted with caution. SLNB remains standard of care if AUS does not

establish axillary nodal involvement (23,24).

Although the ACOSOG Z11 trial(25) preceded the AUS staging studies, it unavoidably

called into question the clinical value of AUS and UNB in preoperative axillary staging. In the

4
ACCEPTED MANUSCRIPT

‘post-Z11’ era (25,26), not all patients with a positive macrometastatic sentinel lymph node

(SLN) require a completion ALND. Can AUS and UNB reliably identify cases that fall outside

the Z11-eligibility group? Breast cancer care teams who have adopted Z11 may find preoperative

AUS and UNB of uncertain utility. The ability of AUS and UNB to quantify the axillary nodal

PT
burden and consequently identify Z11-(in)eligible patients has been challenged(21). On one

RI
hand, AUS-guided biopsy-proven node-positive patients have been found to carry significantly

higher positive axillary nodal burden in the axilla, compared with SLNB-positive patients. Farrel

SC
and colleagues showed that a single nodal metastasis detected by UNB correlated with a mean of

5.2 nodes on final histopathology after ALND(26). Furthermore, Hieken and colleagues

U
demonstrated that up to 66% of T1 and T2 UNB-positive patients with multiple suspicious
AN
lymph nodes on AUS had >2 positive lymph nodes after axillary surgery, suggesting that a

UNB-positive axillary nodal basin falls outside the Z11 criteria(27). Similarly, Boland and
M

colleagues showed that AUS and UNB can reliably identify Z11-ineligible patients(28).
D

Conversely, in a recent meta-analysis of six studies reporting on the axillary nodal burden of
TE

ultrasound-positive versus ultrasound-negative patients, it was suggested that nearly half of the

ultrasound-positive patients could carry axillary nodal burden low enough to be considered for
EP

omission of ALND (‘Z11 eligible’)(29). Recent data suggest that streamlining axillary US

guided FNA+ patients to ALND may be overtreatment.(30,31). A safe and accurate approach to
C

direct ultrasound node-positive patients towards the appropriate further axillary surgical
AC

management (ALND versus no ALND) is yet to be defined.

The findings of the ACOSOG Z11 randomized controlled trial suggested that completion

ALND in select patients who undergo breast conserving surgery, whole breast radiotherapy and

tailored adjuvant systemic treatment, found to have one or two sentinel macrometastases, does

5
ACCEPTED MANUSCRIPT

not improve survival or the risk of axillary failure during a median follow-up of 9.25

years(25,32). The concept that axillary nodal extirpation by means of ALND may not

significantly impact on patient outcomes is not new. In the NSABAP-04 study, clinically node-

negative patients proceeded to either total mastectomy alone with delayed ALND in case of later

PT
nodal recurrence or total mastectomy with concurrent ALND. Interestingly, the subsequent

RI
failure rate in the total mastectomy-alone group was only about half (19%) that predicted from

the proportion of patients with axillary metastases that underwent ALND (40%) (27,28).

SC
Furthermore, in contemporary multidisciplinary breast cancer care, prognostic information

derived from the number of involved axillary lymph nodes is less likely to impact on systemic

U
treatment decision-making, as opposed to patient characteristics and tumor molecular profiling.
AN
Isolated axillary relapse, even in cases of previously SLNB-positive disease is remarkably low

(around 1%)(16). In this direction, the very necessity of axillary nodal staging in early breast
M

cancer has been challenged, particularly for small, low grade, favorable breast cancers, which are
D

less likely to metastasize regionally. Looking at this challenging hypothesis, the SOUND trial
TE

has been designed to investigate whether omission of primary axillary staging with SLNB in

early, clinically and radiologically node-negative breast cancer will have an impact on patient
EP

outcomes, as compared with standard SLNB. The trial closed accrual in January 2017 and its

results are awaited with great interest(33).


C
AC

Summary of recommendations and controversies (Group 1)

• In primary operable breast cancer, SLNB is standard of care in patients with a clinically

and radiologically negative axilla. ALND is no longer recommended in these patients, as

it only adds to arm morbidity without conferring any prognostic or staging benefit(11).

6
ACCEPTED MANUSCRIPT

• During triage of patients with newly diagnosed breast cancer, axillary ultrasound with

biopsy of the abnormal lymph node(s) can fast-track patients to ALND without upfront

SLNB, in cases where nodal biopsy is positive for regional metastasis. Although several

studies have correlated a US-guided biopsy-proven positive axilla with a higher nodal

PT
burden at completion ALND, as compared with the US-negative axilla, a significant

RI
subgroup of patients with UNB+ axillary node(s) may have only one or two involved

nodes in the ALND specimen (‘Z11 eligible’). Therefore, there is a need for establishing

SC
an approach to properly identify UNB+ patients in which ALND would be

overtreatment(20,22,29).


U
Complete omission of SLNB in early, small, favorable breast cancers is under
AN
investigation, provided that a clinical and ultrasound axillary follow-up proves non-
M

inferior in patients with clinically node negative breast cancers at presentation(33).


D
TE

Group 2

SLNB in various clinical scenarios


EP

Repeat SLNB after previous SLNB


C

Data on repeat SLNB after previous SLNB are scarce, however they suggest that SLNB
AC

may be feasible after previous limited axillary staging. Albeit, repeat SLNB is likely to yield

lower identification rates, compared to standard primary SLNB in the previously non-operated

axilla. Kothari and colleagues reviewed a total of 6 studies encompassing 327 patients

undergoing SLNB, of which 60% had previous SLNB or limited axillary staging. The median

overall repeat sentinel identification rate was 70%. No axillary recurrences were observed in

7
ACCEPTED MANUSCRIPT

patients who had a negative repeat SLNB at a follow-up of maximum 46 months(34). In a

systematic review and meta-analysis, Maaskant-Brant and colleagues looked at patients who

underwent repeat SLNB for ipsilateral breast cancer local recurrence. The authors reviewed 692

patients who either had previous SLNB (301/692), or previous ALND (361/692), or no previous

PT
axillary surgery (30/692). The medial overall identification rate was 65% across all 692 cases,

RI
but repeat SLNB had a higher identification rate in patients who had previous SLNB, as opposed

to those who had previous ALND(35). At present, repeat SLNB can be considered in patients

SC
who had previous SLNB or limited axillary sampling, but patients must be informed of the

higher rate of SLNB identification failure and therefore should be consented to proceed to

ALND in case an SLN cannot be retrieved(36).


U
AN
M

SLNB in large tumors and locally advanced or inflammatory breast cancer


D

The main body of high-level evidence studies regarding the feasibility of SLNB, looked
TE

at patients with T1 and T2 cancers with a clinically node-negative axilla. The updated ASCO

guidelines in 2016 recommend against SLNB in T3 breast cancers (level of recommendation:


EP

weak, type: informal consensus)(37). However, data from non-randomized studies suggest that

SLNB is accurate in patients with T3 cancers and a clinically negative axilla(38,39). In most
C

breast cancer centers, SLNB is considered acceptable in cT3N0 patients. In locally advanced
AC

(LABC) or inflammatory breast cancer (IBC), upfront SLNB is not routinely recommended,

based on the hypothesis that locally advanced or inflammatory changes may lead to partial

obstruction of dermal or parenchymal lymphatics and hence yield unacceptable FNRs of sentinel

node retrieval(37,40). In patients with non-inflammatory LABC who have excellent clinical and

radiological response to neoadjuvant chemotherapy (NACT), SLNB instead of ALND may be

8
ACCEPTED MANUSCRIPT

considered and is moderately recommended by the most recent edition of the NCCN

guidelines(41). However, in order to increase sensitivity and accuracy of the procedure, at least 3

SLNs should be retrieved and complete ALND is advised even if only one SLN is found to be

positive(56-60).

PT
SLNB in pregnancy

RI
The ASCO updated guideline in 2016 remained unchanged regarding SLNB in pregnant

SC
patients. High-level evidence is lacking and therefore, the panel weakly recommended against

SLNB in this setting(37). However, data from non-randomized studies suggest that SLNB with

U
radioisotope only (no blue dye) is safe in pregnant breast cancer patients(42–44). Sentinel lymph
AN
node biopsy involves a technetium colloid injection, which demonstrates low radioactivity and

limited local uptake with a rapid wash-out, as well as negligible systemic diffusion(45).
M

Furthermore, radioisotope doses absorbed by the fetus have been shown to be less than 20
D

micrograys (µGy) for injections up to 20 megabequerels (Mbq). Conversely, blue dye injection is

not recommended in pregnancy, as it carries a low (1%) but potentially harmful risk of
TE

anaphylactic maternal reaction(46,47). In conclusion, SLNB with omission of blue dye injection
EP

may be considered in pregnant patients.


C

SLNB after neoadjuvant chemotherapy (cN0 patients at diagnosis)


AC

Neoadjuvant chemotherapy (NACT) is being increasingly recommended in patients with

breast cancer(48). Frequent indications for NACT include tumor downsizing to optimize breast

conservation, conversion of large unifocal breast cancers needing a mastectomy to cancers

amenable to breast conserving surgery (BCS) and conversion of large inoperable, locally

9
ACCEPTED MANUSCRIPT

advanced or inflammatory cancers to operable tumors(49). It has been suggested that

pathological complete response to NACT may serve as a good surrogate for disease-free

survival. Also, NACT provides useful information on responses to different chemotherapy

regiments. However, a survival benefit between patients receiving neoadjuvant compared to

PT
adjuvant chemotherapy has not been demonstrated to date(50).

RI
SLNB has been established as a valid modality of axillary staging in trials enrolling

patients who were receiving upfront surgery, followed by tailored adjuvant systemic treatment.

SC
The concept of SLNB instead of ALND to stage patients receiving neoadjuvant chemotherapy

U
was initially met with reluctance(51). There was concern that lymphatic tumor emboli and
AN
chemotherapy-induced fibrosis may cause a non-uniform response across the axillary nodal basin

and potentially lead to unacceptably high SLNB false negative rates. Indeed, early studies on
M

SLNB after NACT for node-negative patients (cN0) reported on identification rates ranging from

63% to 100%, and FNRs ranging from 0% to 33%(52,53). However, these studies included
D

various clinical stages and cases with bulky nodal disease at presentation, which persisted after
TE

completion of NACT. Subsequent studies showed that SLNB after NACT in node-negative

patients is feasible and FNR is comparable to pre-chemotherapy SLNB. The GANEA study
EP

looked at 130 clinically node-negative patients to assess the feasibility of SLNB following
C

NACT. Among the 130 patients with cN0 disease, the SLN identification rate was 95% with an
AC

FNR of 9%(54). A study from MD Anderson Cancer Center looked at 3171 cN0 patients

undergoing upfront SLNB and 575 cN0 patients who underwent SLNB following NACT.

Importantly, both identification rates and false negative rates were similar between pre-

chemotherapy and post-chemotherapy SLNB (identification rate 99% and 97%, FNR 4% and

6%, respectively)(55). Furthermore, data from meta-analyses including more than 5000 patients

10
ACCEPTED MANUSCRIPT

treated with SLNB after NACT have reported on SLN identification rates ranging from 90% to

94% and FNRs ranging from 7% to 12%(56,57). At present, SLNB in clinically node-negative

breast cancer at diagnosis is recommended after NACT, provided that the end-of-chemotherapy

clinical palpation and imaging suggest no evidence of overall progression and the nodes remain

PT
clinically normal. Trials such as Alliance A011202 are ongoing to further address the need for

RI
ALND in SLN+ patients after NACT(58).

SC
SLNB after neoadjuvant chemotherapy (cN+ patients at diagnosis)

U
Interestingly, studies on SLNB after NACT in cN0 patients demonstrated that post-
AN
NACT axillary staging yielded less nodal burden as compared to pre-chemotherapy axillary

staging (54). Moreover, increasing rates of pathological complete response have been observed
M

in patients treated with NACT(59). These findings led investigators to design studies looking at
D

the feasibility of SLNB after NACT in patients who presented with cN+ disease at diagnosis.
TE

The GANEA study looked at 65 patients with cN+ patients who underwent SLNB and back-up

ALND after NACT. The SLNB identification rate was 81.5% and the FNR was 15%(54). In the
EP

SENTINA trial, 592 patients converting from cN+ to cN0 following NACT had an SLN

identification rate of 80%, with an overall FNR of 14%. However, the FNR in the subgroup
C

where 3 or more sentinel nodes were harvested dropped to 7%, compared with 19% with 2 nodes
AC

harvested and 24% with only 1 SLN harvested(60). The ACOSOG Z1071 trial also looked at

patients with cN+ breast cancers at diagnosis who underwent SLNB after NACT. In this trial, a

pre-defined acceptable FNR was set to 10%. Unlike the SENTINA trial, conversion from cN+ to

cN0 was not mandatory and only patients where more than 2 harvested SLNs were included in

the analysis to calculate the FNR of the procedure. The overall FNR for patients who had at least

11
ACCEPTED MANUSCRIPT

2 SLNs retrieved was 13%, but an unplanned exploratory analysis showed that the FNR

significantly dropped to 9% when 3 or more SLNs were harvested(61). Similarly, the SN-FNAC

study looked at biopsy proven cN+ patients who received SLNB after NACT. Unlikely the

SENTINA and ACOSOG Z1071 trials, immunohistochemistry (IHC) to detect nodal disease

PT
down to isolated tumor cells (ITCs) was mandatory in the SN-FNAC trial. In their results, the

RI
investigators report on an FNR of 8% provided that patients with ITC post-NACT were

considered node-positive, whereas the FNR was 13% when these patients were considered node-

SC
negative(62). Lastly, a Swedish group reported on 195 biopsy-proven node positive breast cancer

patients who underwent SLNB after NACT. The overall FNR in the study was 14% but dropped

U
to 4% when 2 or more SLNs were harvested (58).
AN
In summary, four studies have reported on SLNB after NACT in cN+ patients at
M

diagnosis. They all demonstrate that SLNB is technically feasible provided a good clinical and

radiological response is achieved after NACT. A very important common finding of these studies
D

is that FNRs are unacceptably high unless at least 3 SLNs are identified(60–63). The questions
TE

which remain unanswered are what would be the long-term outcome of cN+ patients having a

negative SLNB (with more than 3 nodes harvested) after NACT, if a completion ALND was
EP

omitted, and whether postmastectomy radiotherapy adds any benefit to cN+ patients who revert
C

to ypN0 after NACT(64). All four studies required a back-up ALND to calculate the FNR of the
AC

SLNB and therefore cannot address this question. At present, SLNB after NACT for cN+

patients is being considered across dedicated breast units and is implemented in the last edition

of the NCCN guidelines as a Level 2b recommendation(41). Pre-NACT clipping or radioactive

seed marking of the biopsy-proven nodes, which are harvested during post-NACT axillary

staging along with the sentinel nodes, is currently under investigation, aiming to provide more

12
ACCEPTED MANUSCRIPT

robust evidence of nodal response to chemotherapy and improve the false negative rates of the

procedure(65–67).

PT
SLNB in DCIS

RI
Ductal breast cancer in situ (DCIS) makes up 20% of all newly diagnosed breast cancers,

and by definition, does not have the potential to spread(68). Axillary failure in patients with

SC
DCIS is rare, as demonstrated in early trials. In the NSABP-B17 trial, only 3/620 of patients with

DCIS recurred in the axilla at 15 years(69). Similarly, in the NSABP-B24 trial, axillary failure

U
events were very infrequent (6/1799 at 12 years)(69). It has been suggested that approximately
AN
20% of DCIS diagnosed on preoperative core biopsy will upgrade to invasive disease on final

histopathology after surgery (70,71). The frequency of axillary metastasis in patients who
M

undergo mastectomy for DCIS has been reported as high as 20%(72). In a meta-analysis by
D

Ansari and colleagues, the incidence of sentinel node metastases in patients with a preoperative
TE

diagnosis of DCIS was 7.4%, compared with 3.7% in patients with a definitive postoperative

diagnosis of pure DCIS(73). In patients with DCIS amenable to breast conserving surgery,
EP

SLNB is not recommended, unless DCIS presents with features suggestive of higher risk of

invasion (palpable mass, suspicious imaging features, large size)(74,75). Conversely, in patients
C

needing mastectomy for extensive DCIS, the risk of non-sampled invasive breast cancer is more
AC

substantive. Additionally, following a mastectomy, the parenchymal lymphatics will have been

extirpated and hence, SLNB as a subsequent procedure carries a higher risk of failure to identify

the sentinel nodes(76). The clinical significance of nodal involvement in pure DCIS remains

unclear(77,78). In summary, although randomized data are not available, SLNB for DCIS should

13
ACCEPTED MANUSCRIPT

be considered in breast-conserving surgery for high-risk DCIS and in patients undergoing

mastectomy for extensive DCIS.

PT
SLNB in multisite BC (multifocal or multicentric)

RI
In multisite breast cancer, it has been suggested that the accuracy of SLNB may be

compromised, as drainage from different primaries in the same breast may complicate the

SC
sentinel node identification and thus result in suboptimal axillary staging. Data on SLNB in

multisite breast cancer are only available from observational studies. Interestingly, no difference

U
in axillary recurrence or survival are noted across these cohorts, when compared with patients
AN
with unifocal disease(79,80). In an unplanned subgroup analysis of the ALMANAC trial, SLNB

in cases of multisite breast cancer did not result in lower FNR or lower identification rates,
M

compared to SLNB performed for unifocal lesions(3). In the AMAROS trial, a SLN was
D

identified successfully in 96% of patients with multisite breast cancer as compared with a 98%
TE

rate in patients with unifocal breast cancer. This difference was not statistically

significant(81,82). At present, SLNB is considered feasible and safe in patients with early,
EP

multisite breast cancer.


C
AC

Summary of recommendations and controversies (Group 2)

• Repeat SLNB after previous SLNB for locally recurrent breast cancer may be considered,

although the false negative rate of repeat axillary staging may be higher than upfront

SLNB in the non-operated axilla. ALND may be needed in these patients in cases where

a sentinel node is not identified(35,36).

14
ACCEPTED MANUSCRIPT

• SLNB with single tracer (radioisotope) is considered safe in pregnant patients. Blue dye

is not recommended due to the small but clinically important risk of maternal

anaphylactic reaction(43,44,46)

• Upfront SLNB in locally advanced or inflammatory breast cancer is not recommended.

PT
SLNB after neoadjuvant chemotherapy in non-inflammatory LABC may be considered,

RI
on the basis of overall and axillary response to neoadjuvant treatment(37,41).

• SLNB is safe and accurate in patients with cN0 breast cancers after neoadjuvant

SC
chemotherapy. In patients with cN+ disease at diagnosis, SLNB may be considered

(Level of evidence 2a) when radiological and clinical complete response is achieved after

U
NACT. There are now four studies supporting this practice, but the long-term outcome of
AN
these patients is yet undefined(60–63).


M

SLNB in multifocal and multicentric breast cancer is deemed safe and accurate, although

randomized data are lacking(80).


D

• SLNB in DCIS patients undergoing breast conservation may be considered for large,
TE

high-risk DCIS, as well as in patients treated by mastectomy on the basis of extensive

DCIS, which portends a higher risk of non-sampled invasive disease(73).


C EP

Group 3
AC

Management of the SLN-positive axilla

Robust evidence suggests that following a negative SLNB, completion ALND is not

necessary, as it only adds on regional morbidity without conferring any clinical benefit in terms

of recurrence risk, disease-free survival or overall survival(11). Axillary local recurrence after a

15
ACCEPTED MANUSCRIPT

negative SLNB is rare (16,25). With the advent of modern histopathology and improvements in

staining and immunohistochemistry techniques (IHC), even small metastatic clusters of regional

nodal spread can be identified, leading to coining of the terms ‘isolated tumor cells, ITCs’ (nodal

foci <0.2mm), ‘micrometastases’ (foci 0.2mm-2mm) and ‘macrometastases’ (foci >2mm).

PT
Consequently, the clinical significance of axillary nodal burden on the basis of size of nodal foci

RI
has been evaluated.

Although isolated tumor cells (ITCs) can be detected on routine hematoxylin and eosin

SC
(H&E) stains, they are usually found on deeper, serial H&E levels or typically during IHC

U
staining. However, routine use of IHC to look for ITCs in the SLNs is not supported(83,84). In
AN
the presence of ITCs in the SLNs, only a small percentage of non-SLNs are involved by residual

disease and the risk of axillary recurrence in cN0(i+) patients who forgo ALND is very
M

low(85,86). According to the updated AJCC-TNM edition for breast cancer, patients with

pN0(i+) only disease (ITCs only) are considered clinically node-negative(41). In patients with
D

ITCs only in the SLNs, completion ALND may be omitted without negatively impacting on their
TE

outcome(85,87). Albeit, the presence of ITCs carries less favorable prognosis than pN0 disease

and tailored systemic treatment may be appropriate(88–90). At present, omission of ALND in


EP

N0(i+) patients is considered when upfront surgery is recommended. The presence of residual
C

ITCs in post-NACT SLNB in patients with clinically positive nodes who have received
AC

neoadjuvant chemotherapy may be of higher clinical significance, compared with ITCs found on

upfront SLNB, as the latter could reflect residual, chemoresistant disease. Interestingly, a recent

study from the Netherlands showed that baseline cN1 patients who had ITCs only found on post-

NACT ALND carried similar prognosis compared with ypN0 patients (where no residual disease

was identified in the ALND specimen)(91).

16
ACCEPTED MANUSCRIPT

Nodal foci with size from 0.2mm to 2mm are termed ‘micrometastases’. The randomized

controlled trial by Galimberti and colleagues challenged the dogma of standard completion

ALND in patients with micrometastatic senitinel involvement. This trial randomized patients

undergoing breast surgery and found to have micrometastases only in the SLN, to undergo either

PT
completion ALND or no further surgical axillary treatment(77). Axillary radiotherapy was not an

RI
alternative to ALND in this study and most patients in both groups received adjuvant systemic

therapy at the discretion of the treating clinician. Completion ALND in patients with sentinel

SC
micrometastases was not found to confer statistically significant benefit in disease-free or overall

survival, compared to no further axillary surgery. Although patients with micrometastases are

U
considered lymph node-positive (as opposed to ITC only+ patients), this trial provided high-level
AN
evidence that surgical extirpation of the axillary nodal basin following micrometastatic SLNB

does not confer clinical benefit. Sola and colleagues independently confirmed the findings by
M

Galimberti and colleagues, albeit recruiting less patients.(92)


D

After a SLNB positive for macrometastases, the standard of care has long been a
TE

completion ALND. However, three randomized controlled trials (ACOSOG Z11, EORTC-

AMAROS, OTOASOR) challenged the dogma of completion ALND in select patients with SLN
EP

macrometastases(25,81,93). In the Z11 trial, patients with T1 and T2 breast cancers and one or
C

two sentinel macrometastases, who underwent breast conserving surgery without completion
AC

ALND, followed by whole breast radiotherapy and appropriate adjuvant systemic treatment, had

similar axillary failure event rates and survival outcomes up to 9.25 years of follow-up, as

compared with patients who underwent completion ALND. The cumulative incidence of axillary

recurrence at 10 years was 0.5% in the ALND arm and 1.5% in the SLND alone arm (p = 0.28).

Ten-year cumulative locoregional recurrence was 6.2% with ALND and 5.3% with SLND alone

17
ACCEPTED MANUSCRIPT

(p = 0.36)(25,32). Importantly, in the SLNB+ALND arm of the trial, only about one out of four

SLN-positive patients were found to have residual disease in the completion ALND specimen.

This finding suggested that SLNB provides adequate local control next to its staging role, whilst

it spares the patient the long-term sequelae of ALND (lymphedema, sensory loss and motor

PT
deficit of the affected arm), without impacting negatively on survival or regional axillary

RI
recurrence. Although the Z11 trial was criticized (lack of homogeneity in the whole breast

radiotherapy tangents, early accrual closure, too many ER-positive patients), it remains a very

SC
well-designed trial and is largely considered as practice-changing. The AMAROS trial enrolled a

patient population similar to the Z11 (T1-2 breast cancers, clinically node negative, undergoing

U
breast conservation with whole breast radiotherapy). In the test arm of the study, axillary
AN
radiotherapy (as opposed to no further regional treatment in the Z11) was offered to patients after

one or two positive (macrometastatic) nodes found during the SLNB. Axillary radiotherapy with
M

omission of ALND conferred similar survival and recurrence outcomes as compared with
D

standard completion ALND. Also, lymphedema rates in the axillary radiotherapy arm were
TE

significantly reduced compared to the ALND arm(81). Similar in design, the OTOASOR trial

independently confirmed the AMAROS findings. Amongst patients with early breast cancer and
EP

low SLNB burden, axillary nodal irradiation was non inferior to completion ALND in terms of

survival and regional recurrence(93).


C
AC

The Z11 trial enrolled patients undergoing breast conserving surgery with whole breast

radiotherapy(25). In the trial by Galimberti and colleages, some 9% of patients in each arm

underwent mastectomy. An unplanned subgroup analysis suggested that omission of ALND may

be acceptable in patients with small invasive primary tumors treated by mastectomy(94).

Notably, in the same trial, 19% of patients in the non-ALND arm underwent breast conserving

18
ACCEPTED MANUSCRIPT

surgery with partial breast irradiation as opposed to whole breast tangents(94). However, there

was no subgroup analysis to look at whether patients who received partial breast irradiation

exhibited any outcome detriment compared with those who received standard whole breast

radiotherapy.. Clinicians should be cautious when extrapolating outcomes from the Z11 trial to

PT
patients who undergo mastectomy or receive partial breast irradiation. In the Z11 trial, one

RI
interpretation of the similarity in outcomes between the ALND and non-ALND groups may be

that in (at least) a proportion of patients, a low axillary tangent (as part of the whole breast

SC
irradiation field) treated partially the axilla. However, a dedicated axillary irradiation field was

not offered in the Z11, as opposed to the AMAROS trial where targeted axillary irradiation was

U
part of the treatment protocol in the test arm. The randomized controlled POSNOC trial is
AN
currently recruiting patients with up to two positive sentinel macrometastases who undergo either

breast conservation or mastectomy, looking at whether omission of ALND in the test group
M

confers similar survival and axillary recurrence outcomes compared with the standard ALND
D

group. The POSNOC trial will therefore attempt to partially resolve the question whether the
TE

Z11 outcomes are reproducible to patients undergoing mastectomy(95).


EP

Summary of recommendations (Group 3)


C

• Completion ALND in patients with sentinel node isolated tumor cells is no longer
AC

recommended. However, pN0(i+) disease confers inferior outcomes compared with pN0

disease and therefore adjuvant chemotherapy may be considered in these patients(88).

• Data from two randomized controlled trials support that ALND is not of clinical benefit

in patients with sentinel micrometastases(92,94).

19
ACCEPTED MANUSCRIPT

• In patients with one or two sentinel macrometastases, completion ALND is non-inferior

to no further axillary treatment, in cases where breast conserving surgery with whole

breast irradiation and appropriate adjuvant systemic therapy is planned. Dedicated

axillary radiotherapy is considered equivalent to completion ALND in these patients, and

PT
axillary radiotherapy confers lower risk of lymphoedema compared to ALND(25,81,93).

RI
• In patients with one or two sentinel node macrometastases undergoing mastectomy, the

findings of the Z11 trial should be extrapolated with caution. The POSNOC trial aims to

SC
address the question of whether ALND is non-inferior to no further axillary surgery in

patients with limited SLN burden undergoing mastectomy(95).

U
AN
Conclusion
M

SLNB has well established indications and remains an operation with minimal morbidity, high
D

accuracy, and clinically valid prognostic significance. Recent advances in multimodality therapy
TE

and dedicated axillary imaging highlight the potential for de-escalating axillary surgery and

SLNB has a cardinal role in such an approach. Furthermore, non-randomized data suggest that
EP

the indication for SLNB may be expanded in patients where standard treatment would include a

complete axillary node dissection (locally advanced and multisite tumors). Clinicians need to be
C

well aware on the recent data supporting the use of SLNB in controversial settings, such as node-
AC

positive patients who respond excellently after neoadjuvant chemotherapy and patients who have

minimal sentinel node involvement.

20
ACCEPTED MANUSCRIPT

References

1. Abrams JS. Adjuvant therapy for breast cancer--results from the USA consensus conference. Breast
Cancer Tokyo Jpn. 2001;8(4):298–304.

2. Samphao S, Eremin JM, El-Sheemy M, Eremin O. Management of the axilla in women with breast
cancer: current clinical practice and a new selective targeted approach. Ann Surg Oncol. 2008

PT
May;15(5):1282–96.

3. Mansel RE, Fallowfield L, Kissin M, Goyal A, Newcombe RG, Dixon JM, et al. Randomized

RI
multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast
cancer: the ALMANAC Trial. J Natl Cancer Inst. 2006 May 3;98(9):599–609.

SC
4. Zavagno G, De Salvo GL, Scalco G, Bozza F, Barutta L, Del Bianco P, et al. A Randomized clinical trial
on sentinel lymph node biopsy versus axillary lymph node dissection in breast cancer: results of
the Sentinella/GIVOM trial. Ann Surg. 2008 Feb;247(2):207–13.

U
5. 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
AN
of a Randomized Clinical Trial. Ann Surg Oncol. 2016 Aug;23(8):2494–500.

6. Veronesi U, Paganelli G, Viale G, Luini A, Zurrida S, Galimberti V, et al. A randomized comparison of


sentinel-node biopsy with routine axillary dissection in breast cancer. N Engl J Med. 2003 Aug
M

7;349(6):546–53.

7. Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Costantino JP, et al. Sentinel-lymph-node
D

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 Oct;11(10):927–33.
TE

8. Krag DN. Minimal access surgery for staging regional lymph nodes: the sentinel-node concept. Curr
Probl Surg. 1998 Nov;35(11):951–1016.
EP

9. Krag D, Weaver D, Ashikaga T, Moffat F, Klimberg VS, Shriver C, et al. The sentinel node in breast
cancer--a multicenter validation study. N Engl J Med. 1998 Oct 1;339(14):941–6.
C

10. Veronesi U, Paganelli G, Galimberti V, Viale G, Zurrida S, Bedoni M, et al. Sentinel-node biopsy to
avoid axillary dissection in breast cancer with clinically negative lymph-nodes. Lancet Lond Engl.
AC

1997 Jun 28;349(9069):1864–7.

11. Bromham N, Schmidt-Hansen M, Astin M, Hasler E, Reed MW. Axillary treatment for operable
primary breast cancer. Cochrane Database Syst Rev. 2017 04;1:CD004561.

12. Kell MR, Burke JP, Barry M, Morrow M. Outcome of axillary staging in early breast cancer: a meta-
analysis. Breast Cancer Res Treat. 2010 Apr;120(2):441–7.

21
ACCEPTED MANUSCRIPT

13. Wang Z, Wu L-C, Chen J-Q. Sentinel lymph node biopsy compared with axillary lymph node
dissection in early breast cancer: a meta-analysis. Breast Cancer Res Treat. 2011 Oct;129(3):675–
89.

14. Fleissig A, Fallowfield LJ, Langridge CI, Johnson L, Newcombe RG, Dixon JM, et al. Post-operative
arm morbidity and quality of life. Results of the ALMANAC randomised trial comparing sentinel
node biopsy with standard axillary treatment in the management of patients with early breast

PT
cancer. Breast Cancer Res Treat. 2006 Feb;95(3):279–93.

15. Del Bianco P, Zavagno G, Burelli P, Scalco G, Barutta L, Carraro P, et al. Morbidity comparison of
sentinel lymph node biopsy versus conventional axillary lymph node dissection for breast cancer

RI
patients: results of the sentinella-GIVOM Italian randomised clinical trial. Eur J Surg Oncol J Eur Soc
Surg Oncol Br Assoc Surg Oncol. 2008 May;34(5):508–13.

SC
16. van der Ploeg IMC, Nieweg OE, van Rijk MC, Valdés Olmos RA, Kroon BBR. 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 J Eur Soc Surg Oncol Br Assoc Surg Oncol. 2008
Dec;34(12):1277–84.

17.
U
Canavese G, Catturich A, Vecchio C, Tomei D, Gipponi M, Villa G, et al. Sentinel node biopsy
AN
compared with complete axillary dissection for staging early breast cancer with clinically negative
lymph nodes: results of randomized trial. Ann Oncol Off J Eur Soc Med Oncol. 2009
Jun;20(6):1001–7.
M

18. Krag DN, Anderson SJ, Julian TB, Brown AM, Harlow SP, Ashikaga T, 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.
D

Lancet Oncol. 2007 Oct;8(10):881–8.


TE

19. Houssami N, Turner RM. Staging the axilla in women with breast cancer: the utility of preoperative
ultrasound-guided needle biopsy. Cancer Biol Med. 2014 Jun;11(2):69–77.

20. Houssami N, Diepstraten SCE, Cody HS, Turner RM, Sever AR. Clinical utility of ultrasound-needle
EP

biopsy for preoperative staging of the axilla in invasive breast cancer. Anticancer Res. 2014
Mar;34(3):1087–97.

21. Schipper RJ, van Roozendaal LM, de Vries B, Pijnappel RM, Beets-Tan RGH, Lobbes MBI, et al.
C

Axillary ultrasound for preoperative nodal staging in breast cancer patients: is it of added value?
Breast Edinb Scotl. 2013 Dec;22(6):1108–13.
AC

22. Houssami N, Ciatto S, Turner RM, Cody HS, Macaskill P. Preoperative ultrasound-guided needle
biopsy of axillary nodes in invasive breast cancer: meta-analysis of its accuracy and utility in staging
the axilla. Ann Surg. 2011 Aug;254(2):243–51.

23. Tucker NS, Cyr AE, Ademuyiwa FO, Tabchy A, George K, Sharma PK, et al. Axillary Ultrasound
Accurately Excludes Clinically Significant Lymph Node Disease in Patients With Early Stage Breast
Cancer. Ann Surg. 2016 Dec;264(6):1098–102.

22
ACCEPTED MANUSCRIPT

24. Nwaogu IY, Yan Y, Appleton CM, Cyr AE, Margenthaler JA. Predictors of false negative axillary
ultrasound in breast cancer. J Surg Res. 2015 Oct;198(2):351–4.

25. Giuliano AE, Hunt KK, Ballman KV, Beitsch PD, Whitworth PW, Blumencranz PW, et al. Axillary
dissection vs no axillary dissection in women with invasive breast cancer and sentinel node
metastasis: a randomized clinical trial. JAMA. 2011 Feb 9;305(6):569–75.

PT
26. Farrell TPJ, Adams NC, Stenson M, Carroll PA, Griffin M, Connolly EM, et al. The Z0011 Trial: Is this
the end of axillary ultrasound in the pre-operative assessment of breast cancer patients? Eur
Radiol. 2015 Sep;25(9):2682–7.

RI
27. Hieken TJ, Trull BC, Boughey JC, Jones KN, Reynolds CA, Shah SS, et al. Preoperative axillary
imaging with percutaneous lymph node biopsy is valuable in the contemporary management of
patients with breast cancer. Surgery. 2013 Oct;154(4):831-838-840.

SC
28. Boland MR, Prichard RS, Daskalova I, Lowery AJ, Evoy D, Geraghty J, et al. Axillary nodal burden in
primary breast cancer patients with positive pre-operative ultrasound guided fine needle
aspiration cytology: management in the era of ACOSOG Z011. Eur J Surg Oncol J Eur Soc Surg Oncol

U
Br Assoc Surg Oncol. 2015 Apr;41(4):559–65.
AN
29. Ahmed M, Jozsa F, Baker R, Rubio IT, Benson J, Douek M. Meta-analysis of tumour burden in pre-
operative axillary ultrasound positive and negative breast cancer patients. Breast Cancer Res Treat.
2017 Jul 28;
M

30. Pilewskie M, Mautner SK, Stempel M, Eaton A, Morrow M. Does a Positive Axillary Lymph Node
Needle Biopsy Result Predict the Need for an Axillary Lymph Node Dissection in Clinically Node-
Negative Breast Cancer Patients in the ACOSOG Z0011 Era? Ann Surg Oncol. 2016 Apr;23(4):1123–
D

8.
TE

31. Pilewskie M, Jochelson M, Gooch JC, Patil S, Stempel M, Morrow M. Is Preoperative Axillary
Imaging Beneficial in Identifying Clinically Node-Negative Patients Requiring Axillary Lymph Node
Dissection? J Am Coll Surg. 2016 Feb;222(2):138–45.
EP

32. Giuliano AE, Ballman K, McCall L, Beitsch P, Whitworth PW, Blumencranz P, 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
C

Sep;264(3):413–20.
AC

33. 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 Edinb Scotl. 2012 Oct;21(5):678–81.

34. Kothari MS, Rusby JE, Agusti AA, MacNeill FA. Sentinel lymph node biopsy after previous axillary
surgery: A review. Eur J Surg Oncol J Eur Soc Surg Oncol Br Assoc Surg Oncol. 2012 Jan;38(1):8–15.

35. Maaskant-Braat AJG, Voogd AC, Roumen RMH, Nieuwenhuijzen GAP. Repeat sentinel node biopsy
in patients with locally recurrent breast cancer: a systematic review and meta-analysis of the
literature. Breast Cancer Res Treat. 2013 Feb;138(1):13–20.

23
ACCEPTED MANUSCRIPT

36. Vugts G, Maaskant-Braat AJG, Voogd AC, van Riet YEA, Luiten EJT, Rutgers EJT, et al. Repeat
sentinel node biopsy should be considered in patients with locally recurrent breast cancer. Breast
Cancer Res Treat. 2015 Oct;153(3):549–56.

37. Lyman GH, Somerfield MR, Bosserman LD, Perkins CL, Weaver DL, Giuliano AE. Sentinel Lymph
Node Biopsy for Patients With Early-Stage Breast Cancer: American Society of Clinical Oncology
Clinical Practice Guideline Update. J Clin Oncol Off J Am Soc Clin Oncol. 2016 Dec

PT
12;JCO2016710947.

38. Chung MH, Ye W, Giuliano AE. Role for sentinel lymph node dissection in the management of large
(> or = 5 cm) invasive breast cancer. Ann Surg Oncol. 2001 Oct;8(9):688–92.

RI
39. Wong SL, Chao C, Edwards MJ, Tuttle TM, Noyes RD, Carlson DJ, et al. Accuracy of sentinel lymph
node biopsy for patients with T2 and T3 breast cancers. Am Surg. 2001 Jun;67(6):522-526-528.

SC
40. Hidar S, Bibi M, Gharbi O, Tebra S, Trabelsi A, Korbi S, et al. Sentinel lymph node biopsy after
neoadjuvant chemotherapy in inflammatory breast cancer. Int J Surg Lond Engl. 2009
Jun;7(3):272–5.

41.
U
Gradishar WJ, Anderson BO, Balassanian R, Blair SL, Burstein HJ, Cyr A, et al. NCCN Guidelines
AN
Insights: Breast Cancer, Version 1.2017. J Natl Compr Cancer Netw JNCCN. 2017 Apr;15(4):433–51.

42. Loibl S, Schmidt A, Gentilini O, Kaufman B, Kuhl C, Denkert C, et al. Breast Cancer Diagnosed During
Pregnancy: Adapting Recent Advances in Breast Cancer Care for Pregnant Patients. JAMA Oncol.
M

2015 Nov;1(8):1145–53.

43. Gentilini O, Cremonesi M, Trifirò G, Ferrari M, Baio SM, Caracciolo M, et al. Safety of sentinel node
D

biopsy in pregnant patients with breast cancer. Ann Oncol Off J Eur Soc Med Oncol. 2004
Sep;15(9):1348–51.
TE

44. Gentilini O, Cremonesi M, Toesca A, Colombo N, Peccatori F, Sironi R, et al. Sentinel lymph node
biopsy in pregnant patients with breast cancer. Eur J Nucl Med Mol Imaging. 2010 Jan;37(1):78–83.
EP

45. Pandit-Taskar N, Dauer LT, Montgomery L, St Germain J, Zanzonico PB, Divgi CR. Organ and fetal
absorbed dose estimates from 99mTc-sulfur colloid lymphoscintigraphy and sentinel node
localization in breast cancer patients. J Nucl Med Off Publ Soc Nucl Med. 2006 Jul;47(7):1202–8.
C

46. Pruthi S, Haakenson C, Brost BC, Bryant K, Reid JM, Singh R, et al. Pharmacokinetics of methylene
blue dye for lymphatic mapping in breast cancer-implications for use in pregnancy. Am J Surg.
AC

2011 Jan;201(1):70–5.

47. Raut CP, Daley MD, Hunt KK, Akins J, Ross MI, Singletary SE, et al. Anaphylactoid reactions to
isosulfan blue dye during breast cancer lymphatic mapping in patients given preoperative
prophylaxis. J Clin Oncol Off J Am Soc Clin Oncol. 2004 Feb 1;22(3):567–8.

48. Ballman KV, Buzdar A, Carey LA, Sikov WM, Strand C, Partridge AH. Age, race, BMI, and pathologic
complete response following neoadjuvant chemotherapy: An analysis of four alliance clinical trials.
J Clin Oncol Off J Am Soc Clin Oncol. 2015 Oct;33(28_suppl):33.

24
ACCEPTED MANUSCRIPT

49. Cortazar P, Kluetz PG. Neoadjuvant breast cancer therapy and drug development. Clin Adv
Hematol Oncol HO. 2015 Nov;13(11):755–61.

50. Steenbruggen TG, van Ramshorst MS, Kok M, Linn SC, Smorenburg CH, Sonke GS. Neoadjuvant
Therapy for Breast Cancer: Established Concepts and Emerging Strategies. Drugs. 2017
Aug;77(12):1313–36.

PT
51. Xing Y, Foy M, Cox DD, Kuerer HM, Hunt KK, Cormier JN. Meta-analysis of sentinel lymph node
biopsy after preoperative chemotherapy in patients with breast cancer. Br J Surg. 2006
May;93(5):539–46.

RI
52. Nason KS, Anderson BO, Byrd DR, Dunnwald LK, Eary JF, Mankoff DA, et al. Increased false negative
sentinel node biopsy rates after preoperative chemotherapy for invasive breast carcinoma. Cancer.
2000 Dec 1;89(11):2187–94.

SC
53. Brady EW. Sentinel lymph node mapping following neoadjuvant chemotherapy for breast cancer.
Breast J. 2002 Apr;8(2):97–100.

U
54. Classe J-M, Bordes V, Campion L, Mignotte H, Dravet F, Leveque J, et al. Sentinel lymph node
biopsy after neoadjuvant chemotherapy for advanced breast cancer: results of Ganglion Sentinelle
AN
et Chimiotherapie Neoadjuvante, a French prospective multicentric study. J Clin Oncol Off J Am Soc
Clin Oncol. 2009 Feb 10;27(5):726–32.

55. Hunt KK, Yi M, Mittendorf EA, Guerrero C, Babiera GV, Bedrosian I, et al. Sentinel lymph node
M

surgery after neoadjuvant chemotherapy is accurate and reduces the need for axillary dissection in
breast cancer patients. Ann Surg. 2009 Oct;250(4):558–66.
D

56. Tan VKM, Goh BKP, Fook-Chong S, Khin L-W, Wong W-K, Yong W-S. The feasibility and accuracy of
sentinel lymph node biopsy in clinically node-negative patients after neoadjuvant chemotherapy
TE

for breast cancer--a systematic review and meta-analysis. J Surg Oncol. 2011 Jul 1;104(1):97–103.

57. Kelly AM, Dwamena B, Cronin P, Carlos RC. Breast cancer sentinel node identification and
classification after neoadjuvant chemotherapy-systematic review and meta analysis. Acad Radiol.
EP

2009 May;16(5):551–63.

58. White J, Mamounas E. Locoregional radiotherapy in patients with breast cancer responding to
neoadjuvant chemotherapy: a paradigm for treatment individualization. J Clin Oncol Off J Am Soc
C

Clin Oncol. 2014 Feb 20;32(6):494–5.


AC

59. Pilewskie M, Morrow M. Axillary Nodal Management Following Neoadjuvant Chemotherapy: A


Review. JAMA Oncol. 2017 Apr 1;3(4):549–55.

60. 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 Jun;14(7):609–18.

61. 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 Oct 9;310(14):1455–61.

25
ACCEPTED MANUSCRIPT

62. Boileau J-F, Poirier B, Basik M, Holloway CMB, Gaboury L, Sideris L, et al. Sentinel node biopsy after
neoadjuvant chemotherapy in biopsy-proven node-positive breast cancer: the SN FNAC study. J
Clin Oncol Off J Am Soc Clin Oncol. 2015 Jan 20;33(3):258–64.

63. Zetterlund LH, Frisell J, Zouzos A, Axelsson R, Hatschek T, de Boniface J, et al. Swedish prospective
multicenter trial evaluating sentinel lymph node biopsy after neoadjuvant systemic therapy in
clinically node-positive breast cancer. Breast Cancer Res Treat. 2017 May;163(1):103–10.

PT
64. Chapman CH, Jagsi R. Postmastectomy Radiotherapy After Neoadjuvant Chemotherapy: A Review
of the Evidence. Oncol Williston Park N. 2015 Sep;29(9):657–66.

RI
65. Donker M, Straver ME, Wesseling J, Loo CE, Schot M, Drukker CA, et al. Marking axillary lymph
nodes with radioactive iodine seeds for axillary staging after neoadjuvant systemic treatment in
breast cancer patients: the MARI procedure. Ann Surg. 2015 Feb;261(2):378–82.

SC
66. Kim EY, Byon WS, Lee KH, Yun J-S, Park YL, Park CH, et al. Feasibility of Preoperative Axillary Lymph
Node Marking with a Clip in Breast Cancer Patients Before Neoadjuvant Chemotherapy: A
Preliminary Study. World J Surg. 2017 Aug 14;

67.
U
Nguyen TT, Hieken TJ, Glazebrook KN, Boughey JC. Localizing the Clipped Node in Patients with
AN
Node-Positive Breast Cancer Treated with Neoadjuvant Chemotherapy: Early Learning Experience
and Challenges. Ann Surg Oncol. 2017 Aug 1;

68. Groen EJ, Elshof LE, Visser LL, Rutgers EJT, Winter-Warnars HAO, Lips EH, et al. Finding the balance
M

between over- and under-treatment of ductal carcinoma in situ (DCIS). Breast Edinb Scotl. 2017
Feb;31:274–83.
D

69. Wapnir IL, Dignam JJ, Fisher B, Mamounas EP, Anderson SJ, Julian TB, et al. Long-term outcomes of
invasive ipsilateral breast tumor recurrences after lumpectomy in NSABP B-17 and B-24
TE

randomized clinical trials for DCIS. J Natl Cancer Inst. 2011 Mar 16;103(6):478–88.

70. Bonev V, De Paz Villanueva CC, Solomon N, Senthil M, Reeves ME, Garberoglio C, et al. Is Sentinel
Lymph Node Dissection Necessary in All Patients with Ductal Carcinoma In Situ Undergoing Total
EP

Mastectomy? Am Surg. 2016 Oct;82(10):982–4.

71. Caswell-Smith P, Wall M. Ductal carcinoma in situ: Is core needle biopsy ever enough? J Med
Imaging Radiat Oncol. 2017 Feb;61(1):29–33.
C

72. Mitchell KB, Lin H, Shen Y, Colfry A, Kuerer H, Shaitelman SF, et al. DCIS and axillary nodal
AC

evaluation: compliance with national guidelines. BMC Surg. 2017 Feb 7;17(1):12.

73. Ansari B, Ogston SA, Purdie CA, Adamson DJ, Brown DC, Thompson AM. Meta-analysis of sentinel
node biopsy in ductal carcinoma in situ of the breast. Br J Surg. 2008 May;95(5):547–54.

74. Heymans C, van Bastelaar J, Visschers RGJ, Vissers YLJ. Sentinel Node Procedure Obsolete in
Lumpectomy for Ductal Carcinoma In Situ. Clin Breast Cancer. 2017 Jun;17(3):e87–93.

26
ACCEPTED MANUSCRIPT

75. van Roozendaal LM, Goorts B, Klinkert M, Keymeulen KBMI, De Vries B, Strobbe LJA, et al. Sentinel
lymph node biopsy can be omitted in DCIS patients treated with breast conserving therapy. Breast
Cancer Res Treat. 2016 Apr;156(3):517–25.

76. Pilewskie M, Karsten M, Radosa J, Eaton A, King TA. Is Sentinel Lymph Node Biopsy Indicated at
Completion Mastectomy for Ductal Carcinoma In Situ? Ann Surg Oncol. 2016 Jul;23(7):2229–34.

PT
77. Lara JF, Young SM, Velilla RE, Santoro EJ, Templeton SF. The relevance of occult axillary
micrometastasis in ductal carcinoma in situ: a clinicopathologic study with long-term follow-up.
Cancer. 2003 Nov 15;98(10):2105–13.

RI
78. Prendeville S, Ryan C, Feeley L, O’Connell F, Browne TJ, O’Sullivan MJ, et al. Sentinel lymph node
biopsy is not warranted following a core needle biopsy diagnosis of ductal carcinoma in situ (DCIS)
of the breast. Breast Edinb Scotl. 2015 Jun;24(3):197–200.

SC
79. Meretoja TJ, Leidenius MH, Heikkilä PS, Joensuu H. Sentinel node biopsy in breast cancer patients
with large or multifocal tumors. Ann Surg Oncol. 2009 May;16(5):1148–55.

U
80. Knauer M, Konstantiniuk P, Haid A, Wenzl E, Riegler-Keil M, Pöstlberger S, et al. Multicentric breast
cancer: a new indication for sentinel node biopsy--a multi-institutional validation study. J Clin
AN
Oncol Off J Am Soc Clin Oncol. 2006 Jul 20;24(21):3374–80.

81. Donker M, van Tienhoven G, Straver ME, Meijnen P, van de Velde CJH, Mansel RE, et al.
Radiotherapy or surgery of the axilla after a positive sentinel node in breast cancer (EORTC 10981-
M

22023 AMAROS): a randomised, multicentre, open-label, phase 3 non-inferiority trial. Lancet


Oncol. 2014 Nov;15(12):1303–10.
D

82. Donker M, Straver ME, van Tienhoven G, van de Velde CJH, Mansel RE, Litière S, et al. Comparison
of the sentinel node procedure between patients with multifocal and unifocal breast cancer in the
TE

EORTC 10981-22023 AMAROS Trial: identification rate and nodal outcome. Eur J Cancer Oxf Engl
1990. 2013 Jun;49(9):2093–100.

83. de Boer M, van Dijck J a. a. M, Bult P, Borm GF, Tjan-Heijnen VCG. Breast cancer prognosis and
EP

occult lymph node metastases, isolated tumor cells, and micrometastases. J Natl Cancer Inst. 2010
Mar 17;102(6):410–25.

84. Weaver DL, Ashikaga T, Krag DN, Skelly JM, Anderson SJ, Harlow SP, et al. Effect of occult
C

metastases on survival in node-negative breast cancer. N Engl J Med. 2011 Feb 3;364(5):412–21.
AC

85. Reed J, Rosman M, Verbanac KM, Mannie A, Cheng Z, Tafra L. Prognostic implications of isolated
tumor cells and micrometastases in sentinel nodes of patients with invasive breast cancer: 10-year
analysis of patients enrolled in the prospective East Carolina University/Anne Arundel Medical
Center Sentinel Node Multicenter Study. J Am Coll Surg. 2009 Mar;208(3):333–40.

86. Degnim AC, Zakaria S, Boughey JC, Sookhan N, Reynolds C, Donohue JH, et al. Axillary recurrence in
breast cancer patients with isolated tumor cells in the sentinel lymph node [AJCC N0(i+)]. Ann Surg
Oncol. 2010 Oct;17(10):2685–9.

27
ACCEPTED MANUSCRIPT

87. Suyoi A, Bains SK, Kothari A, Douek M, Agbaje O, Hamed H, et al. When is a completion axillary
lymph node dissection necessary in the presence of a positive sentinel lymph node? Eur J Cancer
Oxf Engl 1990. 2014 Mar;50(4):690–7.

88. van Diest PJ, de Boer M, van Deurzen CHM, Tjan-Heijnen VCG. Micrometastases and isolated
tumor cells in breast cancer are indeed associated with poorer outcome. J Clin Oncol Off J Am Soc
Clin Oncol. 2010 Mar 20;28(9):e140; author reply e141-142.

PT
89. de Boer M, van Deurzen CHM, van Dijck JAAM, Borm GF, van Diest PJ, Adang EMM, et al.
Micrometastases or isolated tumor cells and the outcome of breast cancer. N Engl J Med. 2009
Aug 13;361(7):653–63.

RI
90. Giuliano AE, Hawes D, Ballman KV, Whitworth PW, Blumencranz PW, Reintgen DS, et al.
Association of occult metastases in sentinel lymph nodes and bone marrow with survival among

SC
women with early-stage invasive breast cancer. JAMA. 2011 Jul 27;306(4):385–93.

91. van Nijnatten TJA, Simons JM, Moossdorff M, de Munck L, Lobbes MBI, van der Pol CC, et al.
Prognosis of residual axillary disease after neoadjuvant chemotherapy in clinically node-positive

U
breast cancer patients: isolated tumor cells and micrometastases carry a better prognosis than
macrometastases. Breast Cancer Res Treat. 2017 May;163(1):159–66.
AN
92. Solá M, Alberro JA, Fraile M, Santesteban P, Ramos M, Fabregas R, 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
M

Oncol. 2013 Jan;20(1):120–7.

93. Sávolt Á, Péley G, Polgár C, Udvarhelyi N, Rubovszky G, Kovács E, et al. Eight-year follow up result
D

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,
TE

non-inferiority trial. Eur J Surg Oncol J Eur Soc Surg Oncol Br Assoc Surg Oncol. 2017
Apr;43(4):672–9.

94. Galimberti V, Cole BF, Zurrida S, Viale G, Luini A, Veronesi P, et al. Axillary dissection versus no
EP

axillary dissection in patients with sentinel-node micrometastases (IBCSG 23-01): a phase 3


randomised controlled trial. Lancet Oncol. 2013 Apr;14(4):297–305.

95. Goyal A, Dodwell D. POSNOC: A Randomised Trial Looking at Axillary Treatment in Women with
C

One or Two Sentinel Nodes with Macrometastases. Clin Oncol R Coll Radiol G B. 2015
Dec;27(12):692–5.
AC

28

Você também pode gostar