ArticlePDF AvailableLiterature Review

Axillary lymph node dissection in early-stage invasive breast cancer: Is it still standard today?

Authors:

Abstract

Evaluation of axillary lymph node status by sentinel lymph node biopsy (SLNB) and complete axillary lymph node dissection (ALND) are an inherent part of breast cancer treatment. Increased understanding of tumor biology has changed the prognostic and therapeutic impact of lymph node status. Non-invasive imaging techniques like axillary ultrasound, FDG-PET, or MRI revealed moderate sensitivity and high specificity in evaluation of lymph node status. Therefore, they are not sufficient for lymph node staging. Otherwise, the impact of remaining micrometastases and even macrometastases for prognosis and treatment decisions is overestimated. Considering tumor biology, the distinction of axillary metastases in isolated tumor cells (ITC, pN0(i+)); micrometastases (pN1mi), and macrometastases (pN1a) is not comprehensible. Increasing data support the thesis that remaining axillary metastases neither increase the axillary recurrence rate nor decrease overall survival. It is doubtful that axillary tumor cells are capable to complete the complex multistep metastatic process. If applied, axillary metastases are sensitive to systemic treatment and are targeted by postoperative tangential breast irradiation. Therefore, the controversy about the clinical relevance of tumor cell clusters or micrometastases in SLN is a sophisticated but not contemporary discussion. Currently, there is no indication for axillary surgery in elderly patients with favorable tumors and clinically tumor-free lymph nodes. Nonetheless, a rational and evidence-based approach to the management of clinically and sonographically N0 patients with planned breast-conserving surgery and limited tumor size is needed now.
1
Axillary lymph node dissection in early-stage invasive breast cancer: Is it still
standard today?
Bernd Gerber, Kristin Heintze, Johannes Stubert, Max Dieterich, Steffi Hartmann,
Angrit Stachs, Toralf Reimer
Department of Obstetrics and Gynecology, University of Rostock, Suedring 81, 18059
Rostock, Germany
Correspondence Address:
Bernd Gerber, MD, PhD
Department of Obstetrics and Gynecology, University of Rostock,
Suedring 81, 18059 Rostock, Germany
e-mail: bernd.gerber@med.uni-rostock.de
Tel: +49 / 381 / 4401-4500
Fax: +49 / 381 / 4401-4599
Short running head: Axillary lymph node extirpation in early breast cancer
Key words: breast cancer, axillary lymph nodes, dissection, sentinel, prognosis,
metastases
2
Responsibilities:
Bernd Gerber
idea, literature search, writing, coordination
Kristin Heintze
literature search, writing, macrometastases
and In which patient is axillary surgery
avoidable?
Johannes Stubert
literature search, writing tumor biology and
do lymph node metastases metastasize?
Max Dieterich
literature search, writing micrometastases
Steffi Hartmann
literature search, writing non-invasive
diagnostics
Angrit Stachs
literature search, writing non-invasive
diagnostics
Toralf Reimer
literature search, reviewing, writing,
coordination
3
Abstract
Evaluation of axillary lymph node status by sentinel lymph node biopsy (SLNB) and
complete axillary lymph node dissection (ALND) are an inherent part of breast cancer
treatment. Increased understanding of tumor biology has changed the prognostic and
therapeutic impact of lymph node status. Non-invasive imaging techniques like
axillary ultrasound, FDG-PET or MRI revealed moderate sensitivity and high
specificity in evaluation of lymph node status. Therefore, they are not sufficient for
lymph node staging. Otherwise the impact of remaining micrometastases and even
macrometastases for prognosis and treatment decisions is overestimated.
Considering tumor biology, the distinction of axillary metastases in isolated tumor
cells (ITC, pN0[i+]); micrometastases (pN1mi), and macrometastases (pN1a) is not
comprehensible. Increasing data support the thesis that remaining axillary
metastases neither increase the axillary recurrence rate nor decrease overall
survival. It is doubtful that axillary tumor cells are capable to complete the complex
multistep metastatic process. If applied, axillary metastases are sensitive to systemic
treatment and are targeted by postoperative tangential breast irradiation. Therefore,
the controversy about the clinical relevance of tumor cell clusters or micrometastases
in SLN is a sophisticated but not contemporary discussion. Currently, there is no
indication for axillary surgery in elderly patients with favorable tumors and clinically
tumor-free lymph nodes. Nonetheless, a rational and evidence-based approach to
the management of clinically and sonographically N0 patients with planned breast-
conserving surgery and limited tumor size is needed now.
4
Introduction
The axillary lymph node dissection (ALND) with removal and histopathological
examination of at least 10 nodes was an inherent part of surgical treatment of breast
cancer for a considerable time. During the last decade ALND was replaced by
sentinel lymph node biopsy (SLNB) in patients with clinically and sonographically
unsuspicious lymph nodes [1-3]. As a consequence the arm morbidity was reduced
markedly [4]. The SLNB requires estimated costs of 200-250 per sentinel labeling
and 1,750 per axillary surgery (Webgrouper DRG Research Group, University of
Muenster, Germany). Due to nationwide mammography screening in most industrial
countries, a greater number of smaller tumors without axillary lymph node
involvement is detected. More than 60% of all primary operable breast cancers do
not have axillary lymph node metastases. This in turn means that even SLNB
represents an overtreatment and is not indicated in the majority of patients. Non-
invasive methods like ultrasound, fluorodeoxyglucose positron emission tomography
(FDG-PET), and magnetic resonance imaging (MRI), have gained more importance
in staging the axillary lymph nodes. With improved insight into primary tumor biology
and metastasis behavior, the relevance of nodal status for adjuvant treatment
decisions is decreasing. Currently, in cases with positive sentinel lymph nodes (SLN)
the need for completion ALND is arguable. Nevertheless, there is an ongoing
discussion about the prognostic impact of isolated tumor cells (ITC, pN0(i+); < 0.2
mm), micrometastases (pN1mi; >0.2 mm to <2.0 mm), and macrometastases (pN1a;
>2.0 mm) in SLNs. The aim of this review was firstly to assess the imaging
techniques in axillary lymph node staging and secondly to evaluate the impact of
pathological lymph node status regarding prognosis and treatment decisions. The
key question to be answered is: Is there a chance to avoid axillary lymph node
5
surgery in patients with early breast cancer? This review is not covering the issue of
axillary surgery following primary systemic treatment.
Materials and methods
We performed a selective literature search on PubMed database using the search
terms “breast cancer & axillary lymph nodes” and “ultrasound or sonography”, “MRI”
or “PET” and “sentinel lymph node and micrometastases” for the period from January
2002 to February 2011. Activated limits for search were Female, Clinical Trial, Meta-
Analysis, Randomized Controlled Trial, English”. Contributions to international
congresses on breast cancer in 2009 (SABCS, San Antonio Breast Cancer
Symposium; ECCO, European Cancer Organization; EBCC, European Breast
Cancer Conference) and 2010 (ASCO, American Society of Clinical Oncology) were
included. The treatment recommendations of AGO-Mamma (the Breast Group of the
German Gynecological Oncology Working Group [5]) and the NCCN (National
Comprehensive Cancer Network) Guidelines [6] together with the Cochrane Library
[7] were also considered.
Imaging techniques
The exclusion of lymph node metastases by using non-invasive methods could
reduce the rate of axillary surgery. However, the experience of examiner and period
of availability are crucial for the diagnostic precision and prediction.
Axillary Ultrasound (AUS)
Clinically palpable axillary nodes are widely considered as contraindication to SLNB
in breast cancer. However, various studies have shown that clinical assessment of
axillary lymph nodes alone is inaccurate with a false-positive rate up to 40% [8, 9].
6
Among patients with clinically uninvolved axillary lymph nodes, AUS has additional
value in detecting pathological axillary nodes [10, 11]. One third of pathological
involved lymph nodes could predicted preoperatively by ultrasound. AUS combined
with fine needle aspiration (FNA) biopsy of suspicious nodes has shown useful
results to guide preoperative surgical planning [12, 13]. However, the reported
sensitivity for AUS-FNA has varied considerably between different studies [14, 15].
Because of the risk of false-negative results, negative FNA findings are almost
followed by SLNB for confirmation.
According to guidelines, a SLNB is only indicated in patients with histologically
proven invasive breast cancer and clinically and sonographically insuspect lymph
nodes. In a systematic review including 16 studies, in which AUS without palpable
lymph nodes was performed, the node size and the morphology were used as criteria
for positivity [15]. For lymph node size sensitivity varied between 48.8% (95%
confidence interval [CI]: 39.6-58%) and 87.1% (76.1-94.3%) and specificity, between
55.6% (44.7-66.3%) and 97.3% (86.1-99.9%). If lymph node morphology was used
as the criterion for positivity, sensitivity ranged from 26.4% (15.3-40.3%) to 75.9%
(56.4-89.7%) and specificity, from 88.4% (82.1-93.1%) to 98.1% (90.1-99.9%). After
sonographically guided node biopsy, sensitivity varied between 30.6% (22.5-39.6%)
and 62.9% (49.7-74.8%) and specificity was nearly 100% (94.8-100%). More recent
studies confirmed these percentage rates for AUS-FNA showing sensitivities from
53% to 59% and specificities of 100% [16, 17]. AUS-FNA seems to be most useful in
the preoperative assessment of patients with large breast tumors (>2 cm) or with
lymph nodes that appear abnormal.
In conclusion, AUS is moderately sensitive and fairly specific in the diagnosis of
axillary metastatic involvement. These scattered variabilities are the result of the
missing clear criteria for evaluation of axillary lymph nodes by ultrasound. The
7
classification of axillary lymph nodes on the basis of cortical thickness (cut-off 2.5
mm) and the appearance of the fatty hilum on sonography was effective for
predicting the presences of metastases in a recent study [18]. In future, addition of
contrast-enhanced color and power doppler ultrasound may improve the sensitivity of
AUS [19]. Malignant lymph nodes showed longer contrast enhancement duration
compared to benign lymph nodes.
FDG-PET
FDG-PET seemed to be an interesting approach as a non-invasive method of staging
the axilla in breast cancer patients. In a meta-analysis, 21 studies for clinical use of
FDG-PET in investigating axillary nodes were graded on methodological quality of
the single studies depending on number and selection of patients, technical details of
examination, pro- or retrospective design, and kind of lymph node biopsy [20]. As
shown in table 1 best quality studies showed a sensitivity, specificity, positive and
negative predictive value of around 80%, whereas in the poorer quality studies the
diagnostic accuracy was higher. However, the variability between study designs has
made it difficult to compare and aggregate the results of these studies. As well,
caution must be exercised when analyzing results from older studies because of the
increased accuracy of the newer scanners [20].
In a current meta-analysis of 25 studies including 2,460 patients a trend to lower
sensitivity (3785%) in the most recent published and higher quality studies (year
2005-2009 and sample size >25) was detectable, whereas the specificity (84100%)
remained high [21]. This meta-analysis also indicated that the sensitivity of PET is
not sufficient to detect small metastatic deposits (micrometastases). Interestingly, the
metabolic activity is variable according to the histopathological subtype of breast
cancer. The mean tumor maximum standardized uptake value SUV(max) in lymph
8
node metastases of estrogen receptor (ER)-positive, triple-negative and human
epidermal growth factor receptor 2 (HER2)-positive tumors was 6.6, 11.6, and 6.6,
respectively [22].
In conclusion, sensitivity and specifity of PET is comparable to AUS, but because
AUS is less expensive, AUS remains standard of care these days. The combination
of AUS and PET seems to be improved the diagnostic accuracy [23]. The additional
benefit of an assessment of distal metastatic spread provided by PET requires further
investigation [21].
MRI
Breast MRI assessment of breast cancer patients considered for primary surgical
treatment is being used more frequently, especially if breast-conserving surgery
(BCS) is planned. During such assessment, regional lymph nodes are usually
included. Using different contrast agents, sensitivity ranged from 63100% and
specificity from 56100% (Tab. 2). Like FDG-PET, contrast-enhanced MRI accuracy
showed a close relationship with histopathologic subtypes of invasive breast cancer
and its limitation in detection of axillary micrometastases [29, 31, 32]. Adding axillary
MRI sequentially after AUS did not significantly improve detection of positive nodes in
AUS-negative cases (false-negative rate of 28% and false-positive rate of 18% for
MRI) [16].
Taken together, all imaging techniques are of high specificity and moderate
sensitivity especially in cases with small lymph node metastases. The value of
combined imaging techniques for improving the diagnostic accuracy should be
considered critically with respect to benefit-cost-ratio.
9
In the following we have to discuss three fundamental questions: Firstly, is it really
important to know the nodal status for treatment decisions? Secondly, is there a
chance to avoid axillary surgery in some cases and how can we select these
patients? And thirdly, how is the biological behavior of metastatic involved lymph
nodes left in situ (separated by tumor cluster size)?
Lymph node status for prognosis and treatment decision
Since Halsted first defined the issue of axillary lymph nodes 100 years ago, the
lymph node status is the basis for prognosis and treatment decision in breast cancer
[33]. It is well known that the prognosis declines with increasing number of metastatic
lymph nodes or more precisely, with an increasing lymph node ratio [34, 35]. Larger
breast tumors tended to be associated with a greater number of metastatic involved
axillary lymph nodes than smaller tumors, indicating that a higher number of involved
lymph nodes reflects a longer tumor growth time. In a multivariable logistic regression
analysis the molecular subtypes of breast tumors (luminal, luminal/HER2-positive,
HER2-positive/ER-negative/progesterone receptor (PgR)-negative, and basal-like)
had a predictive effect for nodal involvement (p=0.000001). Using the luminal
subtype as reference, the basal subtype has an odds ratio for axillary lymph node
involvement of 0.53 (95% CI: 0.41-0.69) in a model using core biopsy data [36]. A
Belgian group reported higher likelihood of being lymph node positive for triple-
positive tumors (HER2 positive/ER positive/PgR positive) compared to other
subtypes (56.2 vs. 35.7%) [37]. Patients older than 70 years were more likely to have
positive nodes with increasing age. The effect of age in older women was most
pronounced for small tumors. On the contrary, for very large tumors, the risk of lymph
node involvement even decreased with increasing age [38]. Other studies reported
no increase of lymph node involvement in elderly patients when investigating the
10
relationship in pT-strata and adjusting for other prognostic factors. Taken together,
axillary lymph node involvement is the result of different factors and lymph node
status at all does not reflect the aggressive behavior of the tumor correctly [39].
Tumor aggressiveness especially in patient without axillary lymph node involvement
has primarily to do with biological features of the tumor cells from the primary tumor.
Based on this, it seems reasonable that poor outcome depends more on the tumor
biology than on lymph node involvement [40-44]. Gene expression analyses of the
primary tumor indicated that 51% of lymph node-negative tumors and 49% of lymph
node-positive tumors have a poor prognostic signature [45]. The value of gene
expression profiling as a prognostic tool in clinical practice is currently being
evaluated in two large, prospective, randomised studies (TAILORx, MINDACT).
Lymph node status is a good indicator of tumor stage related to the time point of
diagnosis, but it does not reflect the real tumor biology, metastatic behavior as well
as sensitivity to systemic treatment. Extensive characterization of the primary tumor
will be available by gene signatures in future [44-46]. Nevertheless, nodal status is
still an established parameter for treatment decisions in current guidelines. According
to the NCCN guidelines the indication for post-mastectomy radiotherapy in all
patients or for adjuvant chemotherapy in ER-positive/HER2-negative disease is
linked to the number of involved lymph nodes [6].
This adherence to treatment recommendations on the basis of axillary nodal status
was also observed in own previous work. We have shown that the knowledge of
pathologic nodal status influences the indications for postoperative chemotherapy
and post-mastectomy radiotherapy [47]. Re-discussion of postoperative tumor board
records without information regarding axillary lymph node status resulted in adjuvant
treatment recommendations different from the current guidelines in one third of all
cases (72 of 207 patients), especially in carcinomas with present lymphangiosis.
11
However, this finding is the result of guidelines which are mostly based on elderly
studies with nodal status as an in- or exclusion criterion. More recent studies have
confirmed the effectiveness of chemotherapy and trastuzumab even in high risk,
node-negative patients [48, 49].
Engel et al. reported 15 reasons to stop axillary surgery entirely [50]. This paper was
hypothesis generating in 2006 without changing current surgical practice. The
author’s discussion is based on the hypothesis that locoregional and distant
metastases develop over time from disseminated tumor cells that originate from the
primary and not from the lymph nodes. The current pros and contras for axillary
lymph node status and associated treatment decisions are summarized in table 3
using some arguments published by Engel et al..
Definition of patient subgroups without any axillary surgery
SLNB is a minimal invasive procedure with the same oncologic safety as ALND and
low (but not zero) morbidity [51-53]. Therefore, SLNB is suggested to be an optimal
approach to reduce the morbidity by the assessment of axillary status in clinically
node-negative breast cancer. But the indication for SLNB in all clinically node-
negative patients is questionable because at least 70% of these patients are
pathologically tumor-free in the axillary nodes. Two randomized trials investigated the
use of ALND versus no axillary surgery in elderly patients [54, 55]. A third trial
randomized patients without any axillary surgery to no axillary treatment versus
axillary radiotherapy (ART) [56]. The results of all three studies showed a very low
rate of axillary recurrences, even in the arms without axillary surgery, and
comparable disease-free and overall survival (Tab. 4).
In an observational study with 671 consecutive patients, aged >70 years, and a
clinically tumor-free axilla, 172 received and 499 did not receive axillary dissection.
12
After a median follow-up of 15 years, there was no significant difference in breast
cancer mortality between the axillary and no axillary clearance groups. Ipsilateral
axillary recurrence rate was zero in the axillary dissection group and relatively low in
the no axillary dissection group: 5.8% overall and 3.7% for pT1 patients [57]. These
axillary recurrence rates in the non-operated group were much lower than the rate of
pathological nodal involvement in the axillary dissection group (33.7% pN+ overall
and 29% pN+ for pT1 patients). The authors concluded, that due to a very low
cumulative incidence of axillary recurrence in elderly patients with BCS both, SLNB
and ALND can be avoided. Axillary dissection should be restricted to the small
number of patients who will later develop apparent axillary disease.
Axillary recurrence is rare, although four-times more common in younger women
(<40 years) than in older patients (50-60 years) [58]. Current NCCN guidelines
consider the performance of ALND as optional in patients who have particularly
favorable tumors, in patients for whom the selection of adjuvant systemic therapy is
unlikely to be affected, for the elderly, or those with severe comorbid conditions [6].
There is no clear statement in the NCCN guidelines in which patients the SLNB can
be avoided.
ART has been used instead of or in addition to axillary sampling or ALND. In 2004,
Louis-Sylvestre et al. published results of a prospective randomized trial comparing
lumpectomy plus ART versus lumpectomy plus ALND (n=658; median follow-up 180
months; 21% node-positive in the ALND group) [59]. Overall survival rates were
identical in both groups, recurrences in the axillary nodes were less frequent in the
ALND group (1% versus 3%, p=0.04). As an alternative to completion ALND for
SLNB positive patients, ART has been suggested [60]. This concept is undergoing
prospective evaluation by the EORTC trial, “After Mapping of the Axilla: Radiotherapy
versus Surgery?” (AMAROS) [61]. The trial is full recruited, but definitive data will not
13
be available for years. ART in the setting of no prior axillary surgery has generally
been associated with less morbidity than that seen with ALND. The incidence of
morbidity after SLNB combined with ART has not been well characterized [62].
In situ left metastatic involved axillary lymph nodes and prognosis
Macrometastases
SLNB trial arms with immediate conventional ALND showed false-negative rates up
to 9.8% regarding accuracy of SLNB [2, 63]. Even after ALND of level I and II, up to
30% of positive lymph nodes remain in the axilla, 15% of these present as 'skip
metastases' [64, 65]. Rudenstam et al. reported nodal involvement in 28% of the
patients who had axillary dissection [54]. Isolated lymph node recurrences are rare
(<2%) in patients with BCS and lymph node-positive disease [58, 66, 67].
The presentation of the prospective, multicentric ACOSOG Z0011 trial at the ASCO
Congress 2010 was a landmark for the discussion about surgical options in pN+
patients after SLNB. According to the protocol, nearly 900 patients with clinically T1-2
cN0 cM0 breast cancer and BCS with metastatic involved sentinel nodes (routine
hematoxylin and eosin [H&E] detected) were randomized to no further axillary
dissection or completion ALND [68]. Targeted enrollment was 1,900 women, but the
trial closed earlier due to lower than expected accrual and event rates. Both arms
were comparable with respect to pathological tumor size, grading, histological tumor
type, hormone receptor status and systemic treatment. After a median follow up of
6.3 years there were no significant differences between SLNB alone and SLNB plus
ALND arms concerning 5-year local recurrence rate (1.6% versus 3.1%), 5-year
disease-free survival (83.9% versus 82.2%), or 5-year overall survival (92.5% versus
91.8%). The local axillary recurrence rate was 0.9% for SLNB alone and 0.5% for
SLNB+ALND [68, 69]. Because all patients were treated with opposing and
14
tangential field irradiation, a local effect on the axilla could not be excluded
completely, however it seems very unlikely in this dimension [58, 66].
According to these findings the German AGO Breast Group has adapted the current
guidelines in March 2011. The completion ALND is not longer recommended (only
“+/- grade of recommendation) in patients with positive sentinel nodes and: cT1/2,
cN0, less than three sentinel nodes involved, BCS plus tangential breast irradiation,
and no alternative ART [5]. The ACOSOG Z0011 data are supported by a
retrospective SEER database analysis (1998-2004) including 26,986 patients with
positive sentinel nodes [70]. Among those, 4,425 (16.4%) underwent SLNB alone,
and 22,561 (83.6%) underwent SLNB with completion ALND. Contrary to the Z0011
study, the SEER analysis was not restricted to BCS (21.2% with total mastectomy in
the SLNB alone arm). At a median follow-up of 50 months, there were no statistically
significant differences in overall survival between patients with SLNB alone versus
complete ALND.
Of 97,314 patients from the US National Cancer Database who underwent SLNB and
who had nodal metastases, 20.8% underwent SLNB alone, and 79.2% underwent
SLNB with complete ALND [71]. In patients with macroscopic nodal metastases and
a median follow-up of 63 months, there was a non-significant trend towards better
outcomes for complete ALND compared to SLNB alone: axillary recurrence HR=0.58
(95% CI: 0.32-1.06) and overall survival HR=0.89 (95% CI: 0.76-1.04). A randomized
controlled trial with 30 years follow-up indicated that clearing the internal mammary
or axillary lymph nodes has no impact on long-term survival [72]. Moreover, tumor
location affects the frequency of lymph node metastases but not the survival. Janni et
al. showed that 25.8% of patients with a tumor in the medial quadrant of the breast
have axillary lymph node metastases compared with 35.5% of patients with a lateral
localized breast tumor [73]. Despite the 10% difference, survival time was similar for
15
both tumor locations. There is no additional survival advantage in clearing level III of
the axilla compared with clearing restricted to levels I and II [74].
The Memorial Sloan-Kettering Cancer Center (MSKCC) developed nomograms to
predict the likelihood of a positive sentinel node or finding additional positive non-
sentinel nodes in patients with involved SLNs [75, 76]. The nomograms are a
statistical approach for calculation metastatic axillary risk by combining different
histopathologic factors (plus age, which is incorporated in the sentinel nomogram)
and are available online (www.mskcc.org/mskcc/html/15938.cfm). Although the first
published MKSCC nomogram for patients with positive SLN has been validated by
numerous centers, clinicians were unlikely to change their surgical plan based on
nomogram results [77]. Additionally, this model seems not reliable predictive for
positive non-SLN in cases with micrometastatic positive SLN [78].
Micrometastases
The SLN is the most likely site of regional metastasis, and gives pathologists the
opportunity to concentrate detection techniques on one or a few lymph nodes. The
use of step sectioning and immunohistochemistry for SLN analysis results in a higher
detection rate of micrometastases (International Union Against Cancer classification:
pN1mi, >0.2 mm to <2.0 mm) and isolated tumor cells (pN0(i+), <0.2mm) [79-81].
Pathologists should follow published recommendations for the management of SLN
[2, 82, 83].
Breast cancer micrometastases have lower tumor proliferation rates and
angiogenesis than breast cancer macrometastases [84]. These characteristics may
explain a discrimination of micro- and macrometastases. The distinction between
pN1mi and pN0(i+) depends on 0.1 mm. Why should 0.1 mm in tumor cluster size
determine different prognosis? Any tumor spread to axillary lymph nodes indicates
16
that the tumor cells are able to metastasize. Therefore the meaningfulness of
distinction between pN1mi and pN0(i+) and also pN1a must be challenged [85].
Tan et al. reported a significant decreased disease-free and overall survival for
pN0(i+) patients (HR=1.7 [95% CI: 1.0-2.9]) compared to pN0(i-) in a retrospective
analysis with 17.6 years follow-up [86]. Many studies confirmed that the occurrence
of pN1mi and pN0(i+) depends on the tumor biology and the location of
micrometastases (sinusal versus parenchymal) [66, 87-94]. However, the clinical
relevance and therapeutic implications of pN1mi and pN0(i+) in the SLN remain a
matter of debate.
A recent meta-analysis including 297,533 patients from 58 studies published
between 1977 and August 2008 divided patients into three categories according to
the method of pathological assessment of the lymph nodes: cohort studies with
single-section examination (n=285,638 patients), occult metastases studies with
retrospective examination of negative lymph nodes by step sectioning and/or
immunohistochemistry (n=7,740 patients), and SLNB studies with intensified work-up
of the sentinel but not of the non-SLN (n=4,155 patients) [95]. In the cohort studies,
axillary lymph node metastases of 2 mm or less in diameter were associated with
poorer overall survival (HR=1.44, 95% CI: 1.29-1.62), occult metastases were
associated with poorer 5-year disease-free survival (RR=1.55, 95% CI: 1.32-1.82)
and overall survival (RR=1.45, 95% CI: 1.11-1.88), although these endpoints were
not consistently assessed in multivariable analyses. SLNB studies are limited by
small patient numbers, SLNB alone or complete ALND in pN1mi and/or pN0(i+),
different systemic treatments, and short follow-up.
As shown in table 5, nine of 12 studies reporting survival outcome, demonstrate no
associations between occult metastases and overall survival. The MIRROR study
(Micrometastases and Isolated tumor cells: Relevant and Robust or Rubbish) has
17
found pN0(i+) and also pN1mi to be indicators of poor prognosis compared to pN0
[89]. The MIRROR-trial is also a retrospective cohort study including only patients
with favorable tumor characteristics for whom adjuvant systemic treatment was not
indicated according to the Dutch treatment guidelines. ITCs or micrometastases in
regional lymph nodes were associated with a reduced 5-year rate of disease-free
survival among patients who did not receive adjuvant therapy (HR 1.5 [95% CI: 1.15-
1.94] for ITC and HR 1.56 [95% CI: 1.15-2.12] for pN1mi). In patients with ITC or
micrometastases who received adjuvant therapy, disease-free survival was
significantly improved as compared with the node-positive, no-adjuvant-therapy
cohort [89].
A recent study showed that the actual rate of positive non-SLNs for patients with SLN
micrometastases or ITCs who underwent completion ALND was significantly less
than that predicted by the MSKCC nomogram [104]. The rate of axillary recurrence
for included 116 patients with stage I to III breast cancer was negligible, regardless of
the extent of axillary staging. The NSABP trial B-32 investigated the clinical
significance of occult metastatic disease in selected sentinel nodes primary
pathologically negative [96]. Occult metastases were detected in 15.9% of 3,887
patients: 11.1% with ITC clusters, 4.4% with micrometastases, and 0.4% with
macrometastases. Occult metastases were an independent prognostic variable;
however, the magnitude of the difference in overall survival at 5 years was small
(94.6% with and 95.8% without detectable metastases). The authors conclude, that
identification of occult metastases does not appear to be clinically useful for patients
with newly diagnosed disease in whom systemic therapy can be recommended on
the basis of the characteristics of the primary tumor.
Do lymph node metastases metastasize?
18
The key question, whether regional lymphatic metastases can further metastasize to
distant organs like the liver, lung or bone or not, is currently unanswered [105]. The
process of metastatic spread is highly complex involving more than 250 genes,
multiple mutations and cell evolutions, and is organotropic [105-109]. Metastatic
spread of tumor cells to regional lymph nodes via lymphatic vessels is not a passive,
but a highly complex active process with specific interaction of tumor cells and
lymphatic tissue [110]. If radio-labeled tumor cells are injected into the afferent nodal
lymphatic vessel, they rapidly appear in the efferent lymphatic vessel and the
thoracic duct, showing, that the lymph node is not only a simple cell-filter [111].
Metastasis to lymph nodes is positively correlated to enhanced peritumoral
lymphangiogenesis, which results from expression of vascular endothelial growth
factor (VEGF)-C. VEGF-C binds to the VEGF receptor (VEGFR)-3 on lymphatic
endothelial cells and induces lymphangiogenesis [112, 113]. High levels of VEGF-C
and VEGFR-3 are not only associated with an increase of lymph node metastasis but
also with poor overall survival without affecting the growth of the primary tumor.
VEGF-C-induced lymphangiogenesis in SLN promotes tumor metastasis spread to
distant sites [114, 115]. These results implicate the possibility of further metastasis to
distant organs from the lymph nodes via the thoracic duct. According to the soil and
seed hypothesis of Paget the fate of a circulating tumor cell is also strongly controlled
by the organ of metastasis resting upon complex cell-cell and cell-matrix interactions.
Key players in lymph node metastasis are the chemokines CXCL12 and CCL21,
produced by the lymphatic tissue. They are attractants for tumor cells which express
the chemokine receptors CXCR4 or CCR7 [110]. High expression of these receptors
is associated with higher incidence of lymph node metastasis in breast cancer
patients [116, 117]. A significant number of patients show discordant quantitative
expression of molecular markers between primary and nodal disease indicating a
19
organotropy [118]. This concept of organ site specificity corroborates the hypothesis,
that lymph node metastases do not further spread or if they do only into lymph nodes
down stream the metastatic one. The lack of lung metastases in patients with a
palliative peritoneo-venous shunt in cases of malignant ascites impressively
emphasizes this hypothesis [119]. But as a result of genetic instability, metastases
that are initially of clonal origin, continuously acquire a genetic heterogeneity which
could result in metastatic transformation of some cells, that permit a further spread to
distant sites [119]. However, animal studies with inhibition of lymph node metastases
are inconsistent and do not clearly confirm the hypothesis of a “metachronous
seeding” [120].
It is widely accepted that also small tumors could early spread to distant sites.
Micrometastases have occurred in 20-40% of carcinomas in the absence of other
detectable signs of spread [88, 121]. Micrometastases as well as the number of
circulating tumor cells are associated with the incidence of distant metastases as well
as with the overall survival of breast cancer patients [122-124]. For patient’s
prognosis, these parameters of tumor spread could be more relevant than the
likelihood of a metastasis from lymph nodes [125, 126]. Therefore we need effective
treatment strategies for these probably stem cell like tumor cells [127-130]. In
conclusion, there is increasing doubt, that lymph node metastasis are able to
metastasize. The life threatening effect of lymph node metastases is overestimated.
Conclusion:
Increasing data suggest that surgical management of the axilla by SLNB or ALND
has no influence on recurrence-free and overall survival in all breast cancer patients.
However, sample size and length of follow-up are substantially different between
cited studies, so that definitive conclusions can not be made. Case reports suggest
20
that patients with axillary metastases alone after SLNB, who had undergone delayed
ALND, have no prognostic disadvantage. With the exception of the ACOSOG Z0011
trial, there are no data from large randomized clinical trials that support or refuse this
hypothesis. There is high evidence to avoid any axillary surgery in elderly patients
with favorable tumor biology (older than 60 years, pT1, G1-2, hormone sensitivity, no
clinically and sonographically involved axillary lymph nodes).
It might be academically interesting to discuss about micrometastases and
immunohistochemically detected tumor cells or tumor cell clusters in the SLN. On the
other hand, we urgently need to initiate randomized clinical trials to avoid any axillary
surgery in most patients with breast cancer and no clinically considerable enlarged
axillary lymph nodes. As the ACOSOG Z0011 trial has shown, patients can be
motivated to participate in randomized trials with restricted axillary surgery.
Nonetheless, a rational and evidence-based approach to the management of
clinically and sonographically N0 patients with planned BCS and limited tumor size is
needed now. Risk estimates using the MSKCC nomogram to predict likelihood of
SLN metastases may be helpful for the decision regarding pro or contra axillary
surgery outside from clinical trials. In patients with BCS without any axillary surgery
an additional regional treatment effect can be expected by postoperative external
beam radiotherapy because traditionally defined tangential breast fields include the
majority (60%-90%) of level I lymph nodes [62].
Surgeons, radiologists, and pathologists should work together to avoid unnecessary
axillary surgery. There is an urgent need for simple but reproducible and validated
sonographic criteria to categorize patients as cN0/iN0 (imaging N0) correctly in the
preoperative setting. Future studies should also include patients with total
mastectomy and favorable tumor biology to prove the value of axillary clearance for
local recurrence and survival rates. It took some decades to replace the radical
21
Halsted theory by new paradigms including BCS. Similarly, the performance of
axillary surgery has to be questioned.
22
References
1. Schwartz GF, Guiliano AE, Veronesi U: Proceeding of the consensus conference
of the role of sentinel lymph node biopsy in carcinoma or the breast April 19-
22, 2001, Philadelphia, PA, USA. Breast J 8:124-138, 2002
2. Lyman GH, Giuliano AE, Somerfield MR et al: American Society of Clinical
Oncology guideline recommendations for sentinel lymph node biopsy in early-
stage breast cancer. J Clin Oncol 23:7703-7720, 2005
3. 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 11:927-933, 2010
4. Crane-Okada R, Wascher RA, Elashoff D et al: Long-term morbidity of sentinel
node biopsy versus complete axillary dissection for unilateral breast cancer.
Ann Surg Oncol 15:1996-2005, 2008
5. AGO: Diagnostic and treatment of early and metastatic breast cancer.
http://www.ago-online.org. 2011
6. NCCN (National Comprehensive Cancer Network). NCCN Practice Guidelines in
Oncology: Breast Cancer. Version 02.2011. http://www.nccn.org. 2011.
7. Cochrane Collaboration. http://www.cochrane.org. 2010.
8. Lanng C, Hoffmann J, Galatius H et al: Assessment of clinical palpation of the
axilla as a criterion for performing the sentinel node procedure in breast
cancer. Eur J Surg Oncol 33:281-284, 2007
23
9. Specht MC, Fey JV, Borgen PI et al: Is the clinically positive axilla in breast cancer
really a contraindication to sentinel lymph node biopsy? J Am Coll Surg
200:10-14, 2005
10. Mathijssen IM, Strijdhorst H, Kiestra SK et al: Added value of ultrasound in
screening the clinically negative axilla in breast cancer. J Surg Oncol 94:364-
367, 2006
11. Zgajnar J, Hocevar M, Podkrajsek M et al: Patients with preoperatively
ultrasonically uninvolved axillary lymph nodes: a distinct subgroup of early
breast cancer patients. Breast Cancer Res Treat 97:293-299, 2006
12. Boughey JC, Moriarty JP, Degnim AC et al: Cost modeling of preoperative
axillary ultrasound and fine-needle aspiration to guide surgery for invasive
breast cancer. Ann Surg Oncol 17:953-958, 2010
13. Park SH, Kim MJ, Park BW et al: Impact of preoperative ultrasonography and
fine-needle aspiration of axillary lymph nodes on surgical management of
primary breast cancer. Ann Surg Oncol 18:738-744, 2011
14. Baruah BP, Goyal A, Young P et al: Axillary node staging by ultrasonography and
fine-needle aspiration cytology in patients with breast cancer. Br J Surg
97:680-683, 2010
15. Alvarez S, Anorbe E, Alcorta P et al: Role of sonography in the diagnosis of
axillary lymph node metastases in breast cancer: a systematic review. AJR
Am J Roentgenol 186:1342-1348, 2006
16. Garcia Fernandez A, Fraile M, Gimenez N et al: Use of axillary ultrasound,
ultrasound-fine needle aspiration biopsy and magnetic resonance imaging in
24
the preoperative triage of breast cancer patients considered for sentinel node
biopsy. Ultrasound Med Biol 37:16-22, 2011
17. Mainiero MB, Cinelli CM, Koelliker SL et al: Axillary ultrasound and fine-needle
aspiration in the preoperative evaluation of the breast cancer patient: an
algorithm based on tumor size and lymph node appearance. AJR Am J
Roentgenol 195:1261-1267, 2010
18. Cho N, Moon WK, Han W et al: Preoperative sonographic classification of axillary
lymph nodes in patients with breast cancer: node-to-node correlation with
surgical histology and sentinel node biopsy results. AJR Am J Roentgenol
193:1731-1737, 2009
19. Steppan I, Reimer D, Muller-Holzner E et al: Breast cancer in women: evaluation
of benign and malignant axillary lymph nodes with contrast-enhanced
ultrasound. Ultraschall Med 31:63-67, 2010
20. Sloka JS, Hollett PD, Mathews M: A quantitative review of the use of FDG-PET in
the axillary staging of breast cancer. Med Sci Monit 13:RA37-46, 2007
21. Peare R, Staff RT, Heys SD: The use of FDG-PET in assessing axillary lymph
node status in breast cancer: a systematic review and meta-analysis of the
literature. Breast Cancer Res Treat 123:281-290, 2010
22. Straver ME, Aukema TS, Olmos RA et al: Feasibility of FDG PET/CT to monitor
the response of axillary lymph node metastases to neoadjuvant chemotherapy
in breast cancer patients. Eur J Nucl Med Mol Imaging 37:1069-1076, 2010
23. Ahn JH, Son EJ, Kim JA et al: The role of ultrasonography and FDG-PET in
axillary lymph node staging of breast cancer. Acta Radiol 51:859-865, 2010
25
24. Harada T, Tanigawa N, Matsuki M et al: Evaluation of lymph node metastases of
breast cancer using ultrasmall superparamagnetic iron oxide-enhanced
magnetic resonance imaging. Eur J Radiol 63:401-407, 2007
25. Stadnik TW, Everaert H, Makkat S et al: Breast imaging. Preoperative breast
cancer staging: comparison of USPIO-enhanced MR imaging and 18F-
fluorodeoxyglucose (FDG) positron emission tomography (PET) imaging for
axillary lymph node staging--initial findings. Eur Radiol 16:2153-2160, 2006
26. Murray AD, Staff RT, Redpath TW et al: Dynamic contrast enhanced MRI of the
axilla in women with breast cancer: comparison with pathology of excised
nodes. Br J Radiol 75:220-228, 2002
27. Michel SC, Keller TM, Frohlich JM et al: Preoperative breast cancer staging: MR
imaging of the axilla with ultrasmall superparamagnetic iron oxide
enhancement. Radiology 225:527-536, 2002
28. Memarsadeghi M, Riedl CC, Kaneider A et al: Axillary lymph node metastases in
patients with breast carcinomas: assessment with nonenhanced versus uspio-
enhanced MR imaging. Radiology 241:367-377, 2006
29. Suzuma T, Sakurai T, Yoshimura G et al: MR-axillography oriented surgical
sampling for assessment of nodal status in the selection of patients with
breast cancer for axillary lymph nodes dissection. Breast Cancer 9:69-74,
2002
30. Stets C, Brandt S, Wallis F et al: Axillary lymph node metastases: a statistical
analysis of various parameters in MRI with USPIO. J Magn Reson Imaging
16:60-68, 2002
26
31. Kitagawa K, Sakuma H, Ishida N et al: Contrast-enhanced high-resolution MRI of
invasive breast cancer: correlation with histopathologic subtypes. AJR Am J
Roentgenol 183:1805-1809, 2004
32. Bathen TF, Jensen LR, Sitter B et al: MR-determined metabolic phenotype of
breast cancer in prediction of lymphatic spread, grade, and hormone status.
Breast Cancer Res Treat 104:181-189, 2007
33. Gelmon KA: The ongoing debate about nodes. Clin Breast Cancer 10:265-266,
2010
34. Tai P, Yu E, Joseph K: Prognostic significance of number of positive nodes: a
long-term study of one to two nodes versus three nodes in breast cancer
patients. Int J Radiat Oncol Biol Phys 77:180-187, 2010
35. Hatoum HA, Jamali FR, El Saghir NS et al: Ratio between positive lymph nodes
and total excised axillary lymph nodes as an independent prognostic factor for
overall survival in patients with nonmetastatic lymph node-positive breast
cancer. Ann Surg Oncol 16:3388-3395, 2009
36. Crabb SJ, Cheang MC, Leung S et al: Basal breast cancer molecular subtype
predicts for lower incidence of axillary lymph node metastases in primary
breast cancer. Clin Breast Cancer 8:249-256, 2008
37. Van Calster B, Vanden Bempt I, Drijkoningen M et al: Axillary lymph node status
of operable breast cancers by combined steroid receptor and HER-2 status:
triple positive tumours are more likely lymph node positive. Breast Cancer Res
Treat 113:181-187, 2009
27
38. Wildiers H, Van Calster B, van de Poll-Franse LV et al: Relationship between age
and axillary lymph node involvement in women with breast cancer. J Clin
Oncol 27:2931-2937, 2009
39. Botteri E, Bagnardi V, Goldhirsch A et al: Axillary lymph node involvement in
women with breast cancer: does it depend on age? Clin Breast Cancer
10:318-321, 2010
40. Kennecke H, Yerushalmi R, Woods R et al: Metastatic behavior of breast cancer
subtypes. J Clin Oncol 28:3271-3277, 2010
41. Voduc KD, Cheang MC, Tyldesley S et al: Breast cancer subtypes and the risk of
local and regional relapse. J Clin Oncol 28:1684-1691, 2010
42. Foulkes WD, Grainge MJ, Rakha EA et al: Tumor size is an unreliable predictor
of prognosis in basal-like breast cancers and does not correlate closely with
lymph node status. Breast Cancer Res Treat 117:199-204, 2009
43. Blows FM, Driver KE, Schmidt MK et al: Subtyping of breast cancer by
immunohistochemistry to investigate a relationship between subtype and short
and long term survival: a collaborative analysis of data for 10,159 cases from
12 studies. PLoS Med 7:e1000279, 2010
44. Huber KE, Carey LA, Wazer DE: Breast cancer molecular subtypes in patients
with locally advanced disease: impact on prognosis, patterns of recurrence,
and response to therapy. Semin Radiat Oncol 19:204-210, 2009
45. van de Vijver MJ, He YD, van't Veer LJ et al: A gene-expression signature as a
predictor of survival in breast cancer. N Engl J Med 347:1999-2009, 2002
28
46. Sorlie T, Perou CM, Tibshirani R et al: Gene expression patterns of breast
carcinomas distinguish tumor subclasses with clinical implications. Proc Natl
Acad Sci U S A 98:10869-10874, 2001
47. Reimer T, Fietkau R, Markmann S et al: How important is the axillary nodal status
for adjuvant treatment decisions at a breast cancer multidisciplinary tumor
board? A survival analysis. Ann Surg Oncol 15:472-477, 2008
48. Martin M, Segui MA, Anton A et al: Adjuvant docetaxel for high-risk, node-
negative breast cancer. N Engl J Med 363:2200-2210, 2010
49. Untch M, Gelber RD, Jackisch C et al: Estimating the magnitude of trastuzumab
effects within patient subgroups in the HERA trial. Ann Oncol 19:1090-1096,
2008
50. Engel J, Lebeau A, Sauer H et al: Are we wasting our time with the sentinel
technique? Fifteen reasons to stop axilla dissection. Breast 15:452-455, 2006
51. Moon HG, Han W, Noh DY: Comparable survival between pN0 breast cancer
patients undergoing sentinel node biopsy and extensive axillary dissection: a
report from the Korean Breast Cancer Society. J Clin Oncol 28:1692-1699,
2010
52. Kell MR, Burke JP, Barry M et al: Outcome of axillary staging in early breast
cancer: a meta-analysis. Breast Cancer Res Treat 120:441-447, 2010
53. Land SR, Kopec JA, Julian TB et al: Patient-reported outcomes in sentinel node-
negative adjuvant breast cancer patients receiving sentinel-node biopsy or
axillary dissection: National Surgical Adjuvant Breast and Bowel Project phase
III protocol B-32. J Clin Oncol 28:3929-3936, 2010
29
54. Rudenstam CM, Zahrieh D, Forbes JF et al: Randomized trial comparing axillary
clearance versus no axillary clearance in older patients with breast cancer:
first results of International Breast Cancer Study Group Trial 10-93. J Clin
Oncol 24:337-344, 2006
55. Martelli G, Boracchi P, De Palo M et al: A randomized trial comparing axillary
dissection to no axillary dissection in older patients with T1N0 breast cancer:
results after 5 years of follow-up. Ann Surg 242:1-6, 2005
56. Veronesi U, Orecchia R, Zurrida S et al: Avoiding axillary dissection in breast
cancer surgery: a randomized trial to assess the role of axillary radiotherapy.
Ann Oncol 16:383-388, 2005
57. Martelli G, Miceli R, Daidone MG et al: Axillary Dissection Versus No Axillary
Dissection in Elderly Patients with Breast Cancer and No Palpable Axillary
Nodes: Results After 15 Years of Follow-Up. Ann Surg Oncol 18:125-133,
2011
58. Fredriksson I, Liljegren G, Arnesson LG et al: Consequences of axillary
recurrence after conservative breast surgery. Br J Surg 89:902-908, 2002
59. Louis-Sylvestre C, Clough K, Asselain B et al: Axillary treatment in conservative
management of operable breast cancer: dissection or radiotherapy? Results of
a randomized study with 15 years of follow-up. J Clin Oncol 22:97-101, 2004
60. Bourez R, Rutgers E, van de Velde C: Will we need lymph node dissection at all
in the future? Clin Breast Cancer 3:315-322, 2002
30
61. Straver ME, Meijnen P, van Tienhoven G et al: Sentinel node identification rate
and nodal involvement in the EORTC 10981-22023 AMAROS trial. Ann Surg
Oncol 17:1854-1861, 2010
62. Evans SB, Gass J, Wazer DE: Management of the axilla after the finding of a
positive sentinel lymph node: a proposal for an evidence-based risk-adapted
algorithm. Am J Clin Oncol 31:293-299, 2008
63. Krag DN, Anderson SJ, Julian TB et al: Technical outcomes of sentinel-lymph-
node resection and conventional axillary-lymph-node dissection in patients
with clinically node-negative breast cancer: results from the NSABP B-32
randomised phase III trial. Lancet Oncol 8:881-888, 2007
64. Canavese G, Catturich A, Vecchio C et al: Prognostic role of lymph-node level
involvement in patients undergoing axillary dissection for breast cancer. Eur J
Surg Oncol 24:104-109, 1998
65. Erb KM, Julian TB: Completion of axillary dissection for a positive sentinel node:
necessary or not? Curr Oncol Rep 11:15-20, 2009
66. Wo JY, Taghian AG, Nguyen PL et al: The association between biological
subtype and isolated regional nodal failure after breast-conserving therapy. Int
J Radiat Oncol Biol Phys 77:188-196, 2010
67. Wapnir IL, Anderson SJ, Mamounas EP et al: Prognosis after ipsilateral breast
tumor recurrence and locoregional recurrences in five National Surgical
Adjuvant Breast and Bowel Project node-positive adjuvant breast cancer trials.
J Clin Oncol 24:2028-2037, 2006
31
68. Giuliano AE, McCall L, Beitsch P et al: Locoregional recurrence after sentinel
lymph node dissection with or without axillary dissection in patients with
sentinel lymph node metastases: the American College of Surgeons Oncology
Group Z0011 randomized trial. Ann Surg 252:426-432, 2010
69. Giuliano AE, Hunt KK, Ballman KV et al: Axillary dissection vs no axillary
dissection in women with invasive breast cancer and sentinel node
metastasis. JAMA 305:569-575, 2011
70. Yi M, Giordano SH, Meric-Bernstam F et al: Trends in and outcomes from
sentinel lymph node biopsy (SLNB) alone vs. SLNB with axillary lymph node
dissection for node-positive breast cancer patients: experience from the SEER
database. Ann Surg Oncol 17:S343-S351, 2010
71. Bilimoria KY, Bentrem DJ, Hansen NM et al: Comparison of sentinel lymph node
biopsy alone and completion axillary lymph node dissection for node-positive
breast cancer. J Clin Oncol 27:2946-2953, 2009
72. Veronesi U, Marubini E, Mariani L et al: The dissection of internal mammary
nodes does not improve the survival of breast cancer patients. 30-year results
of a randomised trial. Eur J Cancer 35:1320-1325, 1999
73. Janni W, Rack B, Sommer H et al: Intra-mammary tumor location does not
influence prognosis but influences the prevalence of axillary lymph-node
metastases. J Cancer Res Clin Oncol 129:503-510, 2003
74. Sinha PS, Thrush S, Bendall S et al: Does radical surgery to the axilla give a
survival advantage in more severe breast cancer? Eur J Cancer 38:1474-
1477, 2002
32
75. Van Zee KJ, Manasseh DM, Bevilacqua JL et al: A nomogram for predicting the
likelihood in breast cancer patients with a positive sentinel node biopsy. Ann
Surg Oncol 10:1140-1151, 2003
76. Bevilacqua JL, Kattan MW, Fey JV et al: Doctor, what are my chances of having
a positive sentinel node? A validated nomogram for risk estimation. J Clin
Oncol 25:3670-3679, 2007
77. Specht MC, Kattan MW, Fey J et al: Predicting nonsentinel node status after
positive sentinel lymph node biopsy for breast cancer: clinicians versus
nomogram. Ann Surg Oncol 12:654-659, 2005
78. Alran S, De Rycke Y, Fourchotte V et al: Validation and limitations of use of a
breast cancer nomogram predicting the likelihood of non-sentinel node
involvement after positive sentinel node biopsy. Ann Surg Oncol 14:2195-
2201, 2007
79. Cserni G, Gregori D, Merletti F et al: Meta-analysis of non-sentinel node
metastases associated with micrometastatic sentinel nodes in breast cancer.
Br J Surg 91:1245-1252, 2004
80. Langer I, Guller U, Viehl CT et al: Axillary lymph node dissection for sentinel
lymph node micrometastases may be safely omitted in early-stage breast
cancer patients: long-term outcomes of a prospective study. Ann Surg Oncol
16:3366-3374, 2009
81. Sahin AA, Guray M, Hunt KK: Identification and biologic significance of
micrometastases in axillary lymph nodes in patients with invasive breast
cancer. Arch Pathol Lab Med 133:869-878, 2009
33
82. Cserni G, Amendoeira I, Apostolikas N et al: Pathological work-up of sentinel
lymph nodes in breast cancer. Review of current data to be considered for the
formulation of guidelines. Eur J Cancer 39:1654-1667, 2003
83. Turner RR, Weaver DL, Cserni G et al: Nodal stage classification for breast
carcinoma: improving interobserver reproducibility through standardized
histologic criteria and image-based training. J Clin Oncol 26:258-263, 2008
84. Klauber-DeMore N, Van Zee KJ, Linkov I et al: Biological behavior of human
breast cancer micrometastases. Clin Cancer Res 7:2434-2439, 2001
85. de Mascarel I, MacGrogan G, Debled M et al: Distinction between isolated tumor
cells and micrometastases in breast cancer: is it reliable and useful? Cancer
112:1672-1678, 2008
86. Tan LK, Giri D, Hummer AJ et al: Occult axillary node metastases in breast
cancer are prognostically significant: results in 368 node-negative patients
with 20-year follow-up. J Clin Oncol 26:1803-1809, 2008
87. Yegiyants S, Romero LM, Haigh PI et al: Completion axillary lymph node
dissection not required for regional control in patients with breast cancer who
have micrometastases in a sentinel node. Arch Surg 145:564-569, 2010
88. Gerber B, Krause A, Muller H et al: Simultaneous immunohistochemical detection
of tumor cells in lymph nodes and bone marrow aspirates in breast cancer and
its correlation with other prognostic factors. J Clin Oncol 19:960-971, 2001
89. de Boer M, van Deurzen CH, van Dijck JA et al: Micrometastases or isolated
tumor cells and the outcome of breast cancer. N Engl J Med 361:653-663,
2009
34
90. van Diest PJ, de Boer M, van Deurzen CH et al: Micrometastases and isolated
tumor cells in breast cancer are indeed associated with poorer outcome. J Clin
Oncol 28:e140-e142, 2010
91. Hansen NM, Grube B, Ye X et al: Impact of micrometastases in the sentinel node
of patients with invasive breast cancer. J Clin Oncol 27:4679-4684, 2009
92. Reed J, Rosman M, Verbanac KM et al: 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 208:333-340, 2009
93. van der Ploeg I, Nieweg OE, van Rijk MC et al: 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 34:1277-1284,
2008
94. Masci G, Di Tommaso L, Del Prato I et al: Sinusal localization of nodal
micrometastases is a prognostic factor in breast cancer. Ann Oncol 21:1228-
1232, 2010
95. de Boer M, van Dijck JA, Bult P et al: Breast cancer prognosis and occult lymph
node metastases, isolated tumor cells, and micrometastases. J Natl Cancer
Inst 102:410-425, 2010
96. Weaver DL, Ashikaga T, Krag DN et al: Effect of occult metastases on survival in
node-negative breast cancer. N Engl J Med 364:412-421, 2011
35
97. Maaskant-Braat AJ, van de Poll-Franse LV, Voogd AC et al: Sentinel node
micrometastases in breast cancer do not affect prognosis: a population-based
study. Breast Cancer Res Treat 2010 Aug 3 [Epub ahead of print]
98. Montagna E, Viale G, Rotmensz N et al: Minimal axillary lymph node involvement
in breast cancer has different prognostic implications according to the staging
procedure. Breast Cancer Res Treat 118:385-394, 2009
99. Gobardhan PD, Elias SG, Madsen EV et al: Prognostic value of micrometastases
in sentinel lymph nodes of patients with breast carcinoma: a cohort study. Ann
Oncol 20:41-48, 2009
100. Cox CE, Kiluk JV, Riker AI et al: Significance of sentinel lymph node
micrometastases in human breast cancer. J Am Coll Surg 206:261-268, 2008
101. Nagashima T, Sakakibara M, Nakano S et al: Sentinel node micrometastasis
and distant failure in breast cancer patients. Breast Cancer 13:186-191, 2006
102. Imoto S, Ochiai A, Okumura C et al: Impact of isolated tumor cells in sentinel
lymph nodes detected by immunohistochemical staining. Eur J Surg Oncol
32:1175-1179, 2006
103. Fan YG, Tan YY, Wu CT et al: The effect of sentinel node tumor burden on non-
sentinel node status and recurrence rates in breast cancer. Ann Surg Oncol
12:705-711, 2005
104. Cyr A, Gillanders WE, Aft RL et al: Micrometastatic disease and isolated tumor
cells as a predictor for additional breast cancer axillary metastatic burden. Ann
Surg Oncol 17:S303-S311, 2010
36
105. Tait CR, Dodwell D, Horgan K: Do metastases metastasize? J Pathol 203:515-
518, 2004
106. Bernards R, Weinberg RA: A progression puzzle. Nature 418:823, 2002
107. Lu X, Yan CH, Yuan M et al: In vivo dynamics and distinct functions of hypoxia
in primary tumor growth and organotropic metastasis of breast cancer. Cancer
Res 70:3905-3914, 2010
108. Lu X, Kang Y: Organotropism of breast cancer metastasis. J Mammary Gland
Biol Neoplasia 12:153-162, 2007
109. Smid M, Wang Y, Zhang Y et al: Subtypes of breast cancer show preferential
site of relapse. Cancer Res 68:3108-3114, 2008
110. Ran S, Volk L, Hall K et al: Lymphangiogenesis and lymphatic metastasis in
breast cancer. Pathophysiology 17:229-251, 2010
111. Fisher B, Fisher ER: Transmigration of lymph nodes by tumor cells. Science
152:1397-1398, 1966
112. Krishnan J, Kirkin V, Steffen A et al: Differential in vivo and in vitro expression of
vascular endothelial growth factor (VEGF)-C and VEGF-D in tumors and its
relationship to lymphatic metastasis in immunocompetent rats. Cancer Res
63:713-722, 2003
113. Hirakawa S, Brown LF, Kodama S et al: VEGF-C-induced lymphangiogenesis in
sentinel lymph nodes promotes tumor metastasis to distant sites. Blood
109:1010-1017, 2007
37
114. Mohammed RA, Green A, El Shikh S et al: Prognostic significance of vascular
endothelial cell growth factors -A, -C and -D in breast cancer and their
relationship with angio- and lymphangiogenesis. Br J Cancer 96:1092-1100,
2007
115. Nakamura Y, Yasuoka H, Tsujimoto M et al: Lymph vessel density correlates
with nodal status, VEGF-C expression, and prognosis in breast cancer. Breast
Cancer Res Treat 91:125-132, 2005
116. Yasuoka H, Tsujimoto M, Yoshidome K et al: Cytoplasmic CXCR4 expression in
breast cancer: induction by nitric oxide and correlation with lymph node
metastasis and poor prognosis. BMC Cancer 8:340, 2008
117. Cabioglu N, Yazici MS, Arun B et al: CCR7 and CXCR4 as novel biomarkers
predicting axillary lymph node metastasis in T1 breast cancer. Clin Cancer
Res 11:5686-5693, 2005
118. Aitken SJ, Thomas JS, Langdon SP et al: Quantitative analysis of changes in
ER, PR and HER2 expression in primary breast cancer and paired nodal
metastases. Ann Oncol 21:1254-1261, 2010
119. Fidler IJ: The pathogenesis of cancer metastasis: the 'seed and soil' hypothesis
revisited. Nat Rev Cancer 3:453-458, 2003
120. Wong SY, Hynes RO: Lymphatic or hematogenous dissemination: how does a
metastatic tumor cell decide? Cell Cycle 5:812-817, 2006
121. Pantel K, Cote RJ, Fodstad O: Detection and clinical importance of
micrometastatic disease. J Natl Cancer Inst 91:1113-1124, 1999
38
122. Wiedswang G, Borgen E, Schirmer C et al: Comparison of the clinical
significance of occult tumor cells in blood and bone marrow in breast cancer.
Int J Cancer 118:2013-2019, 2006
123. Bidard FC, Mathiot C, Delaloge S et al: Single circulating tumor cell detection
and overall survival in nonmetastatic breast cancer. Ann Oncol 21:729-733,
2010
124. Bidard FC, Vincent-Salomon A, Sigal-Zafrani B et al: Prognosis of women with
stage IV breast cancer depends on detection of circulating tumor cells rather
than disseminated tumor cells. Ann Oncol 19:496-500, 2008
125. Schmidt-Kittler O, Ragg T, Daskalakis A et al: From latent disseminated cells to
overt metastasis: genetic analysis of systemic breast cancer progression. Proc
Natl Acad Sci U S A 100:7737-7742, 2003
126. Schardt JA, Meyer M, Hartmann CH et al: Genomic analysis of single
cytokeratin-positive cells from bone marrow reveals early mutational events in
breast cancer. Cancer Cell 8:227-239, 2005
127. Theodoropoulos PA, Polioudaki H, Agelaki S et al: Circulating tumor cells with a
putative stem cell phenotype in peripheral blood of patients with breast cancer.
Cancer Lett 288:99-106, 2010
128. Liu S, Wicha MS: Targeting breast cancer stem cells. J Clin Oncol 28:4006-
4012, 2010
129. McDermott SP, Wicha MS: Targeting breast cancer stem cells. Mol Oncol 2010
39
130. Kakarala M, Wicha MS: Implications of the cancer stem-cell hypothesis for
breast cancer prevention and therapy. J Clin Oncol 26:2813-2820, 2008
Tab. 1: Diagnostic accuracy of FDG-PET in staging axillary nodes. Summarized
results of a meta-analysis [20].
Quality of
Study*
Sensitivity
(%)
Specificity
(%)
Positive
Predictive
Value (%)
Negative
Predictive
Value (%)
A (best)
78
85
80
84
B
67
89
82
78
C
96
84
78
97
D (poor)
78
99
99
76
* depending on number and selection of patients, technical details of examination,
pro- or retrospective design, method of lymph node biopsy.
Tab. 2: Diagnostic accuracy of MRI in staging axillary nodes.
Author
Number
of pts.
Sensitivity
(%)
Specificity
(%)
Positive Predictive
Value (%)
Negative Predictive
Value (%)
Harada (2007)
[24]
33
86
97
91
96
Stadnik (2006)
[25]
10
100
80
80
100
Murray (2002)
[26]
47
100
56
38
100
Michel (2002)
[27]
20
82
100
100
-
Memarsadeqhi
(2006) [28]
22
100
98
98% accuracy
Suzuma
(2002) [29]
62
91
100
91% accuracy
Stets
(2002) [30]
9
63
86
75% accuracy
40
Tab. 3: Reasons for and against knowledge of lymph node status for treatment
decisions.
Contra
Pro
Tumor biology is more important than
nodal status regarding prognosis and
treatment
Treatment mismatch if node status is
unknown (especially in ER-positive,
HER2-negative disease)
Any axillary surgery increases costs
and morbidity
dose-dense and/or dose-intense
chemotherapy in patients with 4 or more
involved lymph nodes
Axillary recurrences are very seldom,
even in node positive patients
Resection of metastases as a potential
source of distant metastases
Delayed surgery of involved lymph
nodes is without overall survival
disadvantages
Tumor volume reduction
Nodal status as an indicator for or
against radiotherapy after mastectomy
Distant metastasis even in pN0 patients
Seldom axillary metastases in patients
with medial tumor location,
pN for staging, study inclusion and
comparison of outcome necessary
Systemic treatment acts also in axillary
lymph node metastases
There is no evidence that lymph node
metastases are able to metastasize
Leaving involved lymph nodes in situ
does not increase axillary recurrences
or metastasis
Tab. 4: Prospective randomized clinical trials comparing axillary lymph node
dissection versus no axillary surgery. Veronesi et al. randomized patients with
no axillary surgery to none further treatment versus radiotherapy of the axilla.
Pts.
N
Follow
-up (mo)
Axillary
recurrences
DFS
OAS
Rudenstam
(2006) [54]
>60 y, pT1-2*
cN0,
473
78
0.9 vs 2.5%
67% vs 66%
p=.7
75% vs 73%;
p=.8
Martelli
(2005) [55]
>65 y, pT1
cN0, Tam
219
60
0 vs 1.8%
95% both
p=.9
96% both
p=.9
Veronesi
(2005) [56]
>45 y, Tumor
<1.2 cm, cN0,
435
63
0.5 vs 1.5%
97 vs 95%
p=.19
99 vs 97%
p=.23
* 42% pts. with tumors >2 cm, y = years, mo = months
DFS = disease-free survival
OAS = overall survival
41
Tab. 5: Summary of studies reporting outcome (follow-up >24 months) of
patients (case numbers >150) with isolated tumor cells pN0(i+) and
micrometastasis pN1mi (International Union Against Cancer classification, >.2 mm
to <2.0 mm) in sentinel lymph node (SLN)
Author
Study design
Accrual period
N
Follow-up
Conclusions
Weaver
(2011)
[96]
prospective,
multicenter
1999-2004
3,887
95.6 mo
Occult metastases (11.1% pN0(i+), 4.4% pN1mi, 0.4%
pN1a) in initial negative SLNs have a small, but
significant impact on DFS, DMFS, and OAS
Maaskant-Braat
(2011) [97]
population-based
1996-2006
6,803
50 mo,
36 mo for
pN0(i+)
pN1mi or pN0(i+) in the SLNB did not convey any
significant OAS difference compared with pN0 pts.
(discrimination between pN1mi and pN0(i+) since 2003)
Langer
(2009) [80]
prospective cohort
1998-2002
150
77 mo
No significant differences for OAS, LRR, and DMFS
between pN0 and pN1mi groups
de Boer
(2009) [89]
population-based
before 2006
2,707
5.1 y
pN(i+) and pN1mi status were associated with reduced
5y-DFS in cases without adjuvant therapy
Montagna
(2009) [98]
prospective cohort
1997-2002
3,158
6.3 y
Among SLNB group, no difference in outcome between
pts. with pN0 or minimal lymph node involvement (ITC,
micrometastasis 0.2-1 mm, micrometastasis 1-2 mm)
Reed
(2009) [92]
prospective,
multicenter
1996-2005
1,259
4.9 y
pN1mi, but not pN0(i+), was associated with additional
positive nodes (27%) and with increased distance
recurrence rate
Gobardhan
(2009) [99]
prospective cohort
1999-2007
703
40 mo
Risk of distant metastases higher in patients with pN1mi
than in pN0 group, but no significant differences in DFS
or OAS between pN0 and pN1mi
Tan (2008) [86]
retrospective
1976-1978
368
17.6 y
Clear trend toward worse outcome (DFS, BCSS) with
increasing tumor cluster size in the lymph nodes
Cox
(2008) [100]
retrospective review
1997-2004
2,381
1.5-2.1 y
Poorer OAS/DFS for pts. with pN1mi compared with pN0
pts.; additional axillary disease in 9.3% of pts. with
pN0(i+)
Nagashima
(2006) [101]
prospective cohort
1999-2004
375
30 mo
No difference in DMFS between pN0 and pN1mi pts.
Imoto (2006)
[102]
retrospective cohort
1998-2000
165
73 mo
No difference in 6y-DFS between pN0(i+)/pN1mi group
and pN0 pts.
Fan (2005) [103]
retrospective
1997-2002
390
31.1 mo
No difference in LRR between pN0 and pN1mi pts.
OAS = Overall survival, DMFS = Distant metastases-free survival, DFS = Disease-free
survival; BCSS = Breast cancer-specific survival, pts. = patients; LRR = Local recurrence
rate; mo = months; y = years; ITC = isolated tumor cells
... Deeskalation der axillären operativen Therapie Die axilläre Lymphknotendissektion ist einer der Hauptgründe für Langzeitfolgen wie Ödeme oder Funktionseinschränkung des Armes der operierten Seite. Es existieren jedoch bereits einige Studien, die aufgezeigt haben, dass eine gute lokoregionäre Tumorkontrolle ebenfalls erreicht werden kann, wenn keine axilläre Lymphknotendissektion durchgeführt wird [38,39]. In der INSE-MA-Studie der GBG, AGO-B und ABCSG wurde diese Fragestellung in einem randomisierten Studiendesign (▶ Abb. 1) untersucht. ...
... In der INSE-MA-Studie der GBG, AGO-B und ABCSG wurde diese Fragestellung in einem randomisierten Studiendesign (▶ Abb. 1) untersucht. Nun wurden die Lebensqualitätsdaten dieser Studie vorgestellt, die mittels der Fragebögen EORTC-QLQ-C30 und BR23 erhoben wurden [38]. ...
... In Bezug auf die 1. Randomisation konnte durch das Weglassen der SLNB keine klinisch relevante Verbesserung der globalen Lebensqualität erreicht werden [38]. Jedoch zeigten sich deutliche Unterschiede in Bezug auf den Arm-Symptom-Score des EORTC-QLQ-BR23-Fragebogens. ...
Article
Full-text available
Evidence relating to the treatment of breast cancer patients with early-stage disease has increased significantly in the past year. Abemaciclib, olaparib, and pembrolizumab are new drugs with good efficacy in the relevant patient groups. However, some questions remain unanswered. In particular, it remains unclear which premenopausal patients with hormone receptor-positive breast cancer should be spared unnecessary treatment. The question of the degree to which chemotherapy exerts a direct cytotoxic effect on the tumor or reduces ovarian function through chemotherapy could be of key importance. This group of patients could potentially be spared chemotherapy. New, previously experimental biomarker analysis methods, such as spatial analysis of gene expression (spatial transcriptomics) are gradually finding their way into large randomized phase III trials, such as the NeoTRIP trial. This in turn leads to a better understanding of the predictive factors of new therapies, for example immunotherapy. This review summarizes the scientific innovations from recent congresses such as the San Antonio Breast Cancer Symposium 2021 but also from recent publications.
... In the past, routine axillary lymph www.nature.com/scientificreports/ node dissection in BC surgery had no prognostic value for patients in stage N0 but produced a series of complications, affecting their quality of life 30,31 . In the 2010s, the American Association of Surgeons Oncology Group (ACOSOG) Z0011 trial changed the clinical model of axillary treatment, for BC patients with cN0 disease, when SLNB instead of ALND became a possibility [32][33][34] . ...
Article
Full-text available
The status of axillary lymph node metastases determines the treatment and overall survival of breast cancer (BC) patients. Three-dimensional (3D) assessment methods have advantages for spatial localization and are more responsive to morphological changes in lymph nodes than two-dimensional (2D) assessment methods, and we speculate that methods developed using 3D reconstruction systems have high diagnostic efficacy. This exploratory study included 43 patients with histologically confirmed BC diagnosed at Second Xiangya Hospital of Central South University between July 2017 and August 2020, all of whom underwent preoperative CT scans. Patients were divided into a training cohort to train the model and a validation cohort to validate the model. A 3D axillary lymph node atlas was constructed on a 3D reconstruction system to create various methods of assessing lymph node metastases for a comparison of diagnostic efficacy. Receiver operating characteristic (ROC) curve analysis was performed to assess the diagnostic values of these methods. A total of 43 patients (mean [SD] age, 47 [10] years) met the eligibility criteria and completed 3D reconstruction. An axillary lymph node atlas was established, and a correlation between lymph node sphericity and lymph node metastasis was revealed. By continuously fitting the size and characteristics of axillary lymph nodes on the 3D reconstruction system, formulas and models were established to determine the presence or absence of lymph node metastasis, and the 3D method had better sensitivity for axillary lymph node assessment than the 2D method, with a statistically significant difference in the correct classification rate. The combined diagnostic method was superior to a single diagnostic method, with a 92.3% correct classification rate for the 3D method combined with ultrasound. In addition, in patients who received neoadjuvant chemotherapy (NAC), the correct classification rate of the 3D method (72.7%) was significantly higher than that of ultrasound (45.5%) and CT (54.5%). By establishing an axillary lymph node atlas, the sphericity formula and model developed with the 3D reconstruction system achieve a high correct classification rate when combined with ultrasound or CT and can also be applied to patients receiving NAC.
... The higher the stage is, the worse the prognosis[28]. In the past, routine axillary lymph node dissection in BC surgery had no prognostic value for patients in stage N0 but produced a series of complications, affecting their quality of life [29,30]. In the 2010s, the American Association of Surgeons Oncology Group (ACOSOG) Z0011 trial changed the clinical model of axillary treatment, for BC patients with cN0 disease, when SLNB instead of ALND became a possibility [31][32][33]. ...
Preprint
Full-text available
Background: The status of axillary lymph node metastases determines the treatment and overall survival of breast cancer (BC) patients. Three-dimensional (3D) assessment methods have advantages for spatial localization and are more responsive to morphological changes in lymph nodes than two-dimensional (2D) assessment methods, and we speculate that methods developed using 3D reconstruction systems have high diagnostic efficacy. Methods: This exploratory study included 43 patients with histologically confirmed BC diagnosed at Second Xiangya Hospital of Central South University between July 2017 and August 2020, all of whom underwent preoperative CT scans. Patients were divided into a training cohort to train the model and a validation cohort to validate the model. A 3D axillary lymph node atlas was constructed on a 3D reconstruction system to create various methods of assessing lymph node metastases for a comparison of diagnostic efficacy. Receiver operating characteristic (ROC) curve analysis was performed to assess the diagnostic values of these methods. Results: A total of 43 patients (mean [SD] age, 47 [10] years) met the eligibility criteria and completed 3D reconstruction. An axillary lymph node atlas was established, and a correlation between lymph node sphericity and lymph node metastasis was revealed. By continuously fitting the size and characteristics of axillary lymph nodes on the 3D reconstruction system, formulas and models were established to determine the presence or absence of lymph node metastasis, and the 3D method had better sensitivity for axillary lymph node assessment than the 2D method, with a statistically significant difference in the correct classification rate. The combined diagnostic method was superior to a single diagnostic method, with a 92.3% correct classification rate for the 3D method combined with ultrasound. In addition, in patients who received neoadjuvant chemotherapy (NAC), the correct classification rate of the 3D method (72.7%) was significantly higher than that of ultrasound (45.5%) and CT (54.5%). Conclusion: By establishing an axillary lymph node atlas, the sphericity formula and model developed with the 3D reconstruction system achieve a high correct classification rate when combined with ultrasound or CT and can also be applied to patients receiving NAC.
Article
Zusammenfassung Die Erkenntnisse über die Behandlung von Mammakarzinompatientinnen mit frühen Krankheitsstadien haben im letzten Jahr deutlich zugenommen. Abemaciclib, Olaparib und Pembrolizumab sind neue Medikamente mit einer guten Wirksamkeit bei den entsprechenden Patientinnengruppen. Jedoch sind einige Fragestellungen nach wie vor unbeantwortet. Insbesondere welchen Patientinnen unnötige Therapien erspart werden können, ist bei prämenopausalen Patientinnen mit einem hormonrezeptorpositiven Mammakarzinom weiterhin unklar. Die Frage, inwieweit eine Chemotherapie einen direkten zytotoxischen Effekt auf den Tumor hat oder eher dadurch wirkt, dass die Ovarfunktion durch die Chemotherapie reduziert wird, könnte wegweisend sein. Dieser Patientinnengruppe kann möglicherweise eine Chemotherapie erspart bleiben. Neue, bislang experimentelle Biomarker-Analysemethoden, wie die räumliche Analyse der Genexpression (spatial transcriptomics), halten nach und nach Einzug in die großen randomisierten Phase-III-Studien, wie die NeoTRIPStudie. Dies führt wiederum zum besseren Verständnis der prädiktiven Faktoren neuer Therapien, zum Beispiel der Immuntherapie. Diese Übersichtsarbeit fasst die wissenschaftlichen Neuerungen der aktuellen Kongresse wie dem San Antonio Breast Cancer Symposium 2021, aber auch von kürzlich veröffentlichten Publikationen zusammen.
Article
Zusammenfassung Über viele Jahrzehnte war die komplette Ausräumung der axillären Lymphknoten im Sinne einer Axilladissektion ein Standardverfahren in der Therapie des Mammakarzinom. Die Zielsetzung lag in der Bestimmung des histologischen Nodalstatus für die Festlegung der adjuvanten Therapie sowie in der Sicherung der lokoregionären Tumorkontrolle. Neben der Diskussion zur Optimierung der Therapiestrategien in der systemischen Behandlung und in der Strahlentherapie fokussieren aktuelle Diskussionen insbesondere auch auf die Verbesserung der chirurgischen Maßnahmen beim Mammakarzinom. Unter Berücksichtigung der zunehmenden Bedeutung der neoadjuvanten Chemotherapie erfährt die operative Behandlung des Mammakarzinoms sowohl im Bereich der Brust als auch im Bereich der Achselhöhle einen Wandel. Basierend auf der derzeitigen Datenlage wird die SLNE vor einer neoadjuvanten Chemotherapie grundsätzlich nicht empfohlen. Demgegenüber wird die operative axilläre Intervention – von der SLNE über die TAD bis zur ALND – nach der neoadjuvanten Chemotherapie als Vorgehen der Wahl zum axillären Staging angesehen. Zur Verringerung der Falsch-negativ-Rate des operativen Stagings der Axilla bei pN+CNB vor NACT und ycN0 nach NACT sind Targeted axillary Dissection (TAD), die Entfernung von > 2 SLNs (SLNE, kein ungezieltes axilläres Sampling), die Immunhistochemie zur Detektion von isolierten Tumorzellen oder Mikrometastasen und die Markierung von positiven Lymphknoten vor NACT als Standard anzusehen. In dem aktuellen Update zur operativen axillären Intervention wird auf die Bedeutung von isolierten Tumorzellen und Mikrometastasen nach neoadjuvanter Chemotherapie und die klinischen Konsequenzen einer mittels SLNE und TAD diagnostizierten Low Volume residual Disease eingegangen und ein Überblick bez. der diesjährigen AGO-Empfehlungen zum operativen Management der Axilla im Rahmen der Primäroperation und im Zusammenhang mit der neoadjuvanten Chemotherapie gegeben.
Article
Full-text available
For many decades, the standard procedure to treat breast cancer included complete dissection of the axillary lymph nodes. The aim was to determine histological node status, which was then used as the basis for adjuvant therapy, and to ensure locoregional tumour control. In addition to the debate on how to optimise the therapeutic strategies of systemic treatment and radiotherapy, the current discussion focuses on improving surgical procedures to treat breast cancer. As neoadjuvant chemotherapy is becoming increasingly important, the surgical procedures used to treat breast cancer, whether they are breast surgery or axillary dissection, are changing. Based on the currently available data, carrying out SLNE prior to neoadjuvant chemotherapy is not recommended. In contrast, surgical axillary management after neoadjuvant chemotherapy is considered the procedure of choice for axillary staging and can range from SLNE to TAD and ALND. To reduce the rate of false negatives during surgical staging of the axilla in pN+CNB stage before NACT and ycN0 after NACT, targeted axillary dissection (TAD), the removal of > 2 SLNs (SLNE, no untargeted axillary sampling), immunohistochemistry to detect isolated tumour cells and micro-metastases, and marking positive lymph nodes before NACT should be the standard approach. This most recent update on surgical axillary management describes the significance of isolated tumour cells and micro-metastasis after neoadjuvant chemotherapy and the clinical consequences of low volume residual disease diagnosed using SLNE and TAD and provides an overview of this yearʼs AGO recommendations for surgical management of the axilla during primary surgery and in relation to neoadjuvant chemotherapy.
Article
Background In the post-American College of Surgeons Oncology Group Z0011 trial era, radiologists have increasingly focused on excluding high-level or advanced axillary lymph node metastasis (ALNM) by using an additional MRI scan positioned higher than lower axillae; however, the value of these additional scans remains undetermined. Purpose To evaluate whether a standard MRI protocol is sufficient to exclude high-level or advanced ALNM in breast cancer or additional MRI of entire axilla is needed. Materials and Methods This retrospective study evaluated women with invasive breast cancer who underwent breast MRI from April 2015 to December 2016. Some underwent neoadjuvant chemotherapy (NAC) and others underwent upfront surgery. Standard (routine axial scans including the lower axillae) and combined (routine axial scans plus additional scans including the entire axilla) MRI protocols were compared for high-level or advanced ALNM detection. Clinical-pathologic characteristics were analyzed. Uni- and multivariable logistic regression was performed to identify predictors of high-level or advanced ALNM. Results A total of 435 women (mean age ± standard deviation, 52 years ± 11) were evaluated (65 in the NAC group, 370 in the non-NAC group). With the standard MRI protocol, predictors of high-level ALNM were peritumoral edema (odds ratio [OR], 12.3; 95% CI: 3.9, 39.4; P < .001) and positive axilla (OR, 5.9; 95% CI: 2.0, 15.2; P < .001). Only three of 289 women with negative axillae without peritumoral edema had high-level ALNM. Predictors of advanced ALNM were positive axillae (OR, 8.9; 95% CI: 3.7, 21.5; P < .001) and peritumoral edema (OR, 2.8; 95% CI: 1.1, 6.9; P = .03). Only six of 310 women who had negative axillae without peritumoral edema had advanced ALNM. Conclusion The performance of standard MRI was satisfactory in excluding high-level and advanced axillary lymph node metastasis in most patients with breast cancer. However, the presence of peritumoral edema or positive axillae in the MRI findings emphasizes the benefits of a combined MRI protocol. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Abe in this issue.
Article
LBA505 Background: NSABP B-32 is the largest prospective randomized phase III trial designed to determine in SN negative patients that SNR alone results in the same survival and regional control as SNR + AD while reducing morbidity. It was designed to detect a survival difference of 2% between the 2 groups at 5 years. Methods: 5,611 women with operable, clinically N0, invasive breast cancer were randomized to SNR + AD (Group 1) or to SNR alone with AD only if SNs were positive (Group 2). 3,989 (71.1%) of the 5,611 patients were SN negative and followed for events. 99.9% of these SN negative patients had follow-up information: 1,975 in Group 1 and 2,011 in Group 2. Median time on study was 95.3 months. Patients were well balanced across clinical strata. Log-rank tests for unadjusted analyses and Cox proportional hazard models adjusting for study stratification variables were used to compare overall survival (OS) and disease-free survival (DFS) between the two groups. Two-sided p-values were used. HR values > 1 indicate a more favorable outcome in Group 1 (SNR + AD). Results: Comparisons of OS (Group 1 vs. Group 2) yielded an unadjusted HR of 1.20 (p = 0.12) and an adjusted HR of 1.19 (p=0.13). Five-year Kaplan-Meier estimates for OS are 96.4% in Group 1 and 95.0% in Group 2 and the 8-year estimates are 91.8% and 90.3%, respectively. Comparisons of DFS (Group 1 vs. Group 2) yielded an unadjusted HR of 1.05 (p=0.54) and an adjusted HR of1.07 (p=0.57). No substantial differences could be seen across sites for first treatment failure. Five-year Kaplan-Meier estimates for DFS are 89.0% in Group 1, and 88.6% in Group 2 and the 8-year estimates are 82.4% and 81.5%, respectively. Local and Regional Recurrences: There were 54 local recurrences in Group 1 and 49 in Group 2 (p=0.55). There were 8 regional node recurrences as first events in Group 1 and 14 in Group 2 (p=0.22). Conclusions: No significant differences were observed in OS, DFS, or regional control between the trial groups. Within the limits of this trial, SNR without AD is validated as a safe and effective method for regional node treatment of SN negative breast cancer patients. [Table: see text]
Article
Background and objective: The early results of the American College of Surgeons Oncology Group (ACOSOG) Z0011 trial demonstrated no difference in locoregional recurrence for patients with positive sentinel lymph nodes (SLNs) randomized either to axillary lymph node dissection (ALND) or sentinel lymph node dissection (SLND) alone. We now report long-term locoregional recurrence results. Methods: ACOSOG Z0011 prospectively examined overall survival of patients with SLN metastases undergoing breast-conserving therapy randomized to undergo ALND after SLND or no further axillary specific treatment. Locoregional recurrence was prospectively evaluated and compared between the groups. Results: Four hundred forty-six patients were randomized to SLND alone and 445 to SLND and ALND. Both groups were similar with respect to age, Bloom-Richardson score, Estrogen Receptor status, adjuvant systemic therapy, histology, and tumor size. Patients randomized to ALND had a median of 17 axillary nodes removed compared with a median of only 2 SLNs removed with SLND alone (P < 0.001). ALND, as expected, also removed more positive lymph nodes (P < 0.001). At a median follow-up of 9.25 years, there was no statistically significant difference in local recurrence-free survival (P = 0.13). The cumulative incidence of nodal recurrences 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 (P = 0.36). Conclusion: Despite the potential for residual axillary disease after SLND, SLND without ALND offers excellent regional control for selected patients with early metastatic breast cancer treated with breast-conserving therapy and adjuvant systemic therapy.
Article
Background: Circulation of cancer cells in the blood is a necessary step of hematogeneous metastasis while circulating tumor cells (CTC) have been reported to have a low metastatic efficiency in preclinical animal models. After a median follow-up of 18 months, we previously reported that CTC detection influences the distant metastasis-free survival (DMFS) in non-metastatic breast cancer (BC) patients (pts) treated by neoadjuvant chemotherapy (NACT) in a multicenter prospective trial. Updated results are presented here, focusing on overall survival (OS) and predictors of metastatic relapse.Methods: In 115 localized BC pts, CTC were prospectively screened (CellSearch) before and after NACT (REMAGUS02). We analyzed their outcome after a median follow-up of 36 months.Results: At baseline, 23% of pts were CTC-positive, but only 10% had more than 1 CTC per 7.5ml of blood. At an individual level, CTC detection before chemotherapy, used as a test to predict metastatic relapse, exhibited a global accuracy of 77%, higher than that of tumor grade (54%), tumor size (57%), lymph node invasion (40%), triple negative phenotype (76%) and pathological complete response (27%). Multivariate analyses for OS and DMFS showed that CTC detection before chemotherapy was a strong independent prognostic factor for both DMFS (p=0.01, RR=5.0, 95%CI[1.4-17]) and OS (p=0.007, RR=9, 95%CI[1.8-45]), along with tumor size and triple-negative phenotype, while post-chemotherapy CTC detection had a lower significance for both endpoints (p=0.07 and p=0.09 respectively).Conclusion: Biologically, the metastatic efficiency of CTC could be higher than previously thought. Clinically, besides confirming our previously reported results, this study shows that CTC detection may become the main prognostic factor in BC pts treated with NACT. Implementing this technique in everyday management might help to identify high-risk pts in whom innovative strategies should be investigated.Supported by PHRC AOM/2OO2/02117, Pfizer inc., Roche, sanofi-aventis.ISRCTN10059974 Citation Information: Cancer Res 2009;69(24 Suppl):Abstract nr 3017.