Journal List > J Korean Med Sci > v.38(34) > 1516083831

Song, Cho, Cho, Jung, Park, Woo, and Seo: Value of Breast MRI and Nomogram After Negative Axillary Ultrasound for Predicting Axillary Lymph Node Metastasis in Patients With Clinically T1-2 N0 Breast Cancer

Abstract

Background

There are increasing concerns about that sentinel lymph node biopsy (SLNB) could be omitted in patients with clinically T1-2 N0 breast cancers who has negative axillary ultrasound (AUS). This study aims to assess the false negative result (FNR) of AUS, the rate of high nodal burden (HNB) in clinically T1-2 N0 breast cancer patients, and the diagnostic performance of breast magnetic resonance imaging (MRI) and nomogram.

Methods

We identified 948 consecutive patients with clinically T1-2 N0 cancers who had negative AUS, subsequent MRI, and breast conserving therapy between 2013 and 2020 from two tertiary medical centers. Patients from two centers were assigned to development and validation sets, respectively. Among 948 patients, 402 (mean age ± standard deviation, 57.61 ± 11.58) were within development cohort and 546 (54.43 ± 10.02) within validation cohort. Using logistic regression analyses, clinical-imaging factors associated with lymph node (LN) metastasis were analyzed in the development set from which nomogram was created. The performance of MRI and nomogram was assessed. HNB was defined as ≥ 3 positive LNs.

Results

The FNR of AUS was 20.1% (81 of 402) and 19.2% (105 of 546) and the rates of HNB were 1.2% (5/402) and 2.2% (12/546), respectively. Clinical and imaging features associated with LN metastasis were progesterone receptor positivity, outer tumor location on mammography, breast imaging reporting and data system category 5 assessment of cancer on ultrasound, and positive axilla on MRI. In validation cohorts, the positive predictive value (PPV) and negative predictive value (NPV) of MRI and clinical-imaging nomogram was 58.5% and 86.5%, and 56.0% and 82.0%, respectively.

Conclusion

The FNR of AUS was approximately 20% but the rate of HNB was low. The diagnostic performance of MRI was not satisfactory with low PPV but MRI had merit in reaffirming negative AUS with high NPV. Patients who had low probability scores from our clinical-imaging nomogram might be possible candidates for the omission of SLNB.

Graphical Abstract

jkms-38-e251-abf001.jpg

INTRODUCTION

There has been a steady trend toward axillary conservatism in patients with early-stage breast cancer. The surgical approach to the axilla has become less invasive, from complete axillary lymph node dissection (ALND) to sentinel lymph node biopsy (SLNB) to reduce potential morbidity.12 ALND has been the treatment of choice for patients with positive SLNB.3 However, clinical paradigms have been modified after the American College of Surgeons Oncology Group (ACOSOG) Z0011 trial,45 which found that SLNB without ALND did not compromise survival in patients with clinically T1-2 N0 breast cancers who underwent breast conserving therapy (BCT) if there were one or two nodal metastases at SLNB.45 The trial results raised question about the value of SLNB in clinically node-negative patients.6 Although SLNB had lower surgical morbidity compared to ALND, surgical complications still exist in patients with SLNB and omission of SLNB could leave out complications.24 Therefore, several randomized clinical trials such as SOUND (Sentinel Node versus Observation after Axillary Ultrasound),7 INSEMA (Intergroup-Sentinel-Mamma),8 and BOOG 2013-08 (Dutch randomized controlled multicenter trial) are currently underway to evaluate whether SLNB could be safely omitted in patients with T1-2 N0 breast cancers who have a negative axillary ultrasound (AUS) findings.68 As AUS was reported to have a high negative predictive value (NPV) of 84%, it has been suggested as a non-invasive alternative to SLNB.9
Given the evidence from the Z0011 trial, the role of breast magnetic resonance imaging (MRI) has also been challenged. Although breast MRI is controversial in early-stage breast cancer, MRI can depict potentially metastatic lymph nodes (LNs) in patients with negative AUS findings because it can provide objective and global views of the axilla irrespective of the patient’s body habitus and operator’s ability.10 Axillary MRI evaluation with a high NPV could result in omission of SLNB, whereas a high positive predictive value (PPV) could further guide ALND or neoadjuvant chemotherapy (NAC).11 Previous studies that retrospectively compared the diagnostic performance of MRI and ultrasound for depicting axillary LN metastasis showed that MRI had comparable performance to ultrasound.1213 However, there has been no published studies which evaluated the role of MRI in detecting additional LN metastases in patients with negative AUS.
For providing evidence for determining which patients can be included in the omission of SLNB, it is important to develop a reliable prediction model to predict axillary LN metastasis. Several nomograms have been developed for assessment of axillary burden.1415 However, these nomograms were mainly aimed at predicting non-sentinel LN metastasis in the presence of positive SLNB. Previous studies using ultrasound or MRI features of primary tumor reported that artificial intelligence might predict axillary LN metastasis.1617181920 However, advanced artificial intelligence is not widely applicable in clinical practice.
Therefore, this study aimed to evaluate the false negative result (FNR) and the rate of high nodal burden (HNB) in patients with clinically T1-2 N0 breast cancers who had negative AUS and assess the diagnostic performance of breast MRI and clinical-imaging nomogram for predicting axillary LN metastasis.

METHODS

Study designs and patients

By retrospectively reviewing two tertiary medical centers’ database (October 2013 and October 2020), we identified 948 clinically T1-2 N0 patients who had negative AUS, subsequent breast MRI, and BCT. At first, a primary development set of 402 patients from Korea University Anam Hospital were retrospectively analyzed. Inclusion criteria were having preoperative breast and AUS by experienced breast radiologists prior to breast MRI and negative axillary result on ultrasound. Exclusion criteria were positive axillary result on ultrasound, ultrasound performed by an outside hospital/resident, NAC before operation, distant metastasis at diagnosis, inadequate information about immunohistochemical staining, and outside MRI. A validation set of 546 patients from Korea University Guro Hospital was further analyzed. The diagram for establishing the development and validation sets is summarized in Fig.1.
Fig. 1

Flow diagram of the study population.

US = ultrasound, MRI = magnetic resonance imaging.
jkms-38-e251-g001

Breast and AUS examinations

To evaluate the clinical T and N stage, one of three breast radiologists (K.R.C., S.E.S., E.K.P., with 21,11, and 8 years of experience in breast imaging, respectively) in Korea University Anam Hospital and one of two breast radiologists (O.H.W. and H.S., with 19 and 7 years of experience) in Korea University Guro Hospital performed preoperative breast and AUS with a Logiq 9 unit (General Electric, Milwaukee, USA), and an Aixplorer system (SuperSonic Imagine, Aix en Provence, France) within a median of 14 and 12 days before surgery in the two sets. All cancers were assessed using the breast imaging reporting and data system (BI-RADS) categories 4A, 4B, 4C, and 5 (Fig. 2A). Axillary results on AUS were recorded as either negative or positive (Fig. 2B). Based on BI-RADS, positive axilla was considered when a LN with at least one of the following features was found: eccentric cortical thickening, diffuse cortical thickening of 3 mm or large, rounded hypoechoic, complete or partial effacement of the fatty hilum, and nonhilar cortical blood flow on color Doppler images.2123 If the assessment for axillary LN were equivocal or borderline, two breast radiologists (K.R.C., S.E.S. with 21 and 11 years of experience in breast imaging, respectively) re-evaluated the axillary LN status and categorized it into negative or positive by consensus. For obtaining clinical T stage, the longest tumor size on ultrasound was measured.
Fig. 2

Findings in a 53-year-old woman with clinically T1N0 breast cancer in her left breast. (A) Breast US shows a 1.3-cm mass with breast imaging reporting and data system category 5 assessment (arrows). (B) Axillary US shows a benign looking LN with an assement of negative axilla (arrows). (C) Axial T1-weighted postcontrast MRI covering entire axilla shows a suspicious metastatic LN (arrow) which was enlarged with cortical thickening, round shape, and a long axis to short axis ratio of less than two at level I of the left axilla. (D) Right craniocaudal view of mammography shows a mass without calcifications (arrow) in the outer breast. (E) Axial T1-weighted postcontrast MRI shows an irregular shaped and marginated, heterogeneously enhancing mass in her left breast.

US = ultrasound, LN = lymph node, MRI = magnetic resonance imaging.
jkms-38-e251-g002

Breast MRI examinations

Breast MRI examinations were performed using two 3.0T scanners (Achieva 3.0T TX; Philips Healthcare, Best, the Netherlands and MAGNETOM Skyra; Siemens Healthcare, Erlangen, Germany) in Korea University Anam Hospital and a 3.0T scanner (MAGNETOM Skyra; Siemens Healthcare) in Korea University Guro Hospital with a dedicated breast array coil in the axial orientation. All MRI scans were taken with the patients in a prone position. Detailed protocols were described in Supplementary Data 1.
One of three breast radiologists in Korea University Anam Hospital and one of two breast radiologists in Korea University Guro Hospital read breast MRI. Axillary MRI results were recorded as negative or positive (Fig. 2C). Positive axilla on MRI was considered when a LN with at least one of the following features was observed: markedly enlarged and morphologically grossly abnormal LNs, especially when they are distinctly different from other visible LNs, cortical irregularity or apparent spiculation, loss of fatty hilum, round shape, or a long axis to short axis ratio of less than two.2324 Axillary LN with equivocal or borderline assessment at radiologic report were re-evaluated in same way as breast ultrasound.

Image analysis

The mammographic and MRI features were retrospectively analyzed in a blinded manner without clinical information by two breast radiologists (K.R.C., S.E.S., with 21 and 11 years of experience in breast imaging, respectively) using the 2013 BI-RADS lexicon.25 Mammographic findings were evaluated with regard to the calcifications (presence vs. absence), and tumor location (outer vs. inner) (Fig. 2D). The shape, margin, density and echogenicity of tumors on mammography and ultrasound were not evaluated because we intended to use those findings from MRI. MRI features were evaluated in terms of mass shape (round to oval vs. irregular), mass margin (circumscribed or irregular vs. spiculated), mass internal enhancement (homogeneous or heterogeneous vs. rim), intratumoral high signal intensity and peritumoral edema on T2-weighted image (presence vs. absence) (Fig. 2E).

Data analysis

Clinical data (menopause, symptom, family history of breast cancer) were reviewed from the electronic medical records. Imaging data were obtained from radiology reports. Pathological data were acquired from histopathological reports of surgery. Pathologic data of primary breast cancer and LN metastasis were evaluated from surgical and pathologic reports. According to the eighth edition of the American Joint Committee on Cancer (AJCC) staging, pN1mi which means micrometastases (larger than 0.2 mm, but none larger than 2.0 mm) at LN was defined as pN1.21 At least one LN metastasis including pN1mi was considered as disease positive. In addition, HNB was defined as ≥ 3 positive LNs and low nodal burden was defined as 1–2 positive LNs according to the Z0011 trial.45 Estrogen receptor (ER) and progesterone receptor (PR) positivity were defined as the presence of positive staining in at least 1% of the nuclei in ten high-power fields. Human epidermal growth factor receptor 2 (HER2) negativity was defined as an immunohistochemical score of 1+ or 2+ staining with negative HER2 gene amplification on fluorescence in situ hybridization. Histologic type was categorized as ductal or other, and histologic grade was categorized as low (grade 1 or 2) or high (grade 3). Molecular subtypes were categorized as luminal (ER and/or PR-positive and HER2-negative), HER2-enriched (ER/PR-positive or negative, HER2-positive), or triple-negative (ER-negative, PR-negative, and HER2-negative).22 The Ki-67 proliferation index was dichotomized as low (< 14%) or high (≥ 14%).22

Statistical analysis

Baseline characteristics were compared using the Mann-Whitney U test, independent t-test, or independent χ2 test. The diagnostic performance of breast MRI and nomogram was assessed using sensitivity, specificity, PPV, and NPV. Logistic regression analysis was performed to identify clinical and imaging factors associated with axillary LN metastasis. For multivariable analysis, covariates with P values < 0.05 in the univariable analysis with forward elimination were used. Based on the adjusted odds ratio (OR) from multivariate analysis, clinical-imaging model was constructed and the nomogram was developed to visualize the results of the predictive model. Internal and external validation of nomogram was performed in development and validation sets. For validation, discrimination and calibration were assessed using area under the curve (AUC) and calibration slope. All statistical analyses were performed by using SPSS for Windows (version 20.0; IBM Corp., Armonk, NY, USA) and open-source R software (version 3.5.1; R Foundation for Statistical Computing, R Foundation, Vienna, Austria).

Ethics statement

The Institutional Review Board (IRB) of Korea University Anam Hospital approved this retrospective study (IRB No.: 2021AN0464). The IRB waived the need for informed consent.

RESULTS

Characteristics of the sets

A total of 948 consecutive female patients were included (mean age ± standard deviation, 55.8 ± 10.9; range, 28–87 years) with 402 female patients (57.61 ± 11.58) in the development set and 546 patients (54.43 ± 10.02) in the validation set. A comparison of characteristics between the two sets is listed in Table 1. Compared to the development set, the validation set was composed of younger patients, pathologic T2 stage, lymphovascular invasion, luminal subtype, higher Ki-67, BI-RADS 5 assessment category on US, and positive axilla on MRI (all P < 0.05).
Table 1

Patient and tumor characteristics in two medical centers

jkms-38-e251-i001
Variables Development set from Korea University Anam Hospital (n = 402) Validation set from Korea University Guro Hospital (n = 546) P value
Patient and tumor characteristics
Age 57.61 ± 11.58 54.43 ± 10.19 < 0.001
Histologic type 0.210
Ductal 349 (86.8) 458 (83.9)
Others 53 (13.2) 47 (16.1)
Histologic grade 0.912
I or II 291 (72.4) 397 (72.7)
III 111 (27.6) 149 (27.3)
Estrogen receptor 0.112
Positive 352 (87.6) 458 (83.9)
Negative 50 (12.4) 88 (16.1)
Progesterone receptor 0.088
Positive 331 (82.3) 425 (77.8)
Negative 71 (17.7) 121 (22.2)
HER2 0.097
Positive 56 (13.9) 98 (17.9)
Negative 346 (86.1) 448 (82.1)
Molecular subtype 0.001
Luminal 315 (78.4) 470 (86.1)
HER2 enriched 56 (13.9) 31 (5.7)
Triple negative 31 (7.7) 45 (8.2)
Ki-67 < 0.001
Low (< 14%) 238 (59.2) 193 (35.3)
High (≥ 14%) 164 (40.8) 353 (64.7)
Pathologic T stage 0.005
T1 307 (76.4) 372 (68.1)
T2 95 (23.6) 174 (31.9)
Pathologic LN status 0.723
No LN metastasis 321 (79.9) 441 (80.8)
Low nodal burden (1–2 positive LNs) 76 (18.9) 93 (17.0)
High nodal burden (≥ 3 positive LNs) 5 (1.2) 12 (2.2)
Lymphovascular invasion 0.005
Yes 42 (10.4) 92 (16.8)
No 360 (89.6) 454 (83.2)
Imaging features
BI-RADS category on US 0.002
C4 252 (62.7) 286 (52.4)
C5 150 (37.3) 260 (47.6)
T stage measured on US < 0.001
T1 337 (83.8) 403 (73.8)
T2 65 (16.2) 143 (26.2)
Positive axilla on MRI 0.046
No 367 (91.3) 476 (87.2)
Yes 35 (8.7) 70 (12.8)
Values are presented as mean ± standard deviation or number (%).
HER2 = human epidermal growth factor receptor 2, LN = lymph node, BI-RADS = breast imaging reporting and data system, US = ultrasound, MRI = magnetic resonance imaging.

FNRs of AUS

The FNRs of AUS were 20.1% (81 of 402 patients) within development set and 19.2% (105 of 546 patients) within validation set. The rates of HNB were 1.2% (5/402) and 2.2% (12/546), respectively. According to the clinical T stage, the rates of HNB were 0.6% (2/337) in T1 stage and 4.6% (3/65) in T2 stage within development set, and 2.5% (10/403) in the T1 stage and 1.4% (2/143) in the T2 stage within validation set.

Predictable factors for HNB

To investigate the factors that can predict HNB, multivariable analysis was performed (Supplementary Table 1). Interestingly, positive axilla on MRI showed marginal significance for predicting HNB (adjusted OR, 2.7; P = 0.059), following after lymphovascular invasion (adjusted OR 3.4; P = 0.023).

Diagnostic performance of breast MRI

FNR of breast MRI were 16.3% (60 of 367 patients) within development set and 13.4% (64 of 476 patients) within validation set. For predicting additional LN metastasis in patients with negative AUS, the NPVs of breast MRI in both development and validation sets were high (83.6% [307/367] and 86.5% [412/476]) while PPVs were low (60.0% [21/35] and 58.5% [41/70]). The sensitivities were low (25.9% [21/81] and 39.0% [41/105]), whereas the specificities were high (95.6% [307/321] and 93.4% [412/441]) (Table 2).
Table 2

Diagnostic performance of breast MRI in patients with negative axillary ultrasound finding

jkms-38-e251-i002
Variables Development set from Korea University Anam Hospital (n = 402) Validation set from Korea University Guro Hospital (n = 546)
Sensitivity 25.9% (21/81) 39.0% (41/105)
Specificity 95.6% (307/321) 93.4% (412/441)
Positive predictive value 60.0% (21/35) 58.5% (41/70)
Negative predictive value 83.6% (307/367) 86.5% (412/476)
False negative result 16.3% (60/367) 13.4% (64/476)

Development of nomogram with clinical-imaging factors

Among clinical factors, lymphovascular invasion, higher pathologic T stage, and PR positivity were factors associated with LN metastasis at univariable analysis (Table 3). Among imaging factors, outer tumor location on mammography, higher clinical T stage measured on US, BI-RADS 5 assessment category on ultrasound, and positive axilla on MRI were significant factors (Table 4). For development of clinical-imaging nomogram which could be used preoperatively, we selected significant clinical and imaging predictors which could be obtained before operation. Therefore, pathologic T stage and lymphovascular invasion which could be assessed after operation were not selected. At multivariable analysis, significant clinical-imaging predictors were PR positivity (adjusted OR, 2.8; P = 0.017), outer tumor location on mammography (adjusted OR, 1.8; P = 0.043), BI-RADS category 5 on US (adjusted OR, 2.6; P = 0.002), and positive axilla on MRI (adjusted OR, 7.8; P < 0.001). However, clinical T2 stage measured on ultrasound did not acquire the significance (adjusted OR, 1.5; P = 0.260) (Table 5). The nomogram integrating clinical and imaging factors was constructed (Fig. 3). The AUC of the clinical-imaging nomogram was 0.76 (95% confidence interval [CI], 0.70–0.81) in the development set and 0.77 (95% CI, 0.70–0.81) in the validation set (Fig. 4). The NPV of clinical-imaging nomogram in the validation set was 82.0%.
Table 3

Univariable analysis of axillary LN metastasis in relation to clinical characteristics in the development set

jkms-38-e251-i003
Clinical characteristics Total patients (n = 402) No axillary LN metastasis (n = 321) Axillary LN metastasis (n = 81) OR 95% CI P value
Age 57.61 ± 11.58 57.97 ± 11.81 56.22 ± 10.63 1.0 0.9–1.0 0.222
Menopause
Yes 264 (65.7) 213 (66.4) 51 (63.0) Reference - -
No 138 (34.3) 108 (33.6) 30 (37.0) 0.9 0.5–1.4 0.566
Symptom
Asymptomatic 228 (56.7) 184 (57.3) 44 (54.3) Reference - -
Palpable or nipple discharge 174 (43.3) 137 (42.7) 37 (45.7) 1.1 0.7–1.8 0.707
Family history of breast cancer
No 360 (89.6) 287 (89.4) 73 (90.1) Reference - -
Yes 42 (10.4) 34 (10.6) 8 (9.9) 0.9 0.4–2.1 0.819
Histologic type
Others 53 (13.2) 48 (15.0) 5 (6.2) Reference - -
Ductal 349 (86.8) 273 (85.0) 76 (93.8) 2.2 0.9–5.3 0.085
Histologic grade
I or II 291 (72.4) 235 (73.2) 56 (69.1) Reference - -
III 111 (27.6) 86 (26.8) 25 (30.9) 1.2 0.7–2.0 0.514
Estrogen receptor
Negative 50 (12.4) 44 (13.7) 6 (7.4) Reference - -
Positive 352 (87.6) 277 (86.3) 75 (92.6) 2.0 0.8–4.9 0.122
Progesterone receptor
Negative 71 (17.7) 63 (19.6) 8 (9.9) Reference - -
Positive 331 (82.3) 258 (80.4) 73 (90.1) 2.3 1.0–4.9 0.040
HER2
Negative 346 (86.1) 276 (86.0) 70 (86.4) Reference - -
Positive 56 (13.9) 45 (14.0) 11 (13.6) 0.9 0.5–1.9 0.880
Molecular subtype
Luminal 315 (78.4) 247 (76.9) 68 (84.0) Reference - -
HER2 enriched 56 (13.9) 45 (14.0) 11 (13.6) 0.8 0.4–1.8 0.705
Triple negative 31 (7.7) 29 (9.1) 2 (2.4) 0.3 0.1–1.1 0.059
Ki-67
Low (< 14%) 238 (59.2) 188 (58.6) 50 (61.7) Reference - -
High (≥ 14%) 164 (40.8) 133 (41.4) 31 (38.3) 0.9 0.6–1.5 0.715
Pathologic T stage
T1 307 (67.9) 252 (78.5) 55 (67.9) Reference - -
T2 95 (23.6) 69 (21.5) 26 (32.1) 1.7 1.1–3.0 0.046
Presence of DCIS
Yes 331 (82.3) 262 (81.6) 69 (85.2) 1.2 0.6–2.2 0.631
No 71 (17.7) 59 (18.4) 12 (14.8) Reference - -
Lymphovascular invasion
Yes 42 (10.4) 22 (6.9) 20 (24.7) 4.4 2.3–8.5 < 0.001
No 360 (89.6) 298 (93.1) 61 (75.3) Reference - -
Values are presented as mean ± standard deviation or number (%).
LN = lymph node, CI = confidence interval, HER2 = human epidermal growth factor receptor 2, DCIS = ductal carcinoma in situ.
Table 4

Univariable analysis of pathologic LN metastasis in relation to imaging features in the development set

jkms-38-e251-i004
Imaging features Total patients (n = 402) No axillary LN metastasis (n = 321) Axillary LN metastasis (n = 81) Odds ratio 95% CI P value
Mammographic features
Mass or asymmetry
No 49 (12.2) 38 (11.8) 11 (13.6) Reference
Yes 353 (87.8) 283 (88.2) 70 (86.4) 0.8 0.4–1.8 0.669
Calcifications
No 314 (78.1) 254 (79.1) 60 (74.1) Reference - -
Yes 88 (21.9) 67 (20.9) 21 (25.9) 1.3 0.7–2.3 0.362
Tumor location
Inner 173 (43.0) 147 (45.8) 26 (32.1) Reference - -
Outer 229 (57.0) 174 (54.2) 55 (67.9) 1.8 1.0–2.9 0.027
Parenchymal density
Fatty 209 (52.0) 165 (51.4) 44 (54.3) Reference - -
Dense 193 (48.0) 156 (48.6) 37 (45.7) 0.9 0.5–1.4 0.864
Sonographic features
BI-RADS category on ultrasound
Category 4 252 (62.7) 217 (67.6) 35 (43.2) Reference - -
Category 5 150 (37.3) 104 (32.4) 46 (56.8) 2.7 1.6–4.3 < 0.001
T stage measured on US
T1 337 (83.8) 277 (86.3) 60 (74.1) Reference
T2 65 (16.2) 44 (13.7) 21 (25.9) 2.2 1.2–3.9 0.009
MRI features
Background parenchymal enhancement
Minimal to mild 294 (73.1) 237 (73.8) 57 (70.4) Reference - -
Moderate to marked 108 (26.9) 108 (26.2) 24 (29.6) 1.2 0.7–2.0 0.582
Lesion type
Massa 396 (98.5) 315 (98.1) 81 (100.0) Reference - -
Non-mass enhancement 6 (1.5) 6 (1.9) 0 (0) 42187539.9 0, … 0.999
Mass shapea
Round or oval 342 (85.1) 276 (86.0) 66 (81.5) Reference - -
Irregular 54 (13.4) 39 (12.1) 15 (18.5) 1.6 0.8–3.0 0.170
Mass margina
Circumscribed or irregular 290 (72.1) 236 (73.5) 54 (66.7) Reference - -
Spiculated 106 (26.4) 79 (24.6) 27 (33.3) 1.5 0.9–2.5 0.159
Mass internal enhancementa
Homogeneous or heterogeneous 311 (77.4) 253 (78.8) 58 (71.6) Reference - -
Rim 85 (21.1) 62 (19.3) 23 (28.4) 1.6 0.9–2.8 0.105
Intratumoral high SI on T2WI
Low or intermediate 216 (53.7) 178 (55.5) 38 (46.9) Reference - -
High 186 (46.3) 143 (44.5) 43 (53.1) 1.4 0.8–2.2 0.210
Peritumoral edema on T2WI
No 335 (83.3) 270 (84.1) 65 (80.2) Reference - -
Yes 67 (16.7) 51 (15.9) 16 (19.8) 1.3 0.7–2.4 0.439
Associated findings on MRI
No 383 (95.3) 305 (95.0) 78 (96.3) Reference - -
Yes 19 (4.7) 16 (5.0) 3 (3.7) 0.7 0.2–2.5 0.611
Positive axilla on MRI
No 367 (91.3) 307 (95.6) 60 (74.1) Reference - -
Yes 35 (8.7) 14 (4.4) 21 (25.9) 7.6 3.7–15.9 < 0.001
Values are presented as number (%).
LN = lymph node, CI = confidence interval, BI-RADS = breast imaging reporting and data system, US = ultrasound, MRI = magnetic resonance imaging.
aMass shape, margin, and internal enhancement were calculated using a denominator of 396 masses.
Table 5

Diagnostic performances of clinical-imaging nomogram in validation set

jkms-38-e251-i005
Model Multivariate analysis Diagnostic performance in the validation set
Adjusted odds ratio 95% CI P value AUC P valueb Sensitivity Specificity Positive predictive value Negative predictive value
Clinical-imaging nomogram 0.76 (0.70–0.81)a < 0.001b 20.0% 96.0% 56.0% 82.0%
Progesterone receptor positivity 2.8 1.2–6.7 0.017
Outer tumor location on MG 1.8 1.0–3.1 0.043
T2 stage measured on US 1.5 0.7–2.9 0.260
BI-RADS category 5 on US 2.3 1.4–4.6 0.003
Positive axilla on MRI 7.8 3.5–17.3 < 0.001
CI = confidence interval, AUC = area under the curve, MG = mammography, US = ultrasound, BI-RADS = breast imaging reporting and data system, MRI = magnetic resonance imaging.
aNumbers in parentheses are 95% CIs.
bP value was acquired from comparison with the reference standard using the Delong method.
Fig. 3

Clinical-imaging nomogram. Each point that corresponds to each feature is on the uppermost point scale, and the sum of all points is the total points. The total points projected at the bottom scale indicate the probability of LN metastasis.

MG = mammography, BI-RADS = breast imaging reporting and data system, US = ultrasound, MRI = magnetic resonance imaging, LN = lymph node.
jkms-38-e251-g003
Fig. 4

Receiver operating characteristic curves (AUCs) of the clinical-imaging nomogram to predict axillary lymph node metastasis in development and validation sets. Numbers in parentheses are AUCs.

AUC = area under the curve, CI = confidence interval.
jkms-38-e251-g004

DISCUSSION

Although imaging strategies for the axilla vary among institutions,20 many centers routinely include AUS as part of the clinical examination because the false negative rate of physical examination has been reported as high as 45%.26 AUS was recently added as a routine workup for axilla in patients with invasive breast cancer according to the National Comprehensive Cancer Network (NCCN) guidelines.22 Moreover, AJCC manual defined clinically suspicious nodes as having palpability on physical examination or suspicious imaging features.21 Considering clinically ongoing trials such as SOUND, INSEMA, and BOOG 2013-08, the role of negative AUS in triaging omission of SLNB has become more important. In our study, the FNR of AUS was 19.2% and 20.1%, which were slightly lower than the 23.4–25% of previous studies.2728 These studies reported that the rate of HNB was 4.2% and 6.7%, which were higher than our rates of 1.2% and 2.2%, respectively.2728 Accordingly, we can conclude that negative AUS can miss LN metastasis in approximately 20% of cases. However, the rate of HNB, according to the Z0011 trial, was low.
Breast MRI is usually recommended for patients with advanced cancers who will receive NAC.2930 Controversy remains regarding the use of MRI in early-stage breast cancer. For omission of SLNB in T1-2 N0 breast cancers, the role of MRI needs to be re-evaluated because it can detect additional LN metastasis missed by AUS.10 In this study, the diagnostic performance of MRI was not satisfactory with low PPVs. However, MRI had merit in reaffirming negative axilla by AUS with high NPVs. Furthermore, predictors for HNB were only lymphovascular invasion and positive axilla on MRI, concordant with a recent study that demonstrated that positive axilla on MRI could predict pathologic N2-3 stage.31 Therefore, surgeons should be careful when omitting SLNB in the presence of positive axilla on MRI.
In our study, three imaging features such as outer tumor location on mammography, BI-RADS 5 assessment category of tumor on ultrasound, and positive axilla on MRI were valuable factors for predicting axillary LN metastasis. Previous study also insisted that outer tumor location was the significant factor affecting LN metastasis in clinically T1-2 N0 breast cancer patients.32 In addition, BI-RADS 5 assessment category on US is known to be associated with larger tumor size, axillary LN metastasis, and lymphovascular invasion.33 Therefore, outer tumor location on mammography and BI-RADS 5 assessment category of tumor on ultrasound, as well as positive axilla on MRI should be considered with significance for predicting LN metastasis.
Among clinicopathological factors, lymphovascular invasion was the most powerful predictor, followed by PR positivity and pathologic T stage. While lymphovascular invasion is a well-known predictor for axillary LN metastasis,141517 the reason why PR positivity affects LN metastasis is less known. It can be assumed that progestins support metastasis of tumor cells,33 and the short PR isoform is a major driver in promoting metastasis of luminal breast cancer by suppressing estrogen/ER action.34 As lymphovascular invasion and pathologic T stage were not factors that could be acquired before surgery, we made a nomogram using significant clinical and imaging predictors which could be obtained preoperatively. Although clinical-imaging nomogram is not perfect considering NPV of 82.0%, our nomogram might be helpful to find possible candidates for the omission of SLNB in patients with clinically T1-2 N0 breast who had negative AUS.
Regarding T stage, the rate of HNB according to the T stage is an important issue in ongoing trials. The inclusion criteria for the SOUND trial were T1 stage, while those for INSEMA and BOOG 2013-08 trials were the T1-2 stages. A recent study advocated the continued use of SLNB in T2 stages because negative AUS has a rate of 6.2% of HNB in T2 stages, higher than 2% in T1 stages.27 In our study, the rates of HNB were 0.3% and 1.9% in the T1 stage and 4.2% and 2.9% in the T2 stage in development and validation sets. Anticipatory to our expectations, clinical T stage measured on ultrasound did not acquire the significance at multivariable analysis. These findings would help provide evidence for determining whether the T2 stage is included in the omission of SLNB in clinical trials.
Our study has several limitations. First, we excluded patients with negative US-guided fine-needle aspiration biopsy (FNAB) results because their AUS findings were positive. FNAB was routinely performed in hospital A, but not hospital B. So, we decided to exclude those patients despite NCCN guidelines and ongoing clinical trials6782135 considered patients with negative FNAB results as negative axilla. Second, this was a retrospective study with small sample size. Third, there were significant differences in the clinical features between the two sets. However, this inherent limitation is unavoidable, and we believe that these differences can reflect the real clinical situation among different institutions. Fourth, the assessment of axillary LN and BI-RADS category depends on the radiologists’ expertise and can be subjective. Because of the nature of retrospective study design, consensus on the criteria was not exactly established and criteria may differ according to the radiologists and institutions. This can limit the application of the clinical-imaging nomogram using imaging features assessed by other radiologists who are likely to have different results interpreting axillary LN status and BI-RADS category.
In conclusion, negative AUS by experienced radiologists missed LN metastasis in approximately 20% of case but the rates of HNB were low in clinically T1-2 N0 breast cancer patients. Breast MRI had merit in reaffirming negative AUS and positive axilla on MRI can help predict HNB. Although clinical-imaging nomogram is not perfect considering NPV of 82.0%, patients who had low probability scores from our nomogram might be candidates for the omission of SLNB. In line with the upcoming results of ongoing trials, our investigation could contribute to future national/international guidelines about axillary LN management.

Notes

Funding: The authors state that this research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (grant number: NRF-2021R1F1A1046016) and a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI) funded by the Ministry of Health & Welfare, Republic of Korea (grant number: HR22C1302).

Disclosure: The authors have no potential conflicts of interest to disclose.

Data Availability Statement: The datasets generated and/or analyzed during the current study are available from the corresponding author on reasonable request.

Author Contributions:

  • Conceptualization: Song SE.

  • Data curation: Cho Y, Cho KR.

  • Formal analysis: Cho Y.

  • Funding acquisition: Song SE.

  • Investigation: Song SE, Woo OH, Seo BK.

  • Methodology: Song SE, Cho Y.

  • Resources: Jung SP.

  • Supervision: Park KH, Cho KR.

  • Validation: Woo OH.

  • Writing - original draft: Song SE.

  • Writing - review & editing: Park KH, Seo BK, Cho KR.

References

1. Caudle AS, Cupp JA, Kuerer HM. Management of axillary disease. Surg Oncol Clin N Am. 2014; 23(3):473–486. PMID: 24882346.
crossref
2. Burak WE Jr, Hollenbeck ST, Zervos EE, Hock KL, Kemp LC, Young DC. Sentinel lymph node biopsy results in less postoperative morbidity compared with axillary lymph node dissection for breast cancer. Am J Surg. 2002; 183(1):23–27. PMID: 11869698.
crossref
3. Lyman GH, Giuliano AE, Somerfield MR, Benson AB 3rd, Bodurka DC, Burstein HJ, et al. American society of clinical oncology guideline recommendations for sentinel lymph node biopsy in early-stage breast cancer. J Clin Oncol. 2005; 23(30):7703–7720. PMID: 16157938.
crossref
4. 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; 305(6):569–575. PMID: 21304082.
crossref
5. Giuliano AE, Ballman KV, McCall L, Beitsch PD, Brennan MB, Kelemen PR, et al. Effect of axillary dissection vs no axillary dissection on 10-year overall survival among women with invasive breast cancer and sentinel node metastasis: the ACOSOG Z0011 (Alliance) randomized clinical trial. JAMA. 2017; 318(10):918–926. PMID: 28898379.
crossref
6. van Roozendaal LM, Vane MLG, van Dalen T, van der Hage JA, Strobbe LJA, Boersma LJ, et al. Clinically node negative breast cancer patients undergoing breast conserving therapy, sentinel lymph node procedure versus follow-up: a Dutch randomized controlled multicentre trial (BOOG 2013-08). BMC Cancer. 2017; 17(1):459. PMID: 28668073.
crossref
7. Gentilini O, Botteri E, Dadda P, Sangalli C, Boccardo C, Peradze N, et al. Physical function of the upper limb after breast cancer surgery. Results from the SOUND (Sentinel node vs. Observation after axillary Ultra-souND) trial. Eur J Surg Oncol. 2016; 42(5):685–689. PMID: 26899941.
crossref
8. Reimer T, Stachs A, Nekljudova V, Loibl S, Hartmann S, Wolter K, et al. Restricted axillary staging in clinically and sonographically node-negative early invasive breast cancer (c/it1-2) in the context of breast conserving therapy: first results following commencement of the Intergroup-Sentinel-Mamma (INSEMA) trial. Geburtshilfe Frauenheilkd. 2017; 77(2):149–157. PMID: 28331237.
crossref
9. 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; 264(6):1098–1102. PMID: 26779976.
crossref
10. Chang JM, Leung JW, Moy L, Ha SM, Moon WK. Axillary nodal evaluation in breast cancer: state of the art. Radiology. 2020; 295(3):500–515. PMID: 32315268.
crossref
11. Kuijs VJ, Moossdorff M, Schipper RJ, Beets-Tan RG, Heuts EM, Keymeulen KB, et al. The role of MRI in axillary lymph node imaging in breast cancer patients: a systematic review. Insights Imaging. 2015; 6(2):203–215. PMID: 25800994.
crossref
12. van Nijnatten TJ, Ploumen EH, Schipper RJ, Goorts B, Andriessen EH, Vanwetswinkel S, et al. Routine use of standard breast MRI compared to axillary ultrasound for differentiating between no, limited and advanced axillary nodal disease in newly diagnosed breast cancer patients. Eur J Radiol. 2016; 85(12):2288–2294. PMID: 27842679.
crossref
13. Abe H, Schacht D, Kulkarni K, Shimauchi A, Yamaguchi K, Sennett CA, et al. Accuracy of axillary lymph node staging in breast cancer patients: an observer-performance study comparison of MRI and ultrasound. Acad Radiol. 2013; 20(11):1399–1404. PMID: 24119352.
14. Barranger E, Coutant C, Flahault A, Delpech Y, Darai E, Uzan S. An axilla scoring system to predict non-sentinel lymph node status in breast cancer patients with sentinel lymph node involvement. Breast Cancer Res Treat. 2005; 91(2):113–119. PMID: 15868438.
crossref
15. Degnim AC, Reynolds C, Pantvaidya G, Zakaria S, Hoskin T, Barnes S, et al. Nonsentinel node metastasis in breast cancer patients: assessment of an existing and a new predictive nomogram. Am J Surg. 2005; 190(4):543–550. PMID: 16164917.
crossref
16. Bae MS, Shin SU, Song SE, Ryu HS, Han W, Moon WK. Association between US features of primary tumor and axillary lymph node metastasis in patients with clinical T1-T2N0 breast cancer. Acta Radiol. 2018; 59(4):402–408. PMID: 28748712.
crossref
17. Yi A, Moon WK, Cho N, Chang JM, Bae MS, Kim SJ, et al. Association of tumour stiffness on sonoelastography with axillary nodal status in T1 breast carcinoma patients. Eur Radiol. 2013; 23(11):2979–2987. PMID: 23783782.
crossref
18. Zhou LQ, Wu XL, Huang SY, Wu GG, Ye HR, Wei Q, et al. Lymph node metastasis prediction from primary breast cancer US images using deep learning. Radiology. 2020; 294(1):19–28. PMID: 31746687.
crossref
19. Ha R, Chang P, Karcich J, Mutasa S, Fardanesh R, Wynn RT, et al. Axillary lymph node evaluation utilizing convolutional neural networks using MRI dataset. J Digit Imaging. 2018; 31(6):851–856. PMID: 29696472.
crossref
20. Han L, Zhu Y, Liu Z, Yu T, He C, Jiang W, et al. Radiomic nomogram for prediction of axillary lymph node metastasis in breast cancer. Eur Radiol. 2019; 29(7):3820–3829. PMID: 30701328.
crossref
21. Hortobagyi GN, Connolly JL, D'Orsi CJ, Edge SB, Mittendorf EA, Rugo HS. Breast. American joint committee on cancer. Amin MB, Edge SB, editors. AJCC cancer staging manual. 8th ed. Chicago IL: Springer;2017. p. 589–628.
22. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology: breast cancer. Accessed November 1, 2019. https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf .
23. Ecanow JS, Abe H, Newstead GM, Ecanow DB, Jeske JM. Axillary staging of breast cancer: what the radiologist should know. Radiographics. 2013; 33(6):1589–1612. PMID: 24108553.
crossref
24. Baltzer PA, Dietzel M, Burmeister HP, Zoubi R, Gajda M, Camara O, et al. Application of MR mammography beyond local staging: is there a potential to accurately assess axillary lymph nodes? evaluation of an extended protocol in an initial prospective study. AJR Am J Roentgenol. 2011; 196(5):W641-7. PMID: 21512057.
crossref
25. American college of radiology. ACR BI-RADS Atlas: breast imaging reporting and data system. 5th ed. Reston, VA: American college of radiology;2013.
26. Sacre RA. Clinical evaluation of axillar lymph nodes compared to surgical and pathological findings. Eur J Surg Oncol. 1986; 12(2):169–173. PMID: 3709822.
27. Keelan S, Heeney A, Downey E, Hegarty A, Roche T, Power C, et al. Breast cancer patients with a negative axillary ultrasound may have clinically significant nodal metastasis. Breast Cancer Res Treat. 2021; 187(2):303–310. PMID: 33837870.
crossref
28. Neal CH, Daly CP, Nees AV, Helvie MA. Can preoperative axillary US help exclude N2 and N3 metastatic breast cancer? Radiology. 2010; 257(2):335–341. PMID: 20807849.
crossref
29. Boughey JC, Alvarado MD, Lancaster RB, Fraser Symmans W, Mukhtar R, Wong JM, et al. Surgical standards for management of the axilla in breast cancer clinical trials with pathological complete response endpoint. NPJ Breast Cancer. 2018; 4(1):26. PMID: 30131975.
crossref
30. Pilewskie M, Morrow M. Axillary nodal management following neoadjuvant chemotherapy: a review. JAMA Oncol. 2017; 3(4):549–555. PMID: 27918753.
crossref
31. Byon JH, Park YV, Yoon JH, Moon HJ, Kim EK, Kim MJ, et al. Added value of MRI for Invasive breast cancer including the Entire axilla for evaluation of high-level or advanced axillary lymph node metastasis in the post-ACOSOG Z0011 trial era. Radiology. 2021; 300(1):46–54. PMID: 33904772.
crossref
32. Zhang Y, Li J, Fan Y, Li X, Qiu J, Zhu M, et al. Risk factors for axillary lymph node metastases in clinical stage T1-2N0M0 breast cancer patients. Medicine (Baltimore). 2019; 98(40):e17481. PMID: 31577783.
crossref
33. Liang Y, Benakanakere I, Besch-Williford C, Hyder RS, Ellersieck MR, Hyder SM. Synthetic progestins induce growth and metastasis of BT-474 human breast cancer xenografts in nude mice. Menopause. 2010; 17(5):1040–1047. PMID: 20461021.
crossref
34. Rosati R, Oppat K, Huang Y, Kim S, Ratnam M. Clinical association of progesterone receptor isoform a with breast cancer metastasis consistent with its unique mechanistic role in preclinical models. BMC Cancer. 2020; 20(1):512. PMID: 32493230.
crossref
35. Chang JM, Shin HJ, Choi JS, Shin SU, Choi BH, Kim MJ, et al. Imaging protocol and criteria for evaluation of axillary lymph nodes in the NAUTILUS trial. J Breast Cancer. 2021; 24(6):554–560. PMID: 34877830.
crossref

SUPPLEMENTARY MATERIALS

Supplementary Data 1

Supplementary Methods
jkms-38-e251-s001.doc

Supplementary Table 1

Patient characteristics and imaging features for predicting high nodal burden in patients with pathologic LN metastasis
jkms-38-e251-s002.doc
TOOLS
Similar articles