Abstracting and Indexing

  • PubMed NLM
  • Google Scholar
  • CrossRef
  • WorldCat
  • ResearchGate
  • Academic Keys
  • DRJI
  • Microsoft Academic
  • Academia.edu
  • OpenAIRE

Neoadjuvant Chemotherapy for Breast Cancer: Evolution of Clinical Practice in a French Cancer Center Over 16 Years and Pathologic Response Rates According to Tumor Subtypes and Clinical Tumor Size: Retrospective Cohort Study

Article Information

Gilles Houvenaeghel1*, Alexandre de Nonneville2, Monique Cohen1, Frédéric Viret2, Sandrine Rua1, Laura Sabiani1, Max Buttarelli1, Emmanuelle Charaffe3, Audrey Monneur2, Aurélie Jalaguier-Coudray4, Marie Bannier1, Renaud Sabatier2, Anthony Gonçalves2

1Aix-Marseille University, CNRS (National Center of Scientific Research), INSERM (National Institute of Health and Medical Research), Paoli-Calmettes Institute, Department of Surgical Oncology, CRCM (Research Cancer Centre of Marseille), 13009 Marseille, France

2Aix-Marseille University, CNRS (National Center of Scientific Research), INSERM (National Institute of Health and Medical Research), Paoli-Calmettes Institute, Department of Medical Oncology, CRCM (Research Cancer Centre of Marseille), 13009 Marseille, France

3Aix-Marseille University, CNRS (National Center of Scientific Research), INSERM (National Institute of Health and Medical Research), Paoli-Calmettes Institute, Department of Pathology, CRCM (Research Cancer Centre of Marseille), 13009 Marseille, France

4Aix-Marseille University, CNRS (National Center of Scientific Research), INSERM (National Institute of Health and Medical Research), Paoli-Calmettes Institute, Department of Radiology, CRCM (Research Cancer Centre of Marseille), 13009 Marseille, France

*Corresponding Author: Gilles Houvenaeghel, Aix-Marseille University, CNRS (National Center of Scientific Research), INSERM (Na-tional Institute of Health and Medical Research), Paoli-Calmettes Institute, Department of Surgical Oncology, CRCM (Research Cancer Centre of Marseille), 13009 Marseille, France

Received: 07 July 2022; Accepted: 14 July 2022; Published: 14 September 2022

Citation:

Gilles Houvenaeghel, Alexandre de Nonneville, Monique Cohen, Frédéric Viret, Sandrine Rua, Laura Sabiani, Max Buttarelli, Emmanuelle Charaffe, Audrey Monneur, Aurélie Jalaguier-Coudray, Marie Bannier, Renaud Sabatier and Anthony Gonçalves. A Critical Review on Reconstructive Options for Vulvo-Vaginal Defects. Journal of Surgery and Research 5 (2022): 511-525.

View / Download Pdf Share at Facebook

Abstract

We examined characteristics trends in early breast cancer patients receiving neoadjuvant chemotherapy (NAC) over a 16-year period. Our primary objective was to analyze variations in tumor stage and subtype over time. Secondary objectives included analyses of type of surgery and pathological response, from January 2005 to May 2021, 1623 patients receiving NAC were identified. Three periods were determined: 2005-2009 (P1), 2010-2014 (P2), 2015-2021 (P3). Correlations between periods and patient features with cT stage, pathological breast and axillary node response, pathological complete response (pCR), and type of surgery were assessed in univariate and multivariate analyses. We observed a significant increase in cT0-1 and N0 stages with periods (from 6.8% at P1 to 21.2% at P3, and from 43.2% at P1 to 55.9% at P3, respectively) and in the proportion of HER2+ and triple negative (TN) subtypes. In a multivariate analysis, a decrease of cT2-3-4 tumors during P3 was observed for HER2+ (OR:0.174; p=0.004) and TN tumors (OR:0.287; p=0.042). In-breast pCR and pCR were observed in 40.8% and 34.4% of all patients, respectively, with strong association with tumor subtypes, but not with tumor size in multivariate analysis (37.0% pCR for cT0-1 tumors, 36.4% for cT2 tumors, 29.1% for cT3 tumors (cT0-1 versus cT≥2; p=0.222)). pCR was negatively associated with cN1 stage (OR:1.499; p<0.001 for cN1 patients compared to cN0). We observed an increase in the proportion of small cT0- 1 and N0 stages treated with NAC, especially in HER2+ and TN subtypes. No significant impact of tumor size on pCR rates was found.

Keywords

Breast cancer; Neoadjuvant chemotherapy; Clinical practice; Pathologic response

Breast cancer articles; Neoadjuvant chemotherapy articles; Clinical practice articles; Pathologic response articles

Breast cancer articles Breast cancer Research articles Breast cancer review articles Breast cancer PubMed articles Breast cancer PubMed Central articles Breast cancer 2023 articles Breast cancer 2024 articles Breast cancer Scopus articles Breast cancer impact factor journals Breast cancer Scopus journals Breast cancer PubMed journals Breast cancer medical journals Breast cancer free journals Breast cancer best journals Breast cancer top journals Breast cancer free medical journals Breast cancer famous journals Breast cancer Google Scholar indexed journals Neoadjuvant chemotherapy articles Neoadjuvant chemotherapy Research articles Neoadjuvant chemotherapy review articles Neoadjuvant chemotherapy PubMed articles Neoadjuvant chemotherapy PubMed Central articles Neoadjuvant chemotherapy 2023 articles Neoadjuvant chemotherapy 2024 articles Neoadjuvant chemotherapy Scopus articles Neoadjuvant chemotherapy impact factor journals Neoadjuvant chemotherapy Scopus journals Neoadjuvant chemotherapy PubMed journals Neoadjuvant chemotherapy medical journals Neoadjuvant chemotherapy free journals Neoadjuvant chemotherapy best journals Neoadjuvant chemotherapy top journals Neoadjuvant chemotherapy free medical journals Neoadjuvant chemotherapy famous journals Neoadjuvant chemotherapy Google Scholar indexed journals Clinical practice articles Clinical practice Research articles Clinical practice review articles Clinical practice PubMed articles Clinical practice PubMed Central articles Clinical practice 2023 articles Clinical practice 2024 articles Clinical practice Scopus articles Clinical practice impact factor journals Clinical practice Scopus journals Clinical practice PubMed journals Clinical practice medical journals Clinical practice free journals Clinical practice best journals Clinical practice top journals Clinical practice free medical journals Clinical practice famous journals Clinical practice Google Scholar indexed journals Pathologic response articles Pathologic response Research articles Pathologic response review articles Pathologic response PubMed articles Pathologic response PubMed Central articles Pathologic response 2023 articles Pathologic response 2024 articles Pathologic response Scopus articles Pathologic response impact factor journals Pathologic response Scopus journals Pathologic response PubMed journals Pathologic response medical journals Pathologic response free journals Pathologic response best journals Pathologic response top journals Pathologic response free medical journals Pathologic response famous journals Pathologic response Google Scholar indexed journals triple negative articles triple negative Research articles triple negative review articles triple negative PubMed articles triple negative PubMed Central articles triple negative 2023 articles triple negative 2024 articles triple negative Scopus articles triple negative impact factor journals triple negative Scopus journals triple negative PubMed journals triple negative medical journals triple negative free journals triple negative best journals triple negative top journals triple negative free medical journals triple negative famous journals triple negative Google Scholar indexed journals sentinel lymph node biopsy articles sentinel lymph node biopsy Research articles sentinel lymph node biopsy review articles sentinel lymph node biopsy PubMed articles sentinel lymph node biopsy PubMed Central articles sentinel lymph node biopsy 2023 articles sentinel lymph node biopsy 2024 articles sentinel lymph node biopsy Scopus articles sentinel lymph node biopsy impact factor journals sentinel lymph node biopsy Scopus journals sentinel lymph node biopsy PubMed journals sentinel lymph node biopsy medical journals sentinel lymph node biopsy free journals sentinel lymph node biopsy best journals sentinel lymph node biopsy top journals sentinel lymph node biopsy free medical journals sentinel lymph node biopsy famous journals sentinel lymph node biopsy Google Scholar indexed journals axillary lymph node dissection articles axillary lymph node dissection Research articles axillary lymph node dissection review articles axillary lymph node dissection PubMed articles axillary lymph node dissection PubMed Central articles axillary lymph node dissection 2023 articles axillary lymph node dissection 2024 articles axillary lymph node dissection Scopus articles axillary lymph node dissection impact factor journals axillary lymph node dissection Scopus journals axillary lymph node dissection PubMed journals axillary lymph node dissection medical journals axillary lymph node dissection free journals axillary lymph node dissection best journals axillary lymph node dissection top journals axillary lymph node dissection free medical journals axillary lymph node dissection famous journals axillary lymph node dissection Google Scholar indexed journals Endocrine Receptor articles Endocrine Receptor Research articles Endocrine Receptor review articles Endocrine Receptor PubMed articles Endocrine Receptor PubMed Central articles Endocrine Receptor 2023 articles Endocrine Receptor 2024 articles Endocrine Receptor Scopus articles Endocrine Receptor impact factor journals Endocrine Receptor Scopus journals Endocrine Receptor PubMed journals Endocrine Receptor medical journals Endocrine Receptor free journals Endocrine Receptor best journals Endocrine Receptor top journals Endocrine Receptor free medical journals Endocrine Receptor famous journals Endocrine Receptor Google Scholar indexed journals

Article Details

Abbreviations:

NAC : neoadjuvant chemotherapy

pCR : pathological complete response

TN : triple negative

BC : breast cancer

SLNB : sentinel lymph node biopsy

ALND : axillary lymph node dissection

ER : Endocrine Receptor

1. Introduction

Neoadjuvant chemotherapy (NAC), as defined as the administration of cytotoxic treatment before tumor surgical removal, has been implemented in breast cancer (BC) during the last 50 years, encompassing different objectives. Initially dedicated to inoperable BC to convert it into a surgically removable disease [1,2], NAC was then widely developed in operable BC when frontline breast-conserving surgery with “in sano” margins was not feasible [3], based on ac-cumulating data showing a high rate of clinical and pathologic response (pCR) as well as identical survival outcome when compared to adjuvant chemotherapy [4]. Yet, the development of more and more sophisticated oncoplastic techniques could also increase the possibilities of immediate breast-conserving surgery [5,6]. Even though some initial expectations in favor of NAC were not confirmed, including a potential survival advantage over adjuvant chemotherapy by an early impact on the hypothetic surgical stimulation of micro-metastatic disease and tumor shedding [7], the interest in NAC has been further boosted during the last decade in relation with several observations. First, molecular analysis of BC revealed that the disease was composed of different subtypes with distinct survival outcomes [8,9], including HER2-positive and triple-negative BC, two subtypes in which both survival benefits from adjuvant chemotherapy and probability of reaching pCR after NAC are the highest. Second, a tight relationship was demonstrated between the achievement of pCR and survival, most notably in the latter subtypes [10-12]. Third, recent randomized trials found that in those patients with residual invasive disease after preoperative systemic treatment, adjuvant trastuzumab emtansine in HER2-positive BC treated with trastuzumab-based NAC [13] and capecitabine in triple negative (TN) BC treated with anthracycline-taxane NAC [14] both significantly improved survival. These recent practice-changing results have resulted in making NAC a standard of care in most of these BC subtypes, even when initial breast conservation may be achieved by frontline surgery. In this retrospective monocentric study, we have examined how the use of NAC has evolved during the last 16 years in a French comprehensive cancer center, including clinical and pathological features of treated patients, as well as pathologic complete response (pCR) results. The primary objective was to analyze variations in tumor stage and subtype over time. Secondary objectives included analyses of the type of breast and axillary surgery as well as pathological axillary and in-breast response.

2. Materials and Methods

2.1. Patient selection and study design

Medical records of early BC patients treated from January 2005 to May 2021 were retrieved from our institutional clinical databases for retrospective analysis. This cohort study was approved by our institutional review board (Registered study in Clinical research Institute: NAC-TS-IPC 2021-026). Patient and tumor characteristics, periods, treatments, and patho-logical results were collected. We included in the present study 1623 patients treated with NAC, without metastasis at initial diagnosis (figure 1). Patients receiving NAC were staged using clinical examination, mammography and ul-trasonogaphy, breast MRI. Search for distant metastases using either PET-scan or a combination of CT-scan and bone scan. Evaluation of lymph node status was determined by sentinel lymph node biopsy (SLNB) with or without completion axillary lymph node dissection (ALND) or only ALND. SLNB was realized before NAC for cN0 patients with cT ≤ 5 centimeters. The method used for the de-tection of SN was a combined technique or isotopic only detection during the last years. NAC included anthracyclines plus taxanes-based regimen. All patients with HER2+ dis-ease received trastuzumab during NAC. Patients who had residual components of ductal carcinoma in situ were assessed as having a breast pathological complete response based on the National Surgical Adjuvant Breast and Bowel Project criteria [15]. Patients with axillary residual lymph node tumor (ypN1) were assessed as having no pathological complete response. Pathological complete response (pCR) was defined as [ypT0 or ypTis] and [ypN0 or pN0sn or pN1sn without ALND or ypNx (absence of axillary surgery)] [16]. Endocrine Receptor (ER) and HER2 status were determined according to French guide-lines (estrogen and/or progesterone receptors by IHC with a 10% threshold for endocrine re-ceptor (ER) positivity; IHC HER2-positivity score of 3+ and/or HER2 amplification by in situ hybridization) [17,18]. Five IHC tumor subtypes were defined as surrogates for molecular subtypes based on tumor grade, ER and HER2 status: luminal A-like (ER+/HER2-/grade1 or 2), luminal B-like (ER+/HER2-/grade 3), luminal B-like HER2+, HER2+ (ER-/HER2+), and triple-negative (ER-/HER2-) [16]. The work has been reported in line with the STROCSS criteria [17].

Abbrevations: NAC: neoadjuvant chemotherapy; MO, without synchronous distant metastases; cT, clinical tumour size stage; ypT, pathologic tumour status after neoadjuvant chemotherapy

fortune-biomass-feedstock

Figure 1: Flow chart.

2.2. Statistical Analysis

Descriptive statistics were used to describe the categorical (counts and frequencies) and continuous (median and range) variables. Characteristics of patients were compared by different periods: Period 1 (P1) 2005-2009, period 2 (P2) 2010-2014, and period 3 (P3) 2015-2021 by using χ2 test for categorical variables, and Kruskal-Wallis test for continuous variables. The main characteristics of patients and tumors were categorized (cT0-1 stage vs. cT2-3-4, ypT0-is vs. ypT≥1, pCR vs. no pCR, breast conservative surgery vs. mastectomy) and association with other variables were explored in univariate, and multivariate analysis by binary logistic regression for significant criteria in univariate analysis. Statistical significance was set as p ≤ 0.05. Analyses were performed with SPSS version 16.0 (SPSS Inc., Chicago, Illinois).

3. Results

3.1. Characteristics of patients according to periods

Of 1629 patients included, 280 received NAC during P1, 503 during P2, and 846 during P3. Median age for all patients was 50.0 years and was stable among periods. Characteristics of patients are reported in table 1. All the other assessed criteria were significantly associated with periods of treatment. Notably, we observed a significant increase in the proportion of cT0-1 (6.8% at P1 to 21.2% at P3), and node-negative tumors (43.2% at P1 to 55.9% at P3) with periods. The rate of breast conservative surgery increased (36.8% at P1 to 48.5% at P3), whereas ALND decreased significantly with time (74.3% at P1 to 57.7% at P3). Similarly, a decrease in the proportion of Luminal-A (36.1% at P1, 27.4% at P2, and 18.2% at P3), and Luminal-B HER2- (19.3%, 14.9%, and 8.2%) tumors was observed, while HER2+ (15.0%, 16.3%, and 21.0%), Luminal B HER2+ (10.7%, 12.5%, and 15.0%), and triple-negatives (18.6%, 28.6%, and 37.5%) increased. Of note, the rate of residual invasive disease in breast and nodes also significantly decreased across the three periods (Table 1).

 

P1

 

P2

 

P3

 

Chi-2

Total

 

Nb

%

Nb

%

Nb

%

p-value

Nb

%

Age

median

48

 

50

 

50

   

50

 
 

CI 95%

48.0-50.6

49.7-51.9

50.8-52.6

 

50.4-51.6

cT stage

cT0-1

19

6.8

42

8.3

179

21.2

<0.0001

240

14.7

 

cT2

154

55

308

61.2

473

55.9

 

935

57.4

 

cT3

82

29.3

132

26.2

136

16.1

 

350

21.5

 

cT4

25

8.9

21

4.2

58

6.9

 

104

6.4

cN stage

cN0

121

43.2

196

39

473

55.9

<0.0001

790

48.5

 

cN1

143

51.1

302

60

372

44

 

817

50.2

 

cNx

16

5.7

5

1

1

0.1

 

22

1.4

Breast

Conservative

103

36.8

220

43.7

410

48.5

0.003

733

45

surgery

Mastectomy

177

63.2

283

56.3

436

51.5

 

896

55

Axillary

SLNB

5

1.8

82

16.3

265

31.3

<0.0001

352

21.6

surgery

ALND

208

74.3

305

60.6

488

57.7

 

1001

61.4

 

SLNB+ALND

65

23.2

113

22.5

81

9.6

 

259

15.9

 

No

2

0.7

3

0.6

12

1.4

 

17

1

Subtypes

Luminal A

101

36.1

138

27.4

154

18.2

<0.0001

393

24.1

 

Luminal B Her2-

54

19.3

75

14.9

69

8.2

 

198

12.2

 

Luminal Her2- Grade?

1

0.4

1

0.2

1

0.1

 

3

0.2

 

Luminal B Her2+

42

15

82

16.3

178

21

 

302

18.5

 

Her2+

30

10.7

63

12.5

127

15

 

220

13.5

 

Triple Negative

52

18.6

144

28.6

317

37.5

 

513

31.5

ypT

ypT0

95

33.9

145

28.8

280

33.1

0.043

520

31.9

 

ypTis

17

6.1

41

8.2

89

10.5

 

147

9

 

ypT >= 1

168

60

317

63

477

56.4

 

962

59.1

pN

pN0sn

3

1.1

75

15

215

25.7

<0.0001

293

18.1

 

pN1sn

2

0.7

8

1.6

53

6.3

 

63

3.9

 

ypN0

167

60.1

200

40

302

36.1

 

669

41.4

 

ypN1

106

38.1

217

43.4

267

31.9

 

590

36.5

Abbreviations: cT, clinical tumor size stage; cN, clinical lymph node status; SLNB, sentinel lymph node biopsy; ALND, axillary lymph node dissection; ypT, pathologic tumor status after neoadjuvant chemotherapy; pN pathologic lymph node status

Table 1: Characteristics of patients according to three periods of treatment Period 1 (P1) 2005-2009, period 2 (P2) 2010-2014, and period 3 (P3) 2015-2021.

3.2. Axillary surgery details for all patients and for clinically N0 patients

For all patients, axillary surgery type (SLNB, ALND, or SLNB+ALND) was significantly associated with periods, tumor subtypes, cT, and cN stages (Table 2). Considering only cN0 patients, SLNB was performed in 496 cases with completion ALND in 168 patients (33.9%). In cN0 patients, SLNB was significantly associated with the period of treatment (42.1% (51/121) during P1, 71.9% (141/196) during P2, and 64.2% (304/473) during P3 ; p<0.0001), tumor subtypes (49.2% (89/181) in Luminal-A tumors, 63.6% (49/77) in Luminal-B HER2-, 65.7% (94/143) in Luminal B HER2+, 56.0% (60/107) in HER2+, and 72.3% (204/282) in TN ; p<0.0001), and cT stages (63.3% (95/150) for cT0-1 tumors, 71.7% (362/505) for cT2, and 31.7% (38/120) for cT3 ; p<0.0001). In multivariate analysis, SLNB was less frequently per-formed for cT3 and cT4 tumors compared to cT0-1, but more commonly used for TN tumors (OR: 2.332, CI95% 1.511-3.597 ; p<0.0001), and during P2 and P3 periods (OR: 3.567, CI95% 2.120-6.001; p<0.0001 and OR: 1.877, CI95% 1.181-2.983 ; p=0.008, respectively) (Table3, SLNB versus ALND section).

 

SLNB

 

ALND

 

SLNB + ALND

No Surgery

 

Chi-2

Nb

%

Nb

%

Nb

%

Nb

%

p-value

All patients

 

352

 

1001

 

259

 

17

   

Period

P1

5

1.4

208

20.8

65

25.1

2

11.8

<0.0001

 

P2

82

23.3

305

30.5

113

43.6

3

17.6

 
 

P3

265

75.3

488

48.8

81

31.3

12

70.6

 

Subtypes

Luminal A

44

12.5

273

27.3

73

28.2

3

17.6

<0.0001

 

Luminal B Her2-

23

6.5

134

13.4

41

15.8

0

0

 
 

Luminal B Her2+

64

18.2

186

18.6

49

18.9

3

17.6

 
 

Her2+

45

12.8

147

14.7

25

9.7

3

17.6

 
 

Triple Negative

176

50

259

25.9

70

27

8

47.1

 
 

Luminal Her2- Grade?

0

0

2

0.2

1

0.4

0

0

 

cN stage

cN0

328

93.2

282

28.2

168

64.9

12

70.6

<0.0001

 

cN1

23

6.5

702

70.1

88

30.4

4

23.5

 
 

cNx

1

0.3

17

1.7

3

1.2

1

5.9

 

cT stage

cT0-1

82

23.3

129

12.9

24

9.3

5

29.4

<0.0001

 

cT2

254

72.2

481

48.1

190

73.9

8

47.1

 
 

cT3

16

4.5

291

29.1

40

15.6

3

17.6

 
 

cT4

0

0

100

10

3

1.2

1

5.9

 

only cN0 patients

328

 

282

 

168

 

12

   

Period

P1

5

1.5

69

24.5

46

27.4

1

8.3

<0.0001

 

P2

76

23.2

54

19.1

65

38.7

1

8.3

 
 

P3

247

75.3

159

56.4

57

33.9

10

83.3

 

Subtypes

Luminal A

43

13.1

90

31.9

46

27.4

2

16.7

<0.0001

 

Luminal B Her2-

23

7

28

9.9

26

15.5

0

0

 
 

Luminal B Her2+

61

18.6

46

16.3

33

19.6

3

25

 
 

Her2+

44

13.4

46

16.3

16

9.5

1

8.3

 
 

Triple Negative

157

47.9

72

25.5

47

28

6

50

 

cT stage

cT0-1

77

23.5

50

17.7

18

10.7

5

41.7

<0.0001

 

cT2

237

72.3

137

48.6

125

74.4

6

50

 
 

cT3

14

4.3

81

28.7

24

14.3

1

8.3

 
 

cT4

0

0

14

5

1

0.6

0

0

 
                       

Abbreviations: cT, clinical tumor size stage; cN, clinical lymph node status; SLNB, sentinel lymph node biopsy; ALND, axillary lymph node dissection

Table 2: Axillary surgery for all patients and for clinically N0 patients.

     

CI 95%

 

p-value

OR

Inferior

Superior

SLNB versus ALND

       

Luminal A

 

1

   

Luminal B Her2-

0.206

1.462

0.811

2.634

Luminal B Her2+

0.053

1.631

0.994

2.677

Her2+

0.355

1.286

0.754

2.193

Triple Negative

<0.0001

2.332

1.511

3.597

cT0-1

 

1

   

cT2

0.086

1.431

0.95

2.154

cT3

<0.0001

0.254

0.147

0.44

cT4

0.003

0.041

0.005

0.329

P1

 

1

   

P2

<0.0001

3.567

2.12

6.001

P3

0.008

1.877

1.181

2.983

cT2-3-4 versus cT0-1

       

P1

 

1

   

P2

0.371

0.771

0.435

1.365

P3

<0.0001

0.27

0.162

0.45

Luminal A

 

1

   

Lum B Her2-

0.981

1.006

0.606

1.671

Lum B Her2+

0.385

1.211

0.786

1.868

Her2+

0.062

1.617

0.976

2.681

Triple Neg

0.163

1.315

0.895

1.933

cN0

 

1

   

cN>=1

<0.0001

1.748

1.306

2.34

mastectomy versus BCS

     

P1

 

1

   

P2

0.224

0.818

0.591

1.131

P3

0.114

0.778

0.57

1.062

cT0-1

 

1

   

cT2

0.381

0.877

0.653

1.177

cT3

<0.0001

2.974

2.058

4.298

cT4

<0.0001

6.469

3.324

12.589

Luminal A

 

1

   

Lum B Her2-

0.085

0.725

0.503

1.045

Lum B Her2+

0.662

0.93

0.673

1.287

Her2+

0.259

1.234

0.856

1.779

Triple Neg

<0.0001

0.567

0.426

0.756

cN0

 

1

   

cN1

<0.0001

1.518

1.223

1.883

cNx

0.684

1.223

0.464

3.222

Abbreviations: cT, clinical tumor size stage; cN, clinical lymph node status; SLNB, sentinel lymph node biopsy; ALND, axillary lymph node dissection; BCS, breast conservative surgery.

Table 3: Results of regression analysis to determine associations between periods, tumor subtypes, and clinical tumor size for SLNB versus ALND, and clinical lymph node status for cT2-3-4 versus cT0-1, and clinical tumor size and clinical lymph node status for mastectomy versus breast conservative surgery.

3.3. cT Stages according to periods

cT stage (cT0-1 versus cT2-3-4) was significantly associated with periods, and cN stages, while the proportion of different subtypes did not differ significantly by univariate analysis (Table 4). The multivariate analysis revealed a strong association of cT2-3-4 tumors with cN≥1 (OR: 1.748; p<0.0001), and a notable decrease of cT2-3-4 stages during the third period (OR: 0.270 ; p<0.0001) (Table 3, cT2-3-4 versus cT0-1 section). During the third period, cT0-1 rates increased significantly during years 2018-2021 (122/515: 23.7%), in comparison with years 2015-2017: (57/331: 17.2%); p=0.015) for all tumor subtypes, and for HER2-positive/ER-negative (21.4%: 18/84 versus 14.0%: 6/43), Luminal B HER2-positive (25.4% (31/122) versus 17.9% (10/56)), triple-negative tumors (22.5% (43/191) versus 15.1% (19/126)) [23.2% (92/397) versus 15.6% (35/225); p=0.014, considering together these three last subtypes]. There was no significant difference observed in Luminal A and Luminal B HER2-negative tumors: 25.4% (30/118) during years 2018-2021 versus 20.0% (21/105) during years 2015-2017 (p=0.211). Tumor subtypes in depth analysis showed higher rates of cT0-1 tumors during the third period for all tumor subtypes, except for Luminal B HER2-negative tumors (Table S1). In a multivariate analysis adjusted on periods and cN status, a decrease of cT2-3-4 tumors during the third period was observed for Luminal A tumors (OR: 0.197, CI 95% 0.079-0.489; p<0.0001) with an increase of cN1 patients (OR: 1.927, CI 95% 1.072-3.465; p=0.028), for HER2+ (Luminal B HER2+ & HER2+) (OR: 0.174, CI 95% 0.053-0.573; p=0.004) without significant difference for cN status (OR: 1.454, CI 95% 0.865-2.445; p=0.158), for triple-negative tumors (OR:0.287, CI 95% 0.086-0.956; p=0.042) with an increase of cN1 patients (OR: 1.814, CI 95% 1.073-3.066; p=0.026) and without significant difference for Luminal B HER2- tumors (OR: 0.616, CI 95% 0.215-1.768; p=0.368, and OR: 2.067, CI 95% 0.904-4.724; p=0.085, for P3 and cN1 status, respectively).

   

cT0-1

 

cT2-3-4

 

Chi-2

   

Nb

%

Nb

%

p-value

All patients

 

240

14.7

1389

85.3

 

Period

P1

19

7.9

261

18.8

<0.0001

 

P2

42

17.5

461

33.2

 
 

P3

179

74.6

667

48

 

Subtypes

Luminal A

58

24.2

335

24.1

0.154

 

Luminal B Her2-

27

11.2

171

12.3

 
 

Luminal Her2+ G?

2

0.8

1

0.1

 
 

Luminal B Her2+

47

19.6

255

18.4

 
 

Her2+

27

11.2

193

13.9

 
 

Triple Negative

79

32.9

434

31.2

 

cN stage

cN0

150

62.5

640

46.1

<0.0001

 

cN1

90

37.5

727

52.3

 
 

cNx

0

0

22

1.6

 

Luminal A

 

58

14.8

335

85.2

 

Period

P1

6

10.3

95

28.4

<0.0001

 

P2

14

24.1

124

37

 
 

P3

38

65.5

116

34.6

 

cN stage

cN0

35

60.3

146

43.6

0.041

 

cN1

23

39.7

181

54

 
 

cNx

0

0

8

2.4

 

Luminal B Her2-

27

13.6

171

86.4

 

Period

P1

6

22.2

48

28.1

0.295

 

P2

8

29.6

67

39.2

 
 

P3

13

48.1

56

32.7

 

cN stage

cN0

15

55.6

62

36.3

0.135

 

cN1

12

44.4

105

61.4

 
 

cNx

0

0

4

2.3

 

Luminal B Her2+ & Her2+

74

14.2

448

85.8

 

Period

P1

3

4.1

69

15.4

<0.0001

 

P2

6

8.1

139

31

 
 

P3

65

87.8

240

53.6

 

cN stage

cN0

45

60.8

205

45.8

0.051

 

cN1

29

39.2

241

53.8

 
 

cNx

0

0

2

0.4

 

Triple Negative

79

15.4

434

84.6

 

Period

P1

3

3.8

49

11.3

0.003

 

P2

14

17.7

130

30

 
 

P3

62

78.5

255

58.8

 

cN stage

cN0

55

69.6

227

52.3

0.012

 

cN1

24

30.4

199

45.9

 
 

cNx

0

0

8

1.8

 

Abbreviations: cN, clinical lymph node status.

Table 4: Characteristics of patients according to clinical tumor size, cT0-1 versus cT2-3-4.

3.4. Pathological results: in-breast tumor response

An in-breast pathological complete response ypT0-is was reported in 40.8% of all patients, including 9.0% of in-situ residual tumors (ypTis). A trend toward a lower rate of invasive residual tumor was observed in the third period (480/836: 49.7% for P3 versus 485/783: 61.9% for P1-2). For all patients, a significant association between in breast pathological response (ypT0-is versus ypT≥1) and tumor subtypes was observed in univariate analysis (Table 5): 14.2% for Luminal A (56/393), 28.8% for Luminal B HER2-negative (57/198), 49.0% for Luminal B HER2-positive (148/302), 67.7% for HER2-positive/ER-negative (149/220), and 49.1% for triple-negative tumors (252/513) (Figure 2). In patients with Luminal-A tumors, ypT0-is response rates were 2.8% (2/71) and 16.8% (54/322) for grade 1 and grade 2 tumors, respectively. For Luminal-A grade 2 tumors, ypT0-is response rates were 8.9% (11/124), 15.4% (6/39), and 23.3% (37/159) for tumors with Ki67≤20%, >20%, and unknown, respectively (p=0.005). In multivariate analysis, a strong association was observed between ypT0-is and tumor subtypes, but not with tumor size (Table 6) which was distributed as follows: 45.4% (109/240) of ypT0-is for cT0-1, 41.7% (390/935) for cT2 and 37.1% (130/350) for cT3 tumors (cT0-1 versus cT>1: p=0.077).

   

ypT0-is

 

ypT≥1

 

Chi-2

   

Nb

%

Nb

%

p-value

All patients

 

664

40.8

965

59.2

 

Period

P1

112

16.9

168

17.4

0.073

 

P2

186

28

317

32.8

 
 

P3

366

55.1

480

49.7

 

Subtypes

Luminal A

56

8.4

337

34.9

<0.0001

 

Luminal B Her2-

57

8.6

141

14.6

 
 

Luminal B Her2+

148

22.3

154

16

 
 

Her2+

149

22.4

71

7.4

 
 

Triple Negative

252

38

261

27

 
 

Luminal Her2- Grade?

2

0.3

1

0.1

 

cT stage

cT0-1

109

16.4

131

13.6

0.087

 

cT2

390

58.7

545

56.5

 
 

cT3

130

19.6

220

22.8

 
 

cT4

35

5.3

69

7.2

 

cN stage

cN0

322

48.5

468

48.5

1

 

cN1

333

50.2

484

50.2

 
 

cNx

9

1.4

13

1.3

 
 

Luminal A Grade 1

2

3.6

69

20.5

0.001

 

Luminal A Grade 2

54

96.4

268

79.5

 
   

pCR

 

no pCR

 

Chi-2

   

Nb

%

Nb

%

p-value

All patients

 

561

34.4

1068

65.6

 

Period

P1

94

16.8

186

17.4

0.011

 

P2

149

26.6

354

33.1

 
 

P3

318

56.7

528

49.4

 

Subtypes

Luminal A

30

5.3

363

34

<0.0001

 

Luminal B Her2-

41

7.3

157

14.7

 
 

Luminal B Her2+

128

22.8

174

16.3

 
 

Her2+

138

24.6

82

7.7

 
 

Triple Negative

223

39.8

290

27.2

 
 

Luminal Her2- Grade?

1

0.2

2

0.2

 

cT stage

cT0-1

89

15.9

151

14.1

0.045

 

cT2

340

60.6

595

55.7

 
 

cT3

102

18.2

248

23.2

 
 

cT4

30

5.3

74

6.9

 

cN stage

cN0

311

55.4

479

44.9

<0.0001

 

cN1

243

43.3

574

53.7

 
 

cNx

7

1.2

15

1.4

 
 

Luminal A Grade 1

2

6.7

69

19

0.065

 

Luminal A Grade 2

28

93.3

294

81

 

Abbreviations: ypT, pathologic tumor status after NAC; cT, clinical tumor size stage; cN, clinical lymph node status.

Table 5: Factors associated with residual breast tumor and pathologic complete response (pCR) in univariate analysis.

residual tumor versus ypT0-is

p-value

OR

CI 95%

   

Inferior

Superior

cT stage

cT2-3-4

 

1

   
 

cT0-1

0.079

0.766

0.569

1.031

Subtypes

Luminal A

 

1

   
 

Lum B Her2-

<0,0001

0.409

0.269

0.622

 

Lum B Her2+

<0,0001

0.173

0.12

0.248

 

Her2+

<0,0001

0.078

0.052

0.117

 

Triple Neg

<0,0001

0.172

0.123

0.239

no pCR versus pCR

p-value

OR

CI 95%

   

Inferior

Superior

cT stage

cT2-3-4

 

1

   
 

cT0-1

0.447

0.885

0.647

1.212

Subtypes

Luminal A

 

1

   
 

Lum B Her2-

<0,0001

0.305

0.183

0.507

 

Lum B Her2+

<0,0001

0.11

0.071

0.171

 

Her2+

<0,0001

0.048

0.03

0.076

 

Triple Neg

<0,0001

0.109

0.072

0.165

cN stage

cN0

 

1

   
 

cN1

<0,0001

1.499

1.194

1.882

 

cNx

0.845

0.905

0.332

2.468

Abbreviations: ypT, pathologic tumor status after NAC; cT, clinical tumor size stage; cN, clinical lymph node status.

Table 6: Regression analysis to determine factors significantly associated with residual breast tumor and pathologic complete response (pCR).

3.5. Pathological results: axillary lymph node response

Considering cN0 patients, sentinel nodes were involved (pN1sn) in 16.7% (55/330) of cases, and 70.2% (315/449) were ypN0, whereas cN1 patients were ypN0 in 43.8% (344/786) (Table 7). Residual nodal tumor (ypN1) was present in 20.8% (103/496) of ypT0-is tumors and in 63.8% (487/763) of ypT≥1 tumors (p<0.0001), in 6.6% (11/167) and 43.5% (123/283) of ypT0-is and ypT>1 tumors, respectively for cN0 patients, 28.0% (90/322) and 75.8% (354/467) of ypT0-is and ypT>1 tumors, respectively for cN1 patients (20 patients cN unknown) (p<0.0001) (Table 7); in 50.0% (26/52), 30.8% (16/52), 17.2% (20/116), 9.6% (11/115), and 18.2% (29/159) of ypT0-is tumors for Luminal A, Luminal B HER2-negative, Luminal B HER2-positive, HER2-positive/ER-negative and triple negative tumors, respectively (p<0.0001) (Table S2) (Figure 2).

 

Luminal A

Lum B Her2-

Lum B Her2+

Her2+

Triple Neg

Luminal Her2- Grade?

Total

 

Nb

%

Nb

%

Nb

%

Nb

%

Nb

%

Nb

%

Nb

%

cN0

                           

pN0sn

29

16.1

20

26

53

37.9

35

33

138

50

0

0

275

35.3

pN1sn

15

8.3

4

5.2

9

6.4

8

7.5

19

6.9

0

0

55

7.1

ypN0

71

39.4

33

42.9

68

48.6

52

49.1

91

33

0

0

315

40.4

ypN1

65

36.1

20

26

10

7.1

11

10.4

28

10.1

0

0

134

17.2

cN1

                           

pN0sn

0

0

0

0

3

1.9

1

0.9

12

5.5

0

0

16

2

pN1sn

1

0.5

1

0.9

0

0

0

0

5

2.3

0

0

7

0.9

ypN0

37

18.3

41

35

87

55.1

74

66.7

104

47.7

1

33.3

344

42.5

ypN1

164

81.2

75

64.1

68

43

36

32.4

97

44.5

2

66.7

442

54.6

Table 7: Pathologic axillary nodal status. Abbreviations: cN, clinical lymph node status; ypN, pathologic lymph node status after NAC

Abbrevations: ypT, pathologic tumor status after neoadjuvant chemotherapy; ypN pathologic lymph node status afer neoadjuvant chemotherapy; pCR, pathologic complete response.

fortune-biomass-feedstock

Figure 2: pathological breast tumor response, axillary lymph node response, and pathologic complete responses rates after neoadjuvant chemotherapy.

3.6. Pathological results: pathological complete response

Pathological complete response rate was reported in 34.4% of all patients, without corre-lation with clinical tumor size: 37.0% (89/240) pCR for cT0-1 tumors, 36.4% (340/935) for cT2 tumors and 29.1% (102/350) for cT3 tumors (cT0-1 versus cT≥1 ; p=0.222). In univariate analysis, pCR was significantly correlated with tumor subtypes (7.6%, 20.7%, 42.4%, 62.7% and 43.5% for Luminal A, Luminal B HER2-, Luminal B HER2+, HER2+ and triple-negative tumors, respectively; p<0.0001) (Figure 2), cN stage (39.4% and 29.7% for cN0 and cN1, respectively; p<0.0001) and periods (33.6%, 29.6%, 37.6% for P1, P2 and P3, respectively; p=0.011) (Table 5). In multivariate analysis, a strong association was observed between pCR and tumor subtypes, and between pCR and cN1 stage, with more patients with residual breast or nodal tumor for cN1 patients (OR: 1.499, p<0.0001) (Table 6).

3.7. Breast conservative surgery or mastectomy

Breast surgery type was significantly associated with periods, tumor subtypes, cT stages, and cN stages: 36.8% (103/280) of breast conservative surgery during P1, 43.7% (220/503) during P2 and 48.5% (410/846) during P3; 38.4% (151/393) of breast conservative surgery for Luminal-A tumors, 45.4% (90/198) for Luminal-B HER2-, 42.7% (129/302) for Luminal-B HER2+, 34.5% (76/220) for HER2+, and 55.5% (285/513) for triple-negative; 52.9% (127/240) for cT0-1 tumors, 54.4% (509/935) for cT2, and 24.3% (85/350) for cT3 (Table 8). In multi-variate analysis, lower rates of mastectomies were observed for triple-negative tumors (OR: 0.567; p<0.0001), whereas mastectomies rates were higher for cT3, cT4 tumors, and cN1 stages (Table 3). There was no significant difference according to periods of treatment.

   

Conservative

Mastectomy

Chi-2

   

Nb

%

Nb

%

p-value

All patients

 

733

45

896

55

 

Period

P1

103

14.1

177

19.8

0.002

 

P2

220

30

283

31.6

 
 

P3

410

55.9

436

48.7

 

Subtypes

Luminal A

151

20.6

242

27

<0.0001

 

Luminal B Her2-

90

12.3

108

12.1

 
 

Luminal B Her2+

129

17.6

173

19.3

 
 

Her2+

76

10.4

144

16.1

 
 

Triple Negative

285

38.9

228

25.4

 
 

Luminal Her2- Grade?

2

0.3

1

0.1

 

cT stage

cT0-1

127

17.3

113

12.6

<0.0001

 

cT2

509

69.4

426

47.5

 
 

cT3

85

11.6

265

29.6

 
 

cT4

12

1.6

92

10.3

 

cN stage

cN0

423

57.7

367

41

<0.0001

 

cN1

302

41.2

515

57.4

 
 

cNx

8

1.1

14

1.6

 

ypT

ypT0

246

33.6

274

30.6

0.274

 

ypTis

70

9.5

77

8.6

 
 

ypT >= 1

417

56.9

545

60.8

 

pCR

Yes

273

37.2

288

32.1

0.018

 

No

460

62.8

608

67.9

 

Abbreviations: cT, clinical tumor size stage; cN, clinical lymph node status; ypT, pathologic tumor status after neoadjuvant chemotherapy; pCR, pathologic complete response.

Table 8: Significant associations between breast conservative surgery or mastectomy in univariate analysis.

4. Discussion

In this large single-center retrospective study covering a broad 16-year period, we reported an increase in NAC use in successive years for small (cT0-1) tumors, and an increase in the use of SLNB before NAC with no significant difference in pCR and mastectomy rates. In locally advanced and large breast cancers, particularly when mastectomy is required, NAC is recommended to decrease mastectomy rate [19]. In operable patients, NAC has not demonstrated impact on survival in comparison with adjuvant chemotherapy after breast surgery [7,19]. However, NAC allows assessment of pathological response, which has prognostic value, most notably in HER2+ and triple-negative subtypes and can guide the indication of postoperative treatments with a significant reduction in the risk of recurrence, as demonstrated with trastuzumab emtansine and capecitabine in these subtypes, respectively. Consequently, NAC should be preferred for triple-negative and HER2-positive tumors > 2cm [19]. However, NAC could also be discussed for smaller triple-negative and HER2-positive, cT1c, and some-times cT1b tumors, especially for cN1 or pN1sn patients when SLNB is performed before NAC. Indeed, in HER2+ BC, trastuzumab-based adjuvant chemotherapy has demonstrated robust survival benefits in randomized trials in patients with pT1c stage or higher [20-23]. In addition, accumulating data suggest similar benefits even in sub-centimetric tumors, more convincingly in pT1bN0 than pT1aN0 tumors [24]. Of note, the KATHERINE study [13], in which post-operative trastuzumab emtansine was administered in the presence of invasive residual disease after trastuzumab-based NAC, demonstrated a significant improvement in invasive recurrence, 12% of patients had T1 tumors and there was no evidence in favor of a lower benefit compare to higher tumor stage (HR=0.33 [95% CI 0.13-0.88], HR=0.52 [95%CI 0.35-0.78] and HR=0.38 [95%CI 0.23-0.63]) in T1, T2, and T3, respectively). Thus, since trastuzumab-based chemotherapy provides survival advantages and is commonly recommended even in small tumors, and since the absence of pCR may allow an effective rescue, it is tempting to consider a novel paradigm in which virtually any HER2+ tumors > 5mm may be offered NAC. However, during the same period, a relative de-escalation was proposed to T1-T2 (up to 3cm) node-negative HER2-positive BC patients. Thus, in this patient population, a short-duration (12 weeks) anthracycline- and alkylating-free regimen, using weekly paclitaxel in combination with trastuzumab may be proposed with minimal toxicity [25]. Whether such a regimen could similarly be used in the neoadjuvant setting with a trastuzumab emtansine-based rescue strategy warrants further investigation. Similarly, in triple-negative BC patients, adjuvant chemotherapy is largely recommended whatever tumor size (NCCN, ESMO, St-Gallen), even though survival benefit in pT1abN0 remains unclear [26,27]. Once again, in this subtype, adjuvant capecitabine, in patients receiving neoadjuvant anthracycline +/- taxane-based chemotherapy and with the persistence of residual invasive cancer on surgical specimen has been demonstrated to improve survival, according to CREATE-X phase III trial [14]. Of note, in this study, only 14% of patients had initial cT1 stage but in this subgroup, the HR was in favor of capecitabine (HR=0.65 [95%CI 0.30-1.44]) and was similar to the one observed in tumor larger than cT1(HR=0.71 [95%CI 0.53-0.96]), even though it did not reach statistical significance, presumably due to limited sample size. Conflicting results have been reported when considering adjuvant capecitabine in BC patients and most studies have failed to identify significant survival benefits [13,20-23,28-35]. However, subgroup analyses of some individual studies [28] and recently reported meta-analyses have suggested significant improvements in triple-negative BC patients, most of benefits being driven by studies in which NAC was used to select patients with the highest risk [36,37]. Accordingly, adjuvant capecitabine in patients not reaching pCR after NAC has become standard of care in this subtype. Thus, neoadjuvant anthracycline-taxane chemotherapy could be proposed to any pT1b-c triple-negative breast cancer to better define the need for additional cytotoxic such as capecitabine. Recent results of the OLYMPIA trial [38], in which olaparib given to patients with germline BRCA mutation and BC with invasive residual disease (any invasive in triple-negative BC, high CPS+EG score in ER+ BC) after neoadjuvant treatment, was shown to improve significantly survival may further support this approach in this specific subgroup of patients. The rate of conservative surgery appears to be stable over time, with higher rates for tri-ple-negative tumors. However, the rate of conservative surgery depends on many factors, in-cluding the initial tumor size, as we have observed for cT3 and cT4 tumors, and on the patient's choice of whether or not to have a total mastectomy in the presence of hereditary risk or mu-tation, when conservative surgery is feasible. This choice may also have evolved with the increased possibilities and proposals for immediate reconstruction. However, patients must be informed of the aesthetic limitations of immediate or delayed reconstruction [39], postoperative complications [40], and of the risks of altering the aesthetic results in the event of post-mastectomy radiotherapy. As expected, pCR rates following NAC were significantly different according to tumor subtypes, being higher in HER2+ and triple-negative BC. Logically, the use of NAC is currently predominantly dedicated to these subtypes. Importantly, no significant differences in pCR according to tumor size was observed, which also argues in favor of considering NAC even in smaller tumors, as discussed above. There was a trend for a slightly higher pCR in the last period, which did not reach statistical significance. This may be due to the lack of significant change in NAC regimen during the considered periods, most HER2+ patients having access to trastuzumab, while pertuzumab or platinum were not routinely used in France in HER2+ and triple-negative subtypes, respectively. Axillary exploration and treatment have evolved significantly in recent years. The per-formance of an SLNB for cN0 patients (clinical and ultrasound) can be considered before or after NAC [41-43]. SLNB prior NAC for tumors ≤5cm in patients without clinical axillary invasion (cN0) is performed under the same conditions as surgeries performed at the outset. This approach has been proposed for many years [44] and validated more recently by the SENTINA study [41]. In these situations, when the sentinel lymph nodes are not involved, the omission of an additional ALND has been recommended since the results of the NSABP B-32 trial, which validated the sentinel lymph node technique without dissection for immediate surgery [45], and since the results of the SENTINA trial, which included an axillary dissection prior to NAC in one of the randomized arms [41]. In case of sentinel lymph node involvement, completion of ALND after NAC is the attitude that has been usually advocated. However, since the results of the ACOSOG Z0011 and IBCSG 23-01 trials, the omission of completion ALND after NAC can be considered as it is the case for immediate BC surgery, in line with the criteria corresponding to the situations of these trials: presence of at most two macro- or mi-cro-metastases in the sentinel lymph nodes without macroscopic capsular rupture with the realization of breast-conserving surgery, whole-breast radiotherapy, and systemic treatment. Under these conditions, the number of cases with more than 2 sentinel nodes affected being low, an omission of completion ALND would be possible for the vast majority of patients. The presence of micro-metastases or isolated cells is considered to have a prognosis equivalent to pN0sn [19] in case of ER+ tumor [46] but with an unfavorable prognostic value for tri-ple-negative cancers [46]. The disadvantage of this approach is the need for additional surgery, which should not delay the initiation of NAC. On the other hand, the advantage is to precisely know the lymph node stage, in particular to evaluate the indication of regional lymph node radiotherapy, at least in case of macro-metastasis of the sentinel lymph nodes. However, a significant limitation of omitting ALND after initial positive SLND includes the lack of nodal staging allowing to certify pCR status according to the most consensual definition. Yet, in HER2+ and triple-negative BC, a residual nodal involvement when breast pCR is achieved is expected to be uncommon (9.6 to 18.4% in our study). However, the most common approach proposed for cN0 patients, both clinically and ultrasonographically, is to perform an SLNB after NAC [19]. The downstaging linked to the sterilization of initially affected lymph nodes allows considering the omission of an additional ALND in a significant proportion of patients. Conversely, in the case of an invaded node after NAC, a factor associated with poor prognosis, additional ALND is indicated for micro or macro-metastases. The advantage of this approach is to determine the pathologic response of the lymph nodes to NAC. Another advantage is to offer ALND only to a small number of patients, which is probably not higher than the rate of ab-stention from dissection if SLNB is performed before NAC and without dissection for invaded pN0sn and <2 SN. The disadvantage is that the initial lymph node status is not known, which may be the only element determining the indication or not of regional lymph node radiotherapy. In patients with initial limited axillary lymph node involvement confirmed on needle aspiration or biopsy (cN1), SLNB may be considered in selected cases with clinical response in the axillary region, based on the results of the SENTINA and ACOSOG Z1071 trials [41,48]. However, the rate of false negatives remains high overall, from 8% to 14.2 [41,48,49]. This false-negative rate can be reduced by marking the biopsied lymph node (clip or other technique) and by sampling more than 2 sentinel lymph nodes whenever this number is reached by identification [50,51]. In the meta-analysis of Samiei et al. [52] the axillary pCR rate in cN1 patients pathologically proven cN-positive disease was 60% for HER2-positive/ER-negative tumors, 45% for HER2-positive/ER-positive tumors, 48% for triple-negative tumors, and 18% for HER2-negative/ER-positive tumors. These rates were comparable in our study for cN1 patients, 67.3% (74/110) for HER2-positive/ER-negative tumors, 56.1% (87/155) for HER2-positive/ER-positive tumors, 51.7% (104/201) for triple-negative tumors, 35.3% (41/116) for HER2-negative/ER-positive/Grade 3 tumors, and 18.4% (37/201) for HER2-negative/ER-positive/Grade 1-2 tumors. Although our study includes a considerable number of early BC patients receiving NAC on a broad 16-year period, limitations must be addressed. First, potential selection bias and lack of standardization in treatment strategies inherent in the retrospective design of the study. Second, we chose to include patients until May 2021 and thus, assessment of independent causality between pathologic response and outcome was not possible. Third, no specialized pathology review of all cases was undertaken.

5. Conclusions

The pCR rate after NAC was neither higher nor significantly different for tumors <2cm compared with tumors ≥2cm, especially for HER2+ and TN subtypes for which adjuvant therapy can be offered to improve prognosis. The pCR rate appears to correlate with intrinsic tumor characteristics and clinical lymph node status rather than with tumor size. These results suggest that it is possible to propose NAC in patients with these tumor subtypes, in case of clinically invasive axillary lymph nodes (cN1) but also in the absence of suspicious lymph nodes (cN0) when the tumor is smaller than 2cm. An evaluation of overall survival and recurrence-free survival comparing patients with these sizes of tumors and adjusted for other prognostic factors should be performed to confirm these NAC indications.

Acknowledgments

None

Funding

None

Data availability statement

Not applicable.

Authors contribution

Conceptualization: GH, AG and AdN

Methodology: GH, AdN

Formal analysis: GH, AG, AdN

Investigation: GH, AG, AdN

Resources: All authors contributed in patient’s inclusion in the study.

Data curation: GH, AG, AdN

Writing-original draft preparation: GH, AG, AdN

Writing- review and editing: GH, AG, and, MC

Supervision: GH

All authors have read and agreed to the published version of the manuscript

Ethics approval and consent to participate

Not applicable

Patient consent for publication

As this was a retrospective non-interventional study, no formal personal consent was required.

Competing interest

The authors declare that they have no competing interests.

References

  1. Bonadonna G. Karnofsky memorial lecture. Conceptual and practical advances in the management of breast cancer. J Clin Oncol 7 (1989): 1380-1397.
  2. Rubens RD, Sexton S, Tong D, Winter PJ, Knight RK, Hayward JL. Combined chemotherapy and radiotherapy for locally advanced breast cancer. Eur J Cancer 16 (1965): 351-356.
  3. Kaufmann M, Von Minckwitz G, Bear HD, et al. Recommendations from an international expert panel on the use of neoadjuvant (primary) systemic treatment of operable breast cancer: new perspectives 2006. Ann Oncol 18 (2007): 1927-1934.
  4. Asselain B, Barlow W, Bartlett J, et al. Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol 19 (2018): 27-39.
  5. Clough KB, Gouveia PF, Benyahi D, et al. Positive Margins After Oncoplastic Surgery for Breast Cancer. Ann Surg Oncol 22 (2015): 4247-4253.
  6. Houvenaeghel G, Lambaudie E, Bannier M, et al. Positive or close margins: reoperation rate and second conservative resection or total mastectomy? Cancer Manag Res 11 (2019): 2507-2516.
  7. Early Breast Cancer Trialists' Collaborative Group (EBCTCG). Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol 19 (2018): 27-39.
  8. Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci 98 (2001): 10869-10874.
  9. Rouzier R, Perou CM, Symmans WF, et al. Breast Cancer Molecular Subtypes Respond Differently to Preoperative Chemother-apy. Clin Cancer Res 11 (2005): 5678-585.
  10. Cortazar P, Zhang L, Untch M, et al. Patho-logical complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. The Lancet 384 (2014): 164-172.
  11. Broglio KR, Quintana M, Foster M, et al. As-sociation of Pathologic Complete Response to Neoadjuvant Therapy in HER2-Positive Breast Cancer With Long-Term Outcomes: A Meta-Analysis. JAMA Oncol 2 (2016): 751.
  12. Huang M, O’Shaughnessy J, Zhao J, et al. Asso-ciation of Pathologic Complete Response with Long-Term Survival Outcomes in Triple-Negative Breast Cancer: A Meta-Analysis. Cancer Res 80 (2020): 5427-5434.
  13. Von Minckwitz G, Huang CS, Mano MS, et al. Trastuzumab Emtansine for Residual Invasive HER2-Positive Breast Cancer. N Engl J Med 380 (2019): 617-628.
  14. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med 376 (2017): 2147-2159.
  15. Bear HD, Anderson S, Smith RE, et al. Sequential preoperative or postoperative docetaxel added to preoperative doxorubicin plus cy-clophosphamide for operable breast cancer:National Surgical Adjuvant Breast and Bowel Project Protocol B-27. J Clin Oncol Off J Am Soc Clin Oncol 24 (2006): 2019-2027.
  16. Von Minckwitz G, Untch M, Blohmer JU, et al. Definition and Impact of Pathologic Complete Response on Prognosis After Neoad-juvant Chemotherapy in Various Intrinsic Breast Cancer Subtypes. J Clin Oncol 30 (2012): 1796-1804.
  17. Agha R, Abdall-Razak A, Crossley E, et al. The STROCSS 2019 Guideline: Strengthening the Reporting of Cohort Studies in Surgery. International Journal of Surgery 72 (2019): 156-165.
  18. Penault-Llorca F, Balaton A, Sabourin JC, et al. Immunochemistry evaluation of HER2 status in infiltration breast cancer: technical protocol and interpretation guidelines. Ann Pathol 22 (2002): 150-157.
  19. Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol 30 (2019): 1194-220.
  20. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 353 (2005): 1673-1684.
  21. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 353 (2005): 1659-1672.
  22. Slamon D, Eiermann W, Robert N, et al. Adjuvant Trastuzumab in HER2-Positive Breast Cancer. N Engl J Med 365 (2011): 1273-1283.
  23. Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant Docetaxel or Vinorelbine with or without Trastuzumab for Breast Cancer. N Engl J Med 354 (2006): 809-820.
  24. De Nonneville A, Gonçalves A, Zemmour C, et al. Benefit of adjuvant chemotherapy with or without trastuzumab in pT1ab node-negative human epidermal growth factor receptor 2-positive breast carcinomas: results of a national multi-institutional study. Breast Cancer Res Treat (2017).
  25. Tolaney SM, Barry WT, Dang CT, et al. Adjuvant Paclitaxel and Trastuzumab for Node-Negative, HER2-Positive Breast Cancer. N Engl J Med 372 (2015): 134-141.
  26. De Nonneville A, Gonçalves A, Zemmour C, et al. Adjuvant chemotherapy in pT1ab node-negative triple-negative breast carcinomas: Results of a national multi-institutional retrospective study. Eur J Cancer 84 (2017): 34-43.
  27. An X, Lei X, Huang R, et al. Adjuvant chemotherapy for small, lymph node- negative, triple-negative breast cancer: A single-center study and a me-ta-analysis of the published literature. Cancer 126 (2020): 3837-3846.
  28. Lluch A, Barrios CH, Torrecillas L, et al. Phase III Trial of Adjuvant Capecitabine After Standard Neo-/Adjuvant Chemotherapy in Pa-tients With Early Triple-Negative Breast Cancer (GEICAM/2003-11_CIBOMA/2004-01). J Clin Oncol Off J Am Soc Clin Oncol 38 (2020): 203-213.
  29. Joensuu H, Kellokumpu-Lehtinen PL, Huovinen R, et al. Adjuvant Capecitabine in Combination With Docetaxel, Epirubicin, and Cyclophosphamide for Early Breast Cancer. JAMA Oncol 3 (2017): 793-800.
  30. Martín M, Ruiz Simón A, Ruiz Borrego M, et al. Epirubicin Plus Cyclophosphamide Followed by Docetaxel Versus Epirubicin Plus Docetaxel Followed by Capecitabine As Adjuvant Therapy for Node-Positive Early Breast Cancer: Results From the GEICAM/2003-10 Study. J Clin Oncol Off J Am Soc Clin Oncol 33 (2015): 3788-3795.
  31. O’Shaughnessy J, Koeppen H, Xiao Y, et al. Patients with Slowly Proliferative Early Breast Cancer Have Low Five-Year Recurrence Rates in a Phase III Adjuvant Trial of Capecitabine. Clin Cancer Res Off J Am Assoc Cancer Res 21 (2015): 4305-4311.
  32. Von Minckwitz G, Conrad B, Reimer T, Decker T, et al. A randomized phase 2 study comparing EC or CMF versus nab-paclitaxel plus cape-citabine as adjuvant chemotherapy for nonfrail elderly patients with moderate to high-risk early breast cancer (ICE II-GBG 52). Cancer 121 (2015): 3639-3648.
  33. Ohno S, Chow LWC, Sato N, et al. Randomized trial of preoperative docetaxel with or without capecitabine after 4 cycles of 5-fluorouracil- epirubicin- cyclophosphamide (FEC) in early-stage breast cancer: exploratory analyses identify Ki67 as a predictive biomarker for response to neoad-juvant chemotherapy. Breast Cancer Res Treat 142 (2013): 69-80.
  34. Von Minckwitz G, Rezai M, Fasching PA, et al. Survival after adding capecitabine and trastuzumab to neoadjuvant anthracy-cline-taxane-based chemotherapy for primary breast cancer (GBG 40--GeparQuattro). Ann Oncol Off J Eur Soc Med Oncol 25 (2014): 81-89.
  35. Von Minckwitz G, Blohmer JU, Costa SD, et al. Response-Guided Neoadjuvant Chemotherapy for Breast Cancer. J Clin Oncol 31 (2013): 3623-3630.
  36. Xu D, Chen X, Li X, et al. Addition of Capecitabine in Breast Cancer First-line Chemotherapy Improves Survival of Breast Cancer Patients. J Cancer 10 (2019): 418-429.
  37. Natori A, Ethier JL, Amir E, et al. Capecitabine in early breast cancer: A meta-analysis of randomised controlled trials. Eur J Cancer Oxf Engl 77 (2017): 40-47.
  38. Tutt ANJ, Garber JE, Kaufman B, et al. Adjuvant Olaparib for Patients with BRCA1- or BRCA2-Mutated Breast Cancer. N Engl J Med 384 (2021): 2394-2405.
  39. Dauplat J, Kwiatkowski F, Rouanet P, et al. Quality of life after mastectomy with or without immediate breast reconstruction. Br J Surg 104 (2017): 1197-1206.
  40. Quilichini O, Barrou J, Bannier M, et al. Mastectomy with immediate breast reconstruction: Results of a mono-centric 4-years cohort. Ann Med Surg 61 (2012):172-179.
  41. Kuehn T, Bauerfeind I, Fehm T, et al. Senti-nel-lymph-node biopsy in patients with breast cancer before and after neoadjuvant chemotherapy (SENTINA): a prospective, multicentre cohort study. Lancet Oncol 14 (2013): 609-618.
  42. El Hage Chehade H, Headon H, El Tokhy O, et al. Is sen-tinel lymph node biopsy a viable alternative to complete axillary dissection following neoadjuvant chemotherapy in women with node-positive breast cancer at diagnosis? An updated meta-analysis involving 3,398 patients. Am J Surg 212 (2016): 969-981.
  43. Galimberti V, Ribeiro FSK, Maisonneuve P, et al. Sentinel node biopsy after neoadjuvant treatment in breast cancer: Five-year follow-up of patients with clinically node-negative or node-positive disease before treatment. Eur J Surg Oncol J Eur Soc Surg Oncol Br Assoc Surg Oncol 42 (2016): 361-368.
  44. Menard JP, Extra JM, Jacquemier J, et al. Sentinel lymphadenectomy for the staging of clinical axillary node-negative breast cancer before neoadjuvant chemotherapy. Eur J Surg Oncol 35 (2009): 916-920.
  45. 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 (2010): 927-933.
  46. Houvenaeghel G, De Nonneville A, Cohen M, et al. Lack of prognostic impact of sentinel node micro-metastases in endocrine recep-tor-positive early breast cancer: results from a large multicenter cohort. ESMO Open 6 (2021): 100-151.
  47. Houvenaeghel G, Sabatier R, Reyal F, et al. Ax-illary lymph node micrometastases decrease triple-negative early breast cancer survival. Br J Cancer 115 (2016): 1024-1031.
  48. Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, et al. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast cancer: the ACOSOG Z1071 (Alliance) clinical trial. JAMA 310 (2013): 1455-1461.
  49. Classe JM, Loaec C, Gimbergues P, et al. Sentinel lymph node biopsy without axillary lymphadenectomy after neoadjuvant chemo-therapy is accurate and safe for selected patients: the GANEA 2 study. Breast Cancer Res Treat 173 (2019): 343-352.
  50. Caudle AS, Yang WT, Krishnamurthy S, et al. Improved Axillary Evaluation Following Neoadjuvant Therapy for Patients With Node-Positive Breast Cancer Using Selective Evaluation of Clipped Nodes: Imple-mentation of Targeted Axillary Dissection. J Clin Oncol Off J Am Soc Clin Oncol 34 (2016): 1072-1078.
  51. Donker M, Straver ME, Wesseling J, et al. Marking axillary lymph nodes with radioactive iodine seeds for axillary staging after neoad-juvant systemic treatment in breast cancer patients: the MARI procedure. Ann Surg 261 (2015): 378-382.
  52. Samiei S, Simons JM, Engelen SME, et al. Axillary Pathologic Complete Response After Neoadjuvant Systemic Therapy by Breast Cancer Subtype in Patients With Initially Clinically Node-Positive Disease: A Systematic Review and Meta-analysis. JAMA Surg 156 (2021): e210891.

Journal Statistics

Impact Factor: * 4.2

CiteScore: 2.9

Acceptance Rate: 11.01%

Time to first decision: 10.4 days

Time from article received to acceptance: 2-3 weeks

Discover More: Recent Articles

Grant Support Articles

© 2016-2024, Copyrights Fortune Journals. All Rights Reserved!