Predictors of pathological complete response to neoadjuvant treatment in invasive breast cancer with different human epidermal growth factor receptor 2 (HER2) subcategories
Original Article

Predictors of pathological complete response to neoadjuvant treatment in invasive breast cancer with different human epidermal growth factor receptor 2 (HER2) subcategories

Bo Sun1,2,3,4, Yanbo Li2,3,4,5, Jiahui Wang2,3,4,5, Hong Lu2,3,4,5#, Junnan Li2,3,4,5#

1The Second Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China; 2Tianjin’s Clinical Research Center for Cancer, Tianjin, China; 3Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; 4Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; 5Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China

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

#These authors contributed equally to this work.

Correspondence to: Hong Lu, MD; Junnan Li, MD. Department of Breast Imaging, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, West Huan-Hu Rd., Ti Yuan Bei, Hexi District, Tianjin 300060, China; Tianjin’s Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China. Email: luhong_tianjin@163.com; 80280589@qq.com.

Background: Among human epidermal growth factor receptor 2 (HER2)-positive breast cancer patients who receive anti-HER2 treatment, a noteworthy correlation between pathological complete response (pCR) and longer survival has been observed. The rate of pCR varies with the tumor’s degree of HER2 protein expression. The aim of this study was to assess the correlations between clinicopathological characteristics, magnetic resonance imaging (MRI) parameters, and pCR in breast cancer with different HER2 subcategories.

Methods: A total of 281 invasive breast cancer patients diagnosed with HER2-positivity were included. HER2-positive translated to immunohistochemistry (IHC) 3+ or IHC 2+/fluorescence in situ hybridization (FISH)(+). All enrolled patients underwent baseline MRI examination and received neoadjuvant chemotherapy, dual anti-HER2 therapy, and subsequent therapeutic surgery from January 2021 to May 2022. A logistic regression model was used to evaluate the effects of covariates on pCR.

Results: Compared to the IHC 2+/FISH(+) group, patients with IHC 3+ tumors had a higher pCR rate (58.1% vs. 26.7%, P<0.001), clinical stage (58.6% vs. 40%, P=0.038), apparent diffusion coefficient (ADC) value (0.96 vs. 0.88 mm2/s, P=0.004), and were more likely to be estrogen receptor (ER) negative (55.9% vs. 31.1%, P=0.002) and progesterone receptor (PR) negative (72.5% vs. 46.7%, P=0.001). In both groups, univariate analysis showed that the pCR group more often had ER-negative and PR-negative status than the non-pCR group (P<0.001). The final multivariable analysis showed that ER-negativity was associated with pCR in the IHC 2+/FISH(+) group (P=0.004). ER-negativity and the longest diameter were two independent predictors of pCR in the IHC 3+ group (P<0.001 for ER, P=0.026 for longest diameter).

Conclusions: The IHC 3+ group had a higher pCR rate than the IHC 2+/FISH(+) group. Along with clinicopathological characteristics, MRI parameters were supplemental predictors of pCR, particularly in IHC 3+ patients.

Keywords: Human epidermal growth factor receptor 2-positive breast cancer (HER2-positive breast cancer); pathological complete response (pCR); clinicopathological characteristics; magnetic resonance imaging parameter (MRI parameter)


Submitted Feb 29, 2024. Accepted for publication Jul 31, 2024. Published online Aug 19, 2024.

doi: 10.21037/qims-24-397


Introduction

Pathological complete response (pCR), serving as a surrogate for survival evaluation, requires fewer patients and a shorter follow-up period (1,2). Particularly in patients with human epidermal growth factor receptor 2 (HER2)-positive and hormone receptor-negative (HR-negative) tumors, the attainment of a pCR has been strongly correlated with a favorable long-term survival rate after anti-HER2 therapy (3-5).

As research on anti-HER2 therapy has advanced, some studies have found that the response to treatment varies in HER2-positive invasive breast cancer. In previous studies (6,7), a considerable proportion of patients did not respond to dual anti-HER2 therapy. Some experts have emphasized that formulating a personalized anti-HER2 therapy plan based on endocrine responsiveness would be an important challenge in the upcoming years (8). In the KRISTINE study, researchers began to assess the possibility of excluding traditional systemic chemotherapy in the neoadjuvant phase (1). These findings highlight the demand for predictive biomarkers that could indicate the response to new treatment methods.

Predictive biomarkers for response have been investigated in the fields of pathology and radiology. Some researchers have aimed to identify predictive factors for pCR across different HER2-positive categories, and found that HER2 immunohistochemistry (IHC) 3+ and histological grade 3 were independent predictors of pCR for patients receiving anti-HER2 therapy (9). Others similarly revealed that the achievement of pCR was associated with the HER2 IHC expression level and that IHC 3+ was a significant predictor of pCR, in addition to other factors (10). The ACRIN 6657/I-SPY TRIAL observed that compared to clinical assessment, the magnetic resonance imaging (MRI) factor (volumetric of tumor) was a more influential predictor of pathologic response in neoadjuvant chemotherapy (11).

Since the HER2 IHC categories have demonstrated a robust correlation with pCR rates, our objective was to find clinicopathological and MRI features linked to pCR in different categories of HER2 breast cancer. We present this article in accordance with the STROBE reporting checklist (available at https://qims.amegroups.com/article/view/10.21037/qims-24-397/rc).


Methods

Case selection

The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Ethics Committee of Tianjin Medical University Cancer Institute & Hospital (No. bc20240060). The requirement for informed consent was waived due to the retrospective nature of the study. A total of 281 invasive breast cancer patients with HER2 IHC 3+ or IHC 2+/fluorescence in situ hybridization (FISH)(+) were included in this study from January 2021 to May 2022. All patients in our study received trastuzumab and pertuzumab therapy. The exclusion criteria were as follows: (I) lack of FISH results in pretreatment specimens for IHC 2+ tumors; (II) patients who did not receive dual anti-HER2 therapy; (III) lack of baseline magnetic resonance (MR) images in our hospital; (IV) intense artifacts on pretreatment dynamic contrast enhanced-magnetic resonance imaging (DCE-MRI); (V) lack of information on pathological response in the final surgical specimen; and (VI) patients had a history of breast cancer or other cancers.

We assessed the following clinicopathological factors: age, family history, menstrual status, body mass index (BMI), clinical nodal status, clinical stage, histological grade, estrogen receptor (ER), progesterone receptor (PR), and Ki-67. Meanwhile, MR factors included background parenchymal enhancement (BPE), type of lesion, longest diameter, multifocal or multicentric disease, edema, necrosis, kinetics, early enhancement ratio (EER), peak enhancement ratio (PER), late enhancement ratio (LER), time to peak (TTP), and apparent diffusion coefficient (ADC) value.

Pathologic assessment

On the pretreatment specimens of core biopsies, histological grade was assessed according to the Nottingham modification of the Bloom Richardson grading system (12). ER (clone SP1, Zymed) and PR (clone SP2, Zymed) were assessed according to the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) guidelines (13). An expression level on immunohistochemical staining of ≥1% was considered to indicate ER and PR positivity.

At our center, we routinely evaluated the HER2 expression of breast cancer specimens by IHC. HER2 (HercepTest TM; DAKO, Glostrup, Denmark) were also assessed according to ASCO/CAP guidelines. Only membrane staining of the invasive tissues should be considered when scoring HER2. HER2 IHC was scored as positive (3+), equivocal (2+), or negative (0 or 1+) (14). HER2 IHC 3+ tumors required no further testing, and IHC 2+ cases mandated further assessment of HER2 amplification by FISH. HER2 FISH results were interpreted according to the HER2 2018 ASCO/CAP updated guidelines (15).

In all resected specimens, pathologic response was evaluated by an experienced pathologist. It has been demonstrated that a stringent definition of pCR (including lymph node status) is more closely related to better survival than the eradication of tumors from the breast alone (3). In our study, pCR was defined as the absence of detectable residual invasive tumor in breast tissue and the absence of lymph node metastasis, regardless of the presence of residual ductal carcinoma in situ (ypT0/is ypN0).

MRI technique and evaluation

MRI was conducted on a 1.5-T scanner with a dedicated 4-channel phased-array breast coil (Signa Infinity Excite II; GE Healthcare, Chicago, IL, USA) and a 3.0-T scanner with a dedicated 8-channel phased-array breast coil (Discovery MR750; GE Healthcare). The following protocol was used before neoadjuvant chemotherapy: An axial T1-weighted sequence: repetition time (TR) =700 ms; echo time (TE) =10 ms; flip angle =90°; matrix =384×224; field of view (FOV) =30×30 cm; slice thickness =5 mm.

A fat-saturated T2-weighted sequence: TR =4,500 ms, TE =85 ms; flip angle =90°; matrix =384×224; FOV =30×30 cm; slice thickness =5 mm.

Diffusion-weighted imaging (DWI): single-shot echo-planar imaging (EPI) with diffusion-sensitizing gradients: TR/TE =6,300/64 ms; field of view =30×30 cm; slice thickness =5 mm; matrix =128×128; b values =500 and 1,000 s/mm2.

DCE-MRI: A sagittal DCE sequence was obtained before and after administration of contrast agent using the volume imaging for breast assessment (VIBRANT) bilateral breast imaging technique: TR =6.1 ms, TE =2.9 ms, matrix size =256×128, FOV =26 × 26 cm, and slice thickness =1.8 mm.

The paramagnetic contrast agent gadopentetate dimeglumine (Gd-DTPA; 0.2 mL/kg body weight, flow rate 2.0 mL/s) was administered intravenously using a power injector followed by an equal volume of saline solution.

The largest diameter was measured on the slice (plane) as the largest area of the whole tumor region from early post-contrast MRI images that were captured about 90 seconds after the injection of contrast agent. If there were unifocal lesions, the total extent of the lesion was measured; if there were multifocal lesions, the longest diameter of the largest lesion was measured.

Pretreatment MRI examinations were independently interpreted by two radiologists with 4 and 10 respective years of experience in breast imaging, according to the 2013 MRI Breast Imaging Reporting and Data System (BI-RADS) lexicon of the American College of Radiology (16). Both radiologists were blinded to the pathological results. In cases of discordance, consensus was reached through image review and discussion under the BIRADS standard.

Statistical analyses

Statistical analysis was conducted using SPSS 25.0 (IBM Corp., Armonk, NY, USA). Descriptive statistics for each variable are reported. For numeric covariates, the median and interquartile range are presented. Frequencies and percentages are shown for categorical variables. The Mann-Whitney U test was performed to compare numerical covariates, and the chi-square test or Fisher’s exact test was employed to compare categorical covariates (clinicopathological and MRI variables) between different HER2-positive groups.

A logistic regression model was used to evaluate the effect of covariates on pCR in the HER2 IHC 3+ and HER2 IHC 2+/FISH(+) groups. If a variable still had P<0.15, it was incorporated into the final multivariable stepwise regression model. The significance level was set at 0.05.


Results

Clinicopathological and MRI characteristics stratified by HER2 expression

In the pretreatment biopsy specimens, HER2 IHC was 3+ in 236 cases (84%) and IHC 2+/FISH(+) in 45 cases (16%). Most (44/45, 97.8%) of the IHC 2+/FISH(+) cases had a HER2/CEP17 ratio ≥2.0 and ≥4.0 HER2 signals/cell (ASCO/CAP ISH Group 1). One (2.2%) case had a HER2/CEP17 ratio <2.0 but ≥6.0 HER2 signals/cell (ASCO/CAP ISH Group 3). Among the 45 IHC 2+ patients with HER2 amplification detected by FISH, the median HER2/CEP17 ratio by FISH was 2.42 (range, 1.22–6.49), and the median HER2 signals/cell was 6.4 (range, 4.20–16.55).

When we compared the clinicopathological and MRI characteristics of the study cohort across HER2 categories, the key finding was that IHC 3+ tumors had a higher pCR rate than IHC 2+/FISH(+) tumors (58.1% vs. 26.7%, P<0.001) (Figure 1). Similarly, the differences in clinical staging (P=0.04), ER (P=0.002), and PR (P=0.001) expression were statistically significant between IHC 2+/FISH(+) and IHC 3+ groups (Figure 1). Compared with the IHC 2+/FISH(+) group, patients with IHC 3+ tumors had a higher clinical stage (58.6% vs. 40% of clinical stage III, P=0.04), along with more frequent ER-negative status (55.9% vs. 31.1%, P=0.002) and PR-negative status (72.5% vs. 46.7%, P=0.001) (Figure 1). Besides, HER2 IHC 3+ tumors had a higher ADC value than IHC 2+/FISH(+) tumors on MRI (0.96 vs. 0.88 mm2/s, P=0.004) (Figure 1).

Figure 1 Differences in clinicopathological and MRI characteristics between the IHC 2+/FISH(+) and IHC 3+ groups. (A) pCR rate, (B) clinical stage III proportion, (C) ER-negative proportion, (D) PR-negative proportion, and (E) median and IQR of ADC value. pCR, pathological complete response; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; ER, estrogen receptor; PR, progesterone receptor; MRI, magnetic resonance imaging; IQR, interquartile range; ADC, apparent diffusion coefficient.

Identification of predictive factors according to HER2 categories

The associations between clinicopathological and MRI parameters and the attainment of a pCR were examined in the different HER2 categories using univariate and multivariate stepwise regression models.

In the univariate analysis of the IHC 2+/FISH(+) group (Table 1), the differences in ER (P=0.004) and PR (P=0.029) expression were statistically significant between pCR and non-pCR groups (P<0.001 for both). The clinicopathological variables with P<0.15 included family history, clinical stage, and clinical nodal status. Compared with patients in the non-pCR group, patients in the pCR group had a higher frequency of family history (33.3% vs. 9.1%, P=0.06), lower clinical stage (16.7% vs. 48.5% of stage III, P=0.07), and less lymph node metastasis (58.3% vs. 81.8%, P=0.11). Some MRI variables were also incorporated into the multivariate stepwise regression model. In the pCR group, the median longest diameter was smaller (3.4 vs. 5.5 cm, P=0.05), multifocal or multicentric disease was less common (41.7% vs. 72.7%, P=0.06), and TTP was longer (41.7% vs. 15.2% taking 2 mins, P=0.07).

Table 1

Univariate analysis of factors associated with pCR in the IHC2+/FISH(+) group

Parameters Non-pCR pCR OR (95% CI) P value
Age (years) 49 (42, 55.5) 53 (41, 58.5) 1.02 (0.95–1.10) 0.52
Family history of cancer 5.00 (0.93–27.04) 0.06
   No 30 (90.9) 8 (66.7)
   Yes 3 (9.1) 4 (33.3)
Menstrual status 0.91 (0.24–3.49) 0.89
   Postmenopausal 13 (39.4) 5 (41.7)
   Premenopausal 20 (60.6) 7 (58.3)
   Unknown 0 (0) 0 (0)
BMI (kg/m2) 0.86 (0.23–3.26) 0.82
   ≤24 18 (54.5) 7 (58.3)
   >24 15 (45.5) 5 (41.7)
Maximum diameter (cm) 5.5 (3.55, 7.1) 3.4 (2.5, 5.2) 0.70 (0.49–1.00) 0.05
Clinical nodal status 0.31 (0.07–1.33) 0.11
   Negative 6 (18.2) 5 (41.7)
   Positive 27 (81.8) 7 (58.3)
Clinical stage 0.21 (0.04–1.12) 0.07
   II 17 (51.5) 10 (83.3)
   III 16 (48.5) 2 (16.7)
Histological grade 1.37 (0.54–3.50) 0.514
   I/II 19 (57.6) 6 (50.0)
   III 11 (33.3) 4 (33.3)
   Unknown 3 (9.1) 2 (16.7)
ER 0.11 (0.03–0.49) 0.004*
   Negative 6 (18.2) 8 (66.7)
   Positive 27 (81.8) 4 (33.3)
PR 0.19 (0.04–0.84) 0.029*
   Negative 12 (36.4) 9 (75.0)
   Positive 21 (63.6) 3 (25.0)
Ki-67 0.71 (0.06–8.62) 0.79
   ≤20% 2 (6.1) 1 (8.3)
   >20% 31 (93.9) 11 (91.7)
BPE 1.06 (0.28–3.98) 0.93
   Minimal or mild 17 (51.5) 6 (50.0)
   Moderate or marked 16 (48.5) 6 (50.0)
Type of lesion 0.64 (0.24–1.73) 0.38
   NME 4 (12.1) 1 (8.3)
   Mass 13 (39.4) 8 (66.7)
   Mass and NME 16 (48.5) 3 (25.0)
Multifocal or multicentric disease 0.27 (0.07–1.07) 0.06
   Absent 9 (27.3) 7 (58.3)
   Present 24 (72.7) 5 (41.7)
Edema 1.00 (0.25–4.06) 1.00
   Absent 11 (33.3) 4 (33.3)
   Present 22 (66.7) 8 (66.7)
Necrosis 1.12 (0.19–6.72) 0.90
   Absent 28 (84.8) 10 (83.3)
   Present 5 (15.2) 2 (16.7)
Kinetics 0.54 (0.11–2.70) 0.45
   Plateau 5 (15.2) 3 (25.0)
   Washout 28 (84.8) 9 (75.0)
EER (%) 205.0 (180.2, 235.2) 195.4 (167.5, 224.8) 0.10 (0.98–1.02) 0.72
PER (%) 205.0 (185.3, 235.2) 195.4 (173.5, 248.4) 0.10 (0.98–1.02) 0.93
LER (%) 162.4 (152.5, 184.9) 167.9 (140.0, 209.3) 0.10 (0.99–1.02) 0.61
TTP 4.00 (0.90–17.76) 0.07
   1 min 28 (84.8) 7 (58.3)
   2 mins 5 (15.2) 5 (41.7)
ADC value (mm2/s) 0.87 (0.80, 1.0) 0.92 (0.81, 1.08) 14.40 (0.25–840.66) 0.20

Data are presented as median (IQR) or n (%). *, P<0.05. pCR, pathological complete response; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; OR, odds ratio; CI, confidence interval; BMI, body mass index; ER, estrogen receptor; PR, progesterone receptor; BPE, background parenchymal enhancement; NME, non-mass enhancement; EER, early enhancement ratio; PER, peak enhancement ratio; LER, late enhancement ratio; TTP, time to peak; ADC, apparent diffusion coefficient; IQR, interquartile range.

Among those with IHC 3+ tumors (Table 2), univariate analysis confirmed that ER and PR were more often negative in the pCR group than in the non-pCR group (P<0.001 for both). Some MRI features also presented differences between these two groups. The median longest diameter was smaller (5.5 vs. 6.2 cm, P=0.12) and the LER was higher (165.5% vs. 164.5%, P=0.10) in the pCR group than in the non-pCR group. However, these differences were not statistically significant.

Table 2

Univariate analysis of factors associated with pCR in IHC3+ group

Parameters Non-pCR pCR OR (95% CI) P value
Age (years) 50 (39.3, 56.9) 49 (41, 55.5) 0.99 (0.97–1.02) 0.50
Family history of cancer 0.99 (0.49–2.01) 0.98
   No 83 (83.8) 115 (83.9)
   Yes 16 (16.2) 22 (16.1)
Menstrual status 0.91 (0.57–1.45) 0.69
   Postmenopausal 43 (43.4) 61 (44.5)
   Premenopausal 52 (52.5) 73 (53.3)
   Unknown 4 (4.0) 3 (2.2)
BMI (kg/m2) 1.16 (0.69–1.94) 0.58
   ≤24 52 (52.5) 67 (48.9)
   >24 47 (47.5) 70 (51.1)
Maximum diameter (cm) 6.2 (3.7, 7.7) 5.5 (3.3, 7.2) 0.92 (0.83–1.02) 0.12
Clinical nodal status 0.70 (0.38–1.29) 0.25
   Negative 21 (21.2) 38 (27.7)
   Positive 78 (78.8) 99 (72.3)
Clinical stage 0.82 (0.49–1.39) 0.46
   II 40 (40.4) 62 (45.3)
   III 59 (59.6) 75 (54.7)
Histological grade 1.30 (0.91–1.85) 0.15
   I/II 46 (46.5) 57 (41.6)
   III 41 (41.4) 51 (37.2)
   Unknown 12 (12.1) 29 (21.2)
ER 0.29 (0.17–0.49) 0.000*
   Negative 38 (38.4) 94 (68.6)
   Positive 61 (61.6) 43 (31.4)
PR 0.30 (0.17–0.55) 0.000*
   Negative 58 (58.6) 113 (82.5)
   Positive 41 (41.4) 24 (17.5)
Ki-67 0.78 (0.22–2.74) 0.70
   ≤20% 4 (4.0) 7 (5.1)
   >20% 95 (96.0) 130 (94.9)
BPE 0.76 (0.45–1.28) 0.30
   Minimal or mild 57 (57.6) 88 (64.2)
   Moderate or marked 42 (42.4) 49 (35.8)
Type of lesion 0.74 (0.49–1.13) 0.16
   NME 4 (4.0) 15 (10.9)
   Mass 47 (47.5) 62 (45.3)
   Mass and NME 48 (48.5) 60 (43.8)
Multifocal or multicentric disease 1.08 (0.64–1.85) 0.77
   Absent 38 (38.4) 50 (36.5)
   Present 61 (61.6) 87 (63.5)
Edema 1.44 (0.77–2.69) 0.25
   Absent 25 (25.3) 26 (19.0)
   Present 74 (74.7) 111 (81.0)
Necrosis 0.94 (0.43–2.03) 0.87
   Absent 86 (86.9) 120 (87.6)
   Present 13 (13.1) 17 (12.4)
Kinetics 1.27 (0.66–2.44) 0.48
   Plateau 21 (21.2) 24 (17.5)
   Washout 78 (78.8) 113 (82.5)
EER (%) 200.6 (171.8, 229.9) 205.1 (172.4, 241.5) 1.003 (0.997–1.008) 0.30
PER (%) 203.9 (174.9, 233.7) 206.3 (175.7, 245.7) 1.003 (0.997–1.009) 0.28
LER (%) 164.5 (141.1, 186.2) 165.5 (146.5, 196.3) 1.006 (0.999–1.013) 0.10
TTP 0.866 (0.473–1.585) 0.64
   1 min 74 (74.7) 106 (77.4)
   2 mins 25 (25.3) 31 (22.6)
ADC value (mm2/s) 0.96 (0.87, 1.1) 0.96 (0.86, 1.1) 1.23 (0.35–4.32) 0.75

Data are presented as median (IQR) or n (%). *, P<0.05. pCR, pathological complete response; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; OR, odds ratio; CI, confidence interval; BMI, body mass index; ER, estrogen receptor; PR, progesterone receptor; BPE, background parenchymal enhancement; NME, non-mass enhancement; EER, early enhancement ratio; PER, peak enhancement ratio; LER, late enhancement ratio; TTP, time to peak; ADC, apparent diffusion coefficient; IQR, interquartile range.

All variables that still had P<0.15 were incorporated into the final multivariable stepwise regression model. The results showed that ER-negativity was associated with pCR both in the IHC 2+/FISH(+) group [odds ratio (OR) =0.11, 95% confidence interval (CI): 0.03–0.49, P=0.004] and in the IHC 3+ group (OR =0.26, 95% CI: 0.15–0.45, P<0.001). Additionally, the median longest diameter from MRI was also identified as an independent predictor of pCR. The cut-off value was 2.75 cm. A higher pCR rate was identified in the smaller longest diameter group (OR =0.88, 95% CI: 0.79–0.99, P=0.026) (Table 3).

Table 3

Multivariate analysis of factors associated with pCR according to HER2 categories

Parameters Reference B S.E. Wals P value OR (95% CI)
IHC2+ group
   ER −*
+ −2.20 0.76 8.34 0.004 0.11 (0.03–0.49)
IHC3+ group
   ER −*
+ −1.36 0.29 22.59 0.000 0.26 (0.15–0.45)
Maximum diameter −0.12 0.06 4.93 0.026 0.88 (0.79–0.99)

*, control group. pCR, pathological complete response; HER2, human epidermal growth factor receptor 2; S.E., standard error; OR, odds ratio; CI, confidence interval; IHC, immunohistochemistry; ER, estrogen receptor.


Discussion

In this study, we aimed to evaluate clinicopathological and MRI data associated with pCR in different HER2 categories of breast cancer. Our results showed that the pCR rate of IHC 2+/FISH(+) tumors was 26.7%, compared to 58.1% in those with HER2 IHC 3+. This is consistent with earlier studies showing that HER2 IHC 3+ invasive breast cancer had a significantly higher pCR rate than IHC 2+/FISH(+) tumors when double anti-HER2 therapy was given (9,10). Considering that the major targets for anti-HER2 therapy were the extracellular domain of the HER2 protein, the higher pCR rate observed in IHC 3+ tumors may be attributed to higher HER2 protein expression. A previous study (17) found that HER2 IHC 3+ tumors had more HER2 enriched molecular subtype than IHC 2+/FISH(+). Several studies (18,19) concluded that the HER2-enriched biomarker can identify patients who are more likely to achieve pCR after neoadjuvant anti-HER2 therapy.

The pCR rate of HER2 IHC 3+ breast cancer was within the range (38–58%) reported in other clinical trials of single or dual anti-HER2 therapy. In contrast, the pCR rate of IHC 2+/FISH(+) tumors was slightly higher than that in other studies (17–21%) (9,10). This difference may be attributed to the fact that all patients in our study received double anti-HER2 therapy. Trastuzumab and pertuzumab have complementary effects due to their different binding sites. This complementary mechanism of action is reflected in trastuzumab inhibiting ligand-independent signaling, whereas pertuzumab exerts its effects by inhibiting ligand-dependent signaling (20-22).

We found that pathological and MRI characteristics differed between HER2 IHC 3+ and HER2 IHC 2+/FISH(+) tumors: IHC 3+ tumors tended to be larger, to be more often ER-negative and PR-negative, and to have higher ADC values. A previous study reported (23) that HER2 IHC 3+ tumors were also larger, often ER-negative and PR-negative, and had higher histological grades than HER2 IHC 2+/FISH(+) tumors. Combining the results of these studies, IHC 3+ tumors tend to have higher ADC values and histological grades. However, we usually expect higher-grade tumors to generate lower ADC values. Cellularity, as a representative factor for histological grade, has also been confirmed to be inversely correlated with the ADC value by most previous studies (24-26). Although some experts have only found a similar trend without statistical significance (27), 1 study (28) found that the ADC value is positively correlated with tumor cellularity in evaluating the correlation between ADC histogram parameters and prognostic factors and provided an explanation for this result. The assumption that high cellularity would lower ADC values in high-grade tumors was attributed to the subjectivity of the Nottingham modification of the Bloom-Richardson grading system (12,29). However, in addition to subjective evaluation, there were also other related factors, such as the spatial focus of evaluation, the morphology of tumor cells, and cellular displacement. Under the influence of these factors, Kim et al. (27) demonstrated that high-grade tumors had higher ADC values (ADCmax) than low-grade tumors in their study, which was consistent with our result.

In our study, ER-negative status was more common in HER2 IHC 3+ tumors than in the IHC 2+/FISH(+) group. A previous study (30) also observed a negative correlation between ER and HER2 levels in breast cancer. Meanwhile, similar to some previous reports (3,6,9,31), we found that ER status was an independent predictor of pCR in both the IHC 2+/FISH(+) group and the HER2 IHC 3+ group. ER-negative tumors presented a significantly higher pCR rate than ER-positive tumors in both univariate and multivariate analyses. In fact, early preclinical data (32) have shown that estrogen (rather than progestin) was able to change HER-2/neu messenger RNA (mRNA) or protein levels in a dose-dependent manner. This mechanism may further influence the response of HER2-positive tumors to combined chemotherapy and anti-HER2 therapy (33-35). Some previous studies demonstrated that tumors not only exhibited a high dependence on the HER2 gene during growth but also exhibited good response to anti-HER2 therapy in the HR−/HER2+ subtype (35,36).

Since MRI mainly displays cancer through contrast enhancement associated with tumor angiogenesis, we supposed that it could provide both physiological tumor information and more accurate markers of tumor response than simple anatomical imaging. In multiple studies, MRI has better demonstrated the extent of cancer than traditional mammography and ultrasonography (37-40) and is the most accurate modality for evaluating tumor response to neoadjuvant chemotherapy (41-43). To our knowledge, this is the first report to not involve artificial intelligence to predict pCR based on pretreatment MRI data according to the HER2 category of breast cancer. Longest diameter was a significant factor of pCR in the HER2 IHC 3+ group. The ACRIN 6657/I-SPY TRIAL (11) concluded that MR image findings are more predictive of pathologic response to neoadjuvant chemotherapy than clinical assessment. The utilization of volumetric measurements demonstrated the greatest advantage in terms of tumor response early in treatment. Furthermore, another two studies showed that tumor volume measured by MRI was a predictor of recurrence-free survival in breast cancer patients (44,45). Similarly, research on triple-negative breast cancer has indicated that tumor volume measured by pretreatment MRI is an independent predictive factor for pCR (43).

Our study used longest diameter rather than volumetric measurement because we focused on baseline MR images rather than data after neoadjuvant chemotherapy. In another study, initial (before neoadjuvant therapy) MRI diameter and volume measurement methods were substituted in the Cox model, with little loss of predictive value (46). Therefore, MRI diameter can be a reasonable substitute for MRI volume to illustrate the association with pCR before neoadjuvant chemotherapy. This study was also successful in assessing the value of MRI measurements of longest diameter before neoadjuvant chemotherapy treatment for predicting pCR in comparison with established prognostic factors from the clinical and pathological domains.

Our study has some limitations. First, this was a retrospective study, and our small sample was collected from a single center, which may have introduced a selection bias. Second, all pathologic factors were based on core biopsy, which might lead to false-negative results. Previous research demonstrated that core biopsy samples and surgical resection samples may produce discrepant findings with respect to HER2-low status because of intratumoral heterogeneity in protein expression and preanalytical variables (47). Therefore, specimen from core biopsy might lead to false-negative results. Finally, most tumors in the IHC 2+/HER2 amplification group belonged to ASCO/CAP FISH Group 1. As Group 2, 3, and 4 tumors are uncommon (48), our results are not representative of all IHC 2+/HER2 amplification tumors.

In conclusion, the benefit of neoadjuvant anti-HER2 therapy was different between patients with HER2 IHC 3+ tumors and those with IHC 2+/FISH(+) tumors. ER negativity independently predicted pCR in both groups. The median longest diameter measured by MRI was another independent predictor for pCR in the HER2 IHC 3+ group. We anticipate further analyses enrolling larger cohorts to validate these results and the exploration of other research methods, such as artificial intelligence applications, to find more biomarkers for achieving personalized treatment for HER2-positive patients.


Acknowledgments

Funding: This work was funded by Tianjin Key Medical Discipline (Specialty) Construction Project (No. TJYXZDXK-012A) and Cultivation Project of Tianjin Medical University Cancer Institute and Hospital for National Natural Science Foundation of China (Class I, No. 210108).


Footnote

Reporting Checklist: The authors have completed the STROBE reporting checklist. Available at https://qims.amegroups.com/article/view/10.21037/qims-24-397/rc

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://qims.amegroups.com/article/view/10.21037/qims-24-397/coif). All authors report that this work was funded by Tianjin Key Medical Discipline (Specialty) Construction Project (No. TJYXZDXK-012A) and Cultivation Project of Tianjin Medical University Cancer Institute and Hospital for National Natural Science Foundation of China (Class I, No. 210108). The authors have no other conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). The study was approved by the Ethics Committee of Tianjin Medical University Cancer Institute & Hospital (No. bc20240060) and the requirement for informed consent was waived due to the retrospective nature of the study.

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


References

  1. Hurvitz SA, Martin M, Symmans WF, Jung KH, Huang CS, Thompson AM, Harbeck N, Valero V, Stroyakovskiy D, Wildiers H, Campone M, Boileau JF, Beckmann MW, Afenjar K, Fresco R, Helms HJ, Xu J, Lin YG, Sparano J, Slamon D. Neoadjuvant trastuzumab, pertuzumab, and chemotherapy versus trastuzumab emtansine plus pertuzumab in patients with HER2-positive breast cancer (KRISTINE): a randomised, open-label, multicentre, phase 3 trial. Lancet Oncol 2018;19:115-26. [Crossref] [PubMed]
  2. Johnston SRD, Hegg R, Im SA, Park IH, Burdaeva O, Kurteva G, Press MF, Tjulandin S, Iwata H, Simon SD, Kenny S, Sarp S, Izquierdo MA, Williams LS, Gradishar WJ. Phase III, Randomized Study of Dual Human Epidermal Growth Factor Receptor 2 (HER2) Blockade With Lapatinib Plus Trastuzumab in Combination With an Aromatase Inhibitor in Postmenopausal Women With HER2-Positive, Hormone Receptor-Positive Metastatic Breast Cancer: Updated Results of ALTERNATIVE. J Clin Oncol 2021;39:79-89. [Crossref] [PubMed]
  3. Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, et al. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet 2014;384:164-72. [Crossref] [PubMed]
  4. Harbeck N, Gluz O, Christgen M, Kates RE, Braun M, Küemmel S, et al. De-Escalation Strategies in Human Epidermal Growth Factor Receptor 2 (HER2)-Positive Early Breast Cancer (BC): Final Analysis of the West German Study Group Adjuvant Dynamic Marker-Adjusted Personalized Therapy Trial Optimizing Risk Assessment and Therapy Response Prediction in Early BC HER2- and Hormone Receptor-Positive Phase II Randomized Trial-Efficacy, Safety, and Predictive Markers for 12 Weeks of Neoadjuvant Trastuzumab Emtansine With or Without Endocrine Therapy (ET) Versus Trastuzumab Plus ET. J Clin Oncol 2017;35:3046-54. [Crossref] [PubMed]
  5. Chen P, Zhao S, Guo W, Shao G. Dynamic contrast-enhanced magnetic resonance imaging features and apparent diffusion coefficient value of HER2-positive/HR-negative breast carcinoma. Quant Imaging Med Surg 2023;13:4816-25. [Crossref] [PubMed]
  6. Gianni L, Pienkowski T, Im YH, Roman L, Tseng LM, Liu MC, Lluch A, Staroslawska E, de la Haba-Rodriguez J, Im SA, Pedrini JL, Poirier B, Morandi P, Semiglazov V, Srimuninnimit V, Bianchi G, Szado T, Ratnayake J, Ross G, Valagussa P. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol 2012;13:25-32. [Crossref] [PubMed]
  7. Nitz UA, Gluz O, Christgen M, Grischke EM, Augustin D, Kuemmel S, Braun M, Potenberg J, Kohls A, Krauss K, Stefek A, Schumacher C, Forstbauer H, Reimer T, Fischer H, Liedtke C, Wuerstlein R, Schumacher J, Kates R, Kreipe H, Harbeck N. De-escalation strategies in HER2-positive early breast cancer (EBC): final analysis of the WSG-ADAPT HER2+/HR- phase II trial: efficacy, safety, and predictive markers for 12 weeks of neoadjuvant dual blockade with trastuzumab and pertuzumab ± weekly paclitaxel. Ann Oncol 2017;28:2768-72. [Crossref] [PubMed]
  8. Harbeck N. Insights into biology of luminal HER2 vs. enriched HER2 subtypes: Therapeutic implications. Breast 2015;24:S44-8. [Crossref] [PubMed]
  9. Katayama A, Miligy IM, Shiino S, Toss MS, Eldib K, Kurozumi S, et al. Predictors of pathological complete response to neoadjuvant treatment and changes to post-neoadjuvant HER2 status in HER2-positive invasive breast cancer. Mod Pathol 2021;34:1271-81. [Crossref] [PubMed]
  10. Krystel-Whittemore M, Xu J, Brogi E, Ventura K, Patil S, Ross DS, Dang C, Robson M, Norton L, Morrow M, Wen HY. Pathologic complete response rate according to HER2 detection methods in HER2-positive breast cancer treated with neoadjuvant systemic therapy. Breast Cancer Res Treat 2019;177:61-6. [Crossref] [PubMed]
  11. Hylton NM, Blume JD, Bernreuter WK, Pisano ED, Rosen MA, Morris EA, Weatherall PT, Lehman CD, Newstead GM, Polin S, Marques HS, Esserman LJ, Schnall MD. ACRIN 6657 Trial Team and I-SPY 1 TRIAL Investigators. Locally advanced breast cancer: MR imaging for prediction of response to neoadjuvant chemotherapy--results from ACRIN 6657/I-SPY TRIAL. Radiology 2012;263:663-72. [Crossref] [PubMed]
  12. Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 1991;19:403-10. [Crossref] [PubMed]
  13. Allison KH, Hammond MEH, Dowsett M, McKernin SE, Carey LA, Fitzgibbons PL, Hayes DF, Lakhani SR, Chavez-MacGregor M, Perlmutter J, Perou CM, Regan MM, Rimm DL, Symmans WF, Torlakovic EE, Varella L, Viale G, Weisberg TF, McShane LM, Wolff AC. Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Guideline Update. J Clin Oncol 2020;38:1346-66. [Crossref] [PubMed]
  14. Rakha EA, Pinder SE, Bartlett JM, Ibrahim M, Starczynski J, Carder PJ, Provenzano E, Hanby A, Hales S, Lee AH, Ellis IONational Coordinating Committee for Breast Pathology. Updated UK Recommendations for HER2 assessment in breast cancer. J Clin Pathol 2015;68:93-9. [Crossref] [PubMed]
  15. Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, Bilous M, Ellis IO, Fitzgibbons P, Hanna W, Jenkins RB, Press MF, Spears PA, Vance GH, Viale G, McShane LM, Dowsett M. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol 2018;36:2105-22. [Crossref] [PubMed]
  16. A.C.o. Radiology. Breast imaging reporting and data system: BI-RADS atlas. 5th ed., Reston, Va: American College of Radiology, 2013. Available online: https://www.acr.org/Clinical-Resources/Reporting-and-Data-Systems/Bi-Rads#MRI
  17. Atallah NM, Alsaleem M, Toss MS, Mongan NP, Rakha E. Differential response of HER2-positive breast cancer to anti-HER2 therapy based on HER2 protein expression level. Br J Cancer 2023;129:1692-705. [Crossref] [PubMed]
  18. Schettini F, Pascual T, Conte B, Chic N, Brasó-Maristany F, Galván P, et al. HER2-enriched subtype and pathological complete response in HER2-positive breast cancer: A systematic review and meta-analysis. Cancer Treat Rev 2020;84:101965. [Crossref] [PubMed]
  19. Prat A, Pascual T, De Angelis C, Gutierrez C, Llombart-Cussac A, Wang T, et al. HER2-Enriched Subtype and ERBB2 Expression in HER2-Positive Breast Cancer Treated with Dual HER2 Blockade. J Natl Cancer Inst 2020;112:46-54. [Crossref] [PubMed]
  20. Junttila TT, Akita RW, Parsons K, Fields C, Lewis Phillips GD, Friedman LS, Sampath D, Sliwkowski MX. Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell 2009;15:429-40. [Crossref] [PubMed]
  21. Agus DB, Akita RW, Fox WD, Lewis GD, Higgins B, Pisacane PI, Lofgren JA, Tindell C, Evans DP, Maiese K, Scher HI, Sliwkowski MX. Targeting ligand-activated ErbB2 signaling inhibits breast and prostate tumor growth. Cancer Cell 2002;2:127-37. [Crossref] [PubMed]
  22. Nahta R, Hung MC, Esteva FJ. The HER-2-targeting antibodies trastuzumab and pertuzumab synergistically inhibit the survival of breast cancer cells. Cancer Res 2004;64:2343-6. [Crossref] [PubMed]
  23. Giuliani S, Ciniselli CM, Leonardi E, Polla E, Decarli N, Luchini C, Cantaloni C, Gasperetti F, Cazzolli D, Berlanda G, Bernardi D, Pellegrini M, Triolo R, Ferro A, Verderio P, Barbareschi M. In a cohort of breast cancer screened patients the proportion of HER2 positive cases is lower than that earlier reported and pathological characteristics differ between HER2 3+ and HER2 2+/Her2 amplified cases. Virchows Arch 2016;469:45-50. [Crossref] [PubMed]
  24. Razek AA, Gaballa G, Denewer A, Nada N. Invasive ductal carcinoma: correlation of apparent diffusion coefficient value with pathological prognostic factors. NMR Biomed 2010;23:619-23. [Crossref] [PubMed]
  25. Leithner D, Wengert GJ, Helbich TH, Thakur S, Ochoa-Albiztegui RE, Morris EA, Pinker K. Clinical role of breast MRI now and going forward. Clin Radiol 2018;73:700-14. [Crossref] [PubMed]
  26. Hatakenaka M, Soeda H, Yabuuchi H, Matsuo Y, Kamitani T, Oda Y, Tsuneyoshi M, Honda H. Apparent diffusion coefficients of breast tumors: clinical application. Magn Reson Med Sci 2008;7:23-9. [Crossref] [PubMed]
  27. Kim SH, Cha ES, Kim HS, Kang BJ, Choi JJ, Jung JH, Park YG, Suh YJ. Diffusion-weighted imaging of breast cancer: correlation of the apparent diffusion coefficient value with prognostic factors. J Magn Reson Imaging 2009;30:615-20. [Crossref] [PubMed]
  28. Kim EJ, Kim SH, Park GE, Kang BJ, Song BJ, Kim YJ, Lee D, Ahn H, Kim I, Son YH, Grimm R. Histogram analysis of apparent diffusion coefficient at 3.0t: Correlation with prognostic factors and subtypes of invasive ductal carcinoma. J Magn Reson Imaging 2015;42:1666-78. [Crossref] [PubMed]
  29. Rakha EA, El-Sayed ME, Lee AH, Elston CW, Grainge MJ, Hodi Z, Blamey RW, Ellis IO. Prognostic significance of Nottingham histologic grade in invasive breast carcinoma. J Clin Oncol 2008;26:3153-8. [Crossref] [PubMed]
  30. Konecny G, Pauletti G, Pegram M, Untch M, Dandekar S, Aguilar Z, Wilson C, Rong HM, Bauerfeind I, Felber M, Wang HJ, Beryt M, Seshadri R, Hepp H, Slamon DJ. Quantitative association between HER-2/neu and steroid hormone receptors in hormone receptor-positive primary breast cancer. J Natl Cancer Inst 2003;95:142-53. [Crossref] [PubMed]
  31. Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, Tausch C, Seo JH, Tsai YF, Ratnayake J, McNally V, Ross G, Cortés J. Pertuzumab plus trastuzumab in combination with standard neoadjuvant anthracycline-containing and anthracycline-free chemotherapy regimens in patients with HER2-positive early breast cancer: a randomized phase II cardiac safety study (TRYPHAENA). Ann Oncol 2013;24:2278-84. [Crossref] [PubMed]
  32. Read LD, Keith D Jr, Slamon DJ, Katzenellenbogen BS. Hormonal modulation of HER-2/neu protooncogene messenger ribonucleic acid and p185 protein expression in human breast cancer cell lines. Cancer Res 1990;50:3947-51. [PubMed]
  33. Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med 2007;357:39-51. [Crossref] [PubMed]
  34. Kaufman B, Mackey JR, Clemens MR, Bapsy PP, Vaid A, Wardley A, Tjulandin S, Jahn M, Lehle M, Feyereislova A, Révil C, Jones A. Trastuzumab plus anastrozole versus anastrozole alone for the treatment of postmenopausal women with human epidermal growth factor receptor 2-positive, hormone receptor-positive metastatic breast cancer: results from the randomized phase III TAnDEM study. J Clin Oncol 2009;27:5529-37. [Crossref] [PubMed]
  35. Untch M, Gelber RD, Jackisch C, Procter M, Baselga J, Bell R, et al. Estimating the magnitude of trastuzumab effects within patient subgroups in the HERA trial. Ann Oncol 2008;19:1090-6. [Crossref] [PubMed]
  36. Perez EA, Romond EH, Suman VJ, Jeong JH, Sledge G, Geyer CE Jr, Martino S, Rastogi P, Gralow J, Swain SM, Winer EP, Colon-Otero G, Davidson NE, Mamounas E, Zujewski JA, Wolmark N. Trastuzumab plus adjuvant chemotherapy for human epidermal growth factor receptor 2-positive breast cancer: planned joint analysis of overall survival from NSABP B-31 and NCCTG N9831. J Clin Oncol 2014;32:3744-52. [Crossref] [PubMed]
  37. Esserman L, Hylton N, Yassa L, Barclay J, Frankel S, Sickles E. Utility of magnetic resonance imaging in the management of breast cancer: evidence for improved preoperative staging. J Clin Oncol 1999;17:110-9. [Crossref] [PubMed]
  38. Weatherall PT, Evans GF, Metzger GJ, Saborrian MH, Leitch AM. MRI vs. histologic measurement of breast cancer following chemotherapy: comparison with x-ray mammography and palpation. J Magn Reson Imaging 2001;13:868-75. [Crossref] [PubMed]
  39. Van Goethem M, Schelfout K, Dijckmans L, Van Der Auwera JC, Weyler J, Verslegers I, Biltjes I, De Schepper A. MR mammography in the pre-operative staging of breast cancer in patients with dense breast tissue: comparison with mammography and ultrasound. Eur Radiol 2004;14:809-16. [Crossref] [PubMed]
  40. Berg WA, Gutierrez L. NessAiver MS, Carter WB, Bhargavan M, Lewis RS, Ioffe OB. Diagnostic accuracy of mammography, clinical examination, US, and MR imaging in preoperative assessment of breast cancer. Radiology 2004;233:830-49. [Crossref] [PubMed]
  41. Croshaw R, Shapiro-Wright H, Svensson E, Erb K, Julian T. Accuracy of clinical examination, digital mammogram, ultrasound, and MRI in determining postneoadjuvant pathologic tumor response in operable breast cancer patients. Ann Surg Oncol 2011;18:3160-3. [Crossref] [PubMed]
  42. Sheikhbahaei S, Trahan TJ, Xiao J, Taghipour M, Mena E, Connolly RM, Subramaniam RM. FDG-PET/CT and MRI for Evaluation of Pathologic Response to Neoadjuvant Chemotherapy in Patients With Breast Cancer: A Meta-Analysis of Diagnostic Accuracy Studies. Oncologist 2016;21:931-9. [Crossref] [PubMed]
  43. Li Y, Chen Y, Zhao R, Ji Y, Li J, Zhang Y, Lu H. Development and validation of a nomogram based on pretreatment dynamic contrast-enhanced MRI for the prediction of pathologic response after neoadjuvant chemotherapy for triple-negative breast cancer. Eur Radiol 2022;32:1676-87. [Crossref] [PubMed]
  44. Hylton NM, Gatsonis CA, Rosen MA, Lehman CD, Newitt DC, Partridge SC, Bernreuter WK, Pisano ED, Morris EA, Weatherall PT, Polin SM, Newstead GM, Marques HS, Esserman LJ, Schnall MD. ACRIN 6657 Trial Team and I-SPY 1 TRIAL Investigators. Neoadjuvant Chemotherapy for Breast Cancer: Functional Tumor Volume by MR Imaging Predicts Recurrence-free Survival-Results from the ACRIN 6657/CALGB 150007 I-SPY 1 TRIAL. Radiology 2016;279:44-55. [Crossref] [PubMed]
  45. Jafri NF, Newitt DC, Kornak J, Esserman LJ, Joe BN, Hylton NM. Optimized breast MRI functional tumor volume as a biomarker of recurrence-free survival following neoadjuvant chemotherapy. J Magn Reson Imaging 2014;40:476-82. [Crossref] [PubMed]
  46. Partridge SC, Gibbs JE, Lu Y, Esserman LJ, Tripathy D, Wolverton DS, Rugo HS, Hwang ES, Ewing CA, Hylton NM. MRI measurements of breast tumor volume predict response to neoadjuvant chemotherapy and recurrence-free survival. AJR Am J Roentgenol 2005;184:1774-81. [Crossref] [PubMed]
  47. Wang J, Yoon E, Krishnamurthy S. Concordance between pathologists and between specimen types in detection of HER2-low breast carcinoma by immunohistochemistry. Ann Diagn Pathol 2024;70:152288. [Crossref] [PubMed]
  48. Press MF, Sauter G, Buyse M, Fourmanoir H, Quinaux E, Tsao-Wei DD, Eiermann W, Robert N, Pienkowski T, Crown J, Martin M, Valero V, Mackey JR, Bee V, Ma Y, Villalobos I, Campeau A, Mirlacher M, Lindsay MA, Slamon DJ. HER2 Gene Amplification Testing by Fluorescent In Situ Hybridization (FISH): Comparison of the ASCO-College of American Pathologists Guidelines With FISH Scores Used for Enrollment in Breast Cancer International Research Group Clinical Trials. J Clin Oncol 2016;34:3518-28. [Crossref] [PubMed]
Cite this article as: Sun B, Li Y, Wang J, Lu H, Li J. Predictors of pathological complete response to neoadjuvant treatment in invasive breast cancer with different human epidermal growth factor receptor 2 (HER2) subcategories. Quant Imaging Med Surg 2024;14(9):6466-6478. doi: 10.21037/qims-24-397

Download Citation