Next Article in Journal
Mixed Reality Biopsy Navigation System Utilizing Markerless Needle Tracking and Imaging Data Superimposition
Previous Article in Journal
Studying Outcomes after Steroid-Sparing Immunosuppressive Agent vs. Steroid-Only Treatment for Immune-Related Adverse Events in Non-Small-Cell Lung Cancer (NSCLC) and Melanoma: A Retrospective Case-Control Study
Previous Article in Special Issue
Histological Patterns and Mammographic Presentation of Invasive Lobular Carcinoma Show No Obvious Associations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Deciphering the Clinical Behaviour of Invasive Lobular Carcinoma of the Breast Defines an Aggressive Subtype

1
Nottingham Breast Cancer Research Centre, Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
2
Department of Pathology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
3
Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia 32928, Egypt
4
Division of Oncoplastic Surgery, Nottingham Breast Institute, Nottingham University Hospitals, NHS Trust, Nottingham NG5 1PB, UK
5
Department of Histopathology, Nottingham University Hospitals, NHS Trust, Nottingham NG5 1PB, UK
6
Department of Pathology, Hamad Medical Corporation, Doha 3050, Qatar
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(10), 1893; https://doi.org/10.3390/cancers16101893
Submission received: 7 May 2024 / Revised: 11 May 2024 / Accepted: 13 May 2024 / Published: 16 May 2024

Abstract

:

Simple Summary

Invasive lobular carcinoma represents a distinct histological type of breast cancer, characterised by morphological, genetic, and behavioural differences from other types. However, the behaviour of invasive lobular carcinomas is not uniform, and some tumours show poor outcome. This may attribute to the presence of aggressive variants. Therefore, we performed detailed investigation of a large series of cases to characterise this aggressive subtype of invasive lobular carcinoma. This subtype, which includes the pleomorphic and high-grade solid variants accounts for 14% of lobular carcinomas. It showed associations with unfavourable prognostic features, poor patient outcomes, and poor response to therapy when compared to classic lobular carcinoma and invasive ductal carcinoma of no special type.

Abstract

Background: Invasive lobular carcinoma (ILC), the most common special type of breast cancer (BC), has unique clinical behaviour and is different from invasive ductal carcinoma of no special type (IDC-NST). However, ILC further comprises a diverse group of tumours with distinct features. This study aims to examine the clinicopathological and prognostic features of different variants of ILC, with a particular focus on characterising aggressive subtypes. Methods: A large (n = 7140) well-characterised and histologically reviewed BC cohort with treatment and long-term follow-up data was investigated. The cohort was classified based on the WHO classification of tumours into main histological subtypes, including ILC and IDC-NST. ILCs were further classified into variants. Clinicopathological parameters and patient outcomes in terms of BC-specific survival (BCSS) and disease-free survival (DFS) were evaluated. Results: ILC constituted 11% of the cohort. The most common non-classic ILC variants were pleomorphic (pILC) and solid (sILC), constituting 19% of ILC. Compared to classic and related variants (alveolar, trabecular, papillary, and tubulolobular; cILC), pILC and sILC variants were associated with aggressive tumour characteristics. The histologic grade of ILC was an important prognostic variable. The survival patterns identified an aggressive ILC subtype encompassing pILC and high-grade sILC. These tumours, which comprised 14% of the cases, were associated with clinicopathological characteristics of poor prognosis and had high BC-specific death and recurrence rates compared not only to cILC (p < 0.001) but also to IDC-NST (p = 0.02) patients. Contrasting this, cILC patients had significantly longer BCSS and DFS than IDC-NST patients in the first 10 to 15 years of follow-up. Adjuvant chemotherapy did not improve the outcome of patients with aggressive ILC subtypes. Conclusions: pILC and high-grade sILC variants comprise an aggressive ILC subtype associated with poor prognostic characteristics and a poor response to chemotherapy. These results warrant confirmation in randomised clinical trials.

1. Introduction

Invasive lobular carcinoma (ILC) is the second most common breast cancer (BC) histological type after invasive ductal carcinoma of no special type (IDC-NST) [1]. In recent years, the incidence of ILC has increased from 5% to 10–15% due to improved diagnostic and surveillance approaches [2].
ILC has distinct morphological features, molecular signatures, and clinical behaviour that are different from those of IDC-NST and other special types of BC [3]. Loss of E-cadherin function, diffuse growth pattern, and low proliferative activity, in addition to oestrogen receptor (ER) expression (>90%) and human epidermal growth factor 2 (HER2) negativity, are characteristic features of ILC [4,5,6]. ILC also shows unique metastatic patterns with involvement of sites less commonly involved by IDC-NST [7], including the gastrointestinal tract, with the stomach being the most commonly affected site [8], genitourinary tract, retroperitoneum, and peritoneum [9]. Early metastatic ILC also tends to be infiltrative rather than mass-forming [10]. Although classical ILC is characterised by cells with small nuclei and scanty cytoplasm, a dyscohesive growth pattern, arrangement in single strands with minimal stromal reaction, and infiltration around breast ductal units (targetoid lesions) [6,11], cytological and architectural variations exist. ILC cells may exhibit a high degree of pleomorphism, warranting the diagnosis of a pleomorphic ILC variant (pILC) or displaying diffuse solid infiltration in the solid variant (sILC). ILC cells may also be arranged in tubules, clusters, or trabeculae in tubulolobular, alveolar, and trabecular variants, respectively [4,12,13].
As IDC-NST is the most common type of BC, accounting for more than 60% of cases, it has steered the majority of BC research, and differences between various histological types have been less investigated [3]. For a long time, patients with ILC were treated using the same protocols used for IDC-NST. However, recent studies have shown that ILC patients respond less to chemotherapeutic agents [14,15,16]. Therefore, there is now greater awareness of the different invasive BC subtypes, including the unique characteristics of ILC, and a move towards personalised treatment approaches.
Although most of the previous studies have reported the clinical behaviour of ILC based on the most common variant, classical ILC (cILC), and on comparison between the cILC and IDC-NST [2,17,18,19,20,21], there is increasing evidence that some variants of ILC have distinct and aggressive clinical behaviour. pILC is the variant of ILC associated with negative prognostic factors, mainly ER negativity and HER2 positivity [22,23]. However, the consideration of pILC as a separate entity and its impact on patient management remains controversial, and most patients with pILC are managed in the same way as cILC [24]. Similarly, some authors have reported that sILC is an aggressive, highly proliferative variant of ILC compared with cILC [12]; however, studies addressing the characteristics of this variant remain lacking.
Previous studies have demonstrated the poor response of ILC patients to neoadjuvant chemotherapy [3,12,25,26]. However, the response of ILC variants to neoadjuvant therapies remains unclear [27]. Moreover, it is unclear if the difference in the clinical behaviour and response to therapy of ILC variants is related to the degree of proliferation (histological grade), receptor status, or the histological subtypes of ILC [28].
Therefore, this study aims to characterise ILC variants for management purposes utilising a large well-characterised cohort of BC with long-term follow-up coupled with a detailed histological review.

2. Materials and Methods

2.1. Study Cohort

A well-characterised consecutive cohort (n = 7140) of patients presented to Nottingham City Hospital from 1998 to 2018 with early-stage (TNM cT1-2, cN0-3, M0) operable BC was investigated. Patients with locally advanced or metastatic BC or those who did not undergo surgery for any reason or received neoadjuvant therapy were not included in this study. Loco-regional radiological staging was performed using ultrasound, but MRI was used in cases diagnosed as ILC on the core biopsy according to local protocol. Clinicopathological data, including patient age, tumour histological type, tumour size, tumour grade and its components, lymph node (LN) status, lymphovascular invasion (LVI), and Nottingham Prognostic Index (NPI), were retrieved from the data repository. Biomarker expression data, including ER, progesterone receptor (PR), HER2, and Ki67 index, were collected from patient reports. Hormone receptors and HER2 were scored according to updated UK guidelines and American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) guidelines [29,30,31]. Ki67 > 14% was considered a high index [32]. Oncotype Dx recurrence scores (RS) were also available in a subset of tumours (n = 431). RS of <11, 11–25, and >25 were defined as low, intermediate, and high, respectively [33]. Follow-up data, including BC-specific survival (BCSS), defined as the time from the initial surgery to death related to BC, and disease-free survival (DFS), defined as the time from the initial surgery to the development of any recurrence event of disease, in addition to the site of recurrences, were collected.
Adjuvant systemic therapies were given following a multidisciplinary team decision according to ER status, NPI, menopausal status, and associated comorbidities. ER-positive BC patients with good prognostic NPI received endocrine therapy (ET). Chemotherapy regimens were given to ER-negative patients. ER + BC patients with moderate and poor prognostic NPI (>4.4) received ET with combined chemotherapy if patients were fit to tolerate chemotherapy. In later years of the study, HER2 status, Oncotype Dx RS, and TNM stage were also considered in the decision of adjuvant therapy (A subset of patients with NPI 3.4–4.4 were tested using the Oncotype Dx assay for adjuvant chemotherapy, and most HER2 positive patients receive adjuvant Herceptin in addition to chemotherapy). ET was administered as the only adjuvant therapy in 47% of the cohort; 14% of patients received only chemotherapy; and 14% received both ET and chemotherapy. Adjuvant radiotherapy was applied according to local protocols.

2.2. Invasive Lobular Carcinoma Histological Assessment

All cases diagnosed as ILC in routine practice, where histological slides were available (72% of cases), were reviewed by a certified pathologist (SM). In Nottingham, the diagnosis of lobular type and variants is based on morphology, while E-cadherin use is limited to occasional cases that show overlapping features between ILC and ductal carcinoma [34]. None of the cases was designated as pILC in this study unless it was histologically reviewed, and the classification was approved by all the observers. Variants were classified according to the 5th edition of the WHO Classification of Tumours [35] into classic, pleomorphic, solid, tubulolobular, alveolar, trabecular, histiocytoid, and signet ring. Mixed variants encompassing more than one variant were reclassified and assigned the potentially aggressive variant. In cases of discordance with the original reports, discordant cases were reviewed by other experienced pathologists (ER and NA) until a consensus was reached.
cILC exhibited small to medium-sized dyscohesive cells arranged in single files or targetoid patterns with low to intermediate-grade nuclei. ILC cells with marked cellular pleomorphism with or without apocrine morphology were classified as pILC. sILC displayed a solid growth pattern of small lobular cells arranged in large, solid sheets with scarce intervening stroma. sILC often mimics IDC-NST but lacks polarisation of the cells, which are E-cadherin negative. The few cases with solid growth patterns but marked cellular pleomorphism were classified as pILC. If the solid growth pattern shows papillary architecture, ILC is classified as papillary ILC. Arrangement in broad trabeculae or clusters of at least 20 cells was diagnosed as trabecular and alveolar variants, respectively. When cells exhibit abundant granular foamy cytoplasm, histiocytoid differentiation is recognised, while signet ring carcinoma features intracytoplasmic accumulation resembling signet ring cells [3,4,6,11,35,36,37] (Figure 1). In routine practice, E-cadherin immunohistochemistry was used in cases with overlapping features, and in such a case where the tumour was E-cadherin negative, the classification of ILC was confirmed. In cases with classic morphology, the classification of ILC was confirmed without using E-cadherin, as 10% of ILC with classic morphology show E-cadherin membrane expression [34].

2.3. Statistical Analysis

Statistical Package SPSS v28 for Windows (Chicago, IL, USA) was used. The chi-square test was used to compare categorical groups. Inter-observer agreement was determined using the intra-class correlation coefficient (ICC). Kaplan–Meier curves and log-rank tests were used for survival analysis. The mean patient follow-up was 127 months. A p-value of less than 0.05 (two-tailed) was considered significant in all the statistical tests.

3. Results

3.1. Patient and Tumour Characteristics

Eleven percent of the cohort was classified as ILC, 63% as IDC-NST, and 26% as other special or mixed histological types. The mean patient age was 57 years (ranging from 32 to 87), 59 years in ILC, and 56 years in IDC-NST. Eighteen percent of the cohort was classified as grade 1, while 43% and 39% were grade 2 and 3 tumours, respectively. High-grade (grade 3) tumours comprised 56% of IDC-NST and 8% of ILC. ER was positive in 80% of the cohort, 96% in ILC, and 71% in IDC-NST.
The mean tumour size of the cohort was 19 mm. However, ILC had a mean size of 23 mm, while IDC-NST had a mean size of 19 mm. LN metastasis was detected in 34% of the whole cohort, 33% of ILC, and 35% of IDC-NST patients. However, when the analysis was limited to grade-matched cases, the frequency of LN positivity was 33% in ILC compared to 31% in IDC-NST. Of the LN-positive patients, the mean number of positive LN was 3.9 in ILC and 3.0 in IDC-NST. In this early-stage cohort, 54% of patients were treated with breast-conserving therapy, 47% of ILC patients, and 53% of patients with IDC-NST.

3.2. Invasive Lobular Carcinoma Variants

In this study, a few cases were classified as tubulolobular, trabecular, alveolar, and papillary variants, and these were classified as grade 1 and 2 tumours, had small lobular carcinoma cells, and were combined with the classic variant as related variants. The classic and related variants (cILC) comprised 81% of ILC, while 19% of ILC exhibited pleomorphic, solid, histiocytoid, and signet ring morphologies (Figure 1). pILC and sILC constituted most of the non-classic variants (67% and 29%, respectively). The histiocytoid and signet ring variants were insufficiently represented for meaningful statistical analyses and were excluded from subsequent outcome analyses.

3.3. Correlation between ILC Variants and Clinicopathological Characteristics

pILC was significantly associated with poor tumour characteristics, including a higher histological grade, a larger tumour size, LN metastasis, a higher probability of LVI, a poorer NPI prognostic group, ER and PR negativity, HER2 overexpression, a higher Ki67 index, and a younger patient age compared to cILC (Table 1). Compared to IDC-NST, pILC revealed a significant association with larger tumour size, LN metastasis, and poorer NPI; however, it displayed a lower histological grade, mitotic count, and Ki67 index than IDC-NST.
sILC showed a significantly higher histological grade, mitotic count, and Ki67 index than cILC. However, sILC had a lower histological grade and mitotic count than IDC-NST (Table 1). Comparing the two variants, pleomorphic and solid, pILC was significantly associated with a higher grade, larger tumour size, LN metastasis, and poorer NPI than sILC. However, a higher Ki67 index was shown in the solid variant than in the pleomorphic variant.
In contrast to the aggressive features seen in pILC and sILC, cILC was associated with favourable characteristics compared to IDC-NST. cILC was more frequently associated with older age, lower grade and grade components, and good NPI. Moreover, cILC was more frequently of low- and intermediate-risk Oncotype Dx RS than IDC-NST (Table 1).

3.4. Outcome Analyses

cILC patients displayed BC-specific death in 14% and recurrence in 26% of patients, compared to 19% and 29%, respectively, of IDC-NST patients. Patients with the cILC subtype had longer BCSS (HR = 0.7, 95% CI = 0.5–0.8, p < 0.001) and DFS (HR = 0.8, 95% CI = 0.7–0.9, p = 0.008) compared to IDC-NST patients. However, cILC continued to develop events over time, and the significance decreased at 10–15 years (Figure 2).
The clinicopathological characteristics and survival patterns of ILC variants were further examined to identify the aggressive, high-risk ILC variants. As both pILC and sILC showed aggressive features compared to cILC, we aimed to determine whether to include the whole variants or stratify them based on a combination of variant and grade as histological grade played a significant prognostic role (A significantly shorter BCSS was observed in patients with grade 3 ILC subtype compared to those with grade 2 pILC; HR = 2.2, 95% CI = 1.1–4.6, p = 0.04).
Grade 2 pILC was associated with significantly younger patient age, larger tumour size, and LVI than cILC (Supplementary Table S1). With a mean patient follow-up of 127 months, the BC-specific death of pILC was 28%, while 43% of pILC patients experienced recurrence. Within grade 2 pILC, BC-specific death and recurrence were seen in 23% and 40% of the cases, respectively. In grade 3 pILC, 34% and 46% of patients experienced BC-specific death and recurrence, respectively.
Grade 2 sILC did not show significant differences from cILC, apart from the characteristic higher proliferation manifested in the mitotic score and Ki-67 index. sILC patients experienced BC-specific death in 19% of cases and recurrence in 29% of cases. However, grade stratification revealed a more aggressive survival pattern associated with grade 3 sILC, where 22% and 33% of patients died and developed recurrence, respectively, compared to 18% and 27%, respectively, of grade 2 sILC. When compared with cILC patients, no outcome differences were identified for grade 2 sILC, whereas significant BCSS and DFS differences were identified between cILC and pILC grades (p < 0.001) (Supplementary Figure S1).
Therefore, pILC, where both grades displayed aggressive behaviour, and grade 3 sILC patients, that were associated with higher risk, were combined in one group ‘aggressive ILC subtype’, which comprised 14% of ILC and was further analysed.
The aggressive ILC subtype was associated with large tumour size, positive LN metastasis (46%), high grade (grade 3 in 50%), and a high Ki-67 index. Over 80% had NPI scored as moderate and poor prognostic groups. All characteristics are summarised in Table 2.
Statistically significant outcome differences were observed between ILC subtypes and IDC-NST (p = 0.001 and p = 0.004, for BCSS and DFS, respectively) (Figure 3).
Compared to cILC, patients with the aggressive ILC subtype suffered significantly shorter BCSS and DFS (HR = 2.2, 95% CI = 1.4–3.4, p < 0.001, and HR = 1.8, 95% CI = 1.3–2.5, p < 0.001). Moreover, worse DFS was associated with an aggressive ILC subtype compared to IDC-NST patients (HR = 1.4, 95% CI = 1.1–1.9, p = 0.02) (Figure 3).
Multivariate Cox regression analyses involving the ILC subtypes (classic versus aggressive) and adjusted for known prognostic parameters: NPI, ER status, and HER2 status revealed an independent prognostic significance for shorter BCSS (HR = 1.7, 95% CI = 1.1–2.7, p = 0.01) and shorter DFS (HR = 1.5, 95% CI = 1.1–2.1, p = 0.02) associated with aggressive ILC subtype. The same results were associated with the aggressive ILC subtype compared to IDC-NST (HR = 1.6, 95% CI = 1.1–2.3, p = 0.006 for BCSS, HR = 1.5, 95% CI = 1.2–2.0, p = 0.003 for DFS) in a multivariate model. However, cILC could not retain a significant difference from IDC-NST when tested in multivariate analyses (Supplementary Table S2).
In the context of adjuvant therapies, patients with aggressive ILC subtypes who received only adjuvant ET exhibited worse survival outcomes compared to those with cILC (HR = 2.8, 95% CI = 1.5–5.2, p = 0.002 for BCSS and HR = 1.8, 95% CI = 1.1–3.1, p = 0.03 for DFS). Similar results were obtained when survival analyses were restricted to patients who received adjuvant ET and chemotherapy, where patients with aggressive ILC subtypes remained to show shorter BCSS (HR = 2.9, 95% CI = 1.1–8, p = 0.03) compared to cILC patients (Figure 4).
Patients with aggressive ILC subtypes also demonstrated shorter BCSS (HR = 1.8, 95% CI = 1.1–3.0, p = 0.03) compared to IDC-NST patients when both were treated with adjuvant ET alone and shorter DFS (HR = 1.9, 95% CI = 1.1–3.2, p = 0.02) when treated with adjuvant ET and chemotherapy (Figure 5).
Compared to IDC-NST, cILC patients showed a favourable response to adjuvant ET (HR = 0.6, 95% CI = 0.4–0.9, p = 0.01 and HR = 0.7, 95% CI = 0.6–0.9, p = 0.03 for BCSS and DFS, respectively) (Figure 6), but when adjuvant chemotherapy was given, IDC-NST patients’ outcomes improved and the survival differences between cILC and IDC-NST patients became insignificant.

4. Discussion

The most common special histological subtype, ILC, has shown a rise in incidence in recent years because of the significant advancements in imaging modalities [6]. The unique characteristic morphology and behaviour of ILC have captivated the clinical interest in this special subtype [11]. However, the clinical outcomes of ILC vary widely in the literature and are conflicting, mandating further investigation in large, well-characterised cohorts [12].
Investigating a large, well-characterised, and histologically reviewed cohort of early-stage (cT1-2) operable BC patients who were treated uniformly in Nottingham with long-term follow-up (>20 years), we demonstrated favourable survival outcomes of cILC compared to IDC-NST. Nevertheless, a tendency toward BC-specific death and recurrence was noticed at 15 years. These findings are in line with Chamalidou et al. [17], where an excess mortality rate ratio was calculated and more favourable outcomes of ILC than IDC-NST were observed at 5 years while significantly decreasing at 10–15 years after diagnosis and then similar after 20 years [17].
Worse outcomes associated with ILC than IDC-NST were reported [18,20,38], while others revealed similar outcomes [1,2,19,21,39,40]. This variation in outcome findings is likely to reflect the variability in the ILC cohorts with variable representations of ILC histological variants [2,3,17]. Therefore, we aimed to refine the prognosis of ILC variants. We identified various morphological variants, where pILC and sILC comprised 19% of our cohort. Few studies addressed the prognostic relevance of ILC variants, which revealed comparable proportions [12,27].
Thorough analyses of the survival patterns of pILC and sILC could identify a subgroup of ILC with aggressive behaviour. By stratifying ILC variants based on histological grade, none of the cILC were classified as grade 3 tumours. Both grades of pILC seemed to behave worse than cILC, while only grade 3 sILC was more aggressive than cILC. We combined these two tumour types into one group that was termed the “aggressive ILC subtype”. The aggressive subtype accounts for 14% of ILC cases, requiring further clinical research and potentially contributing to over 1000 cases annually in the UK based on recent BC statistics [41].
This subtype showed poorer outcomes not only when compared to the classic ILC and related variants (cILC) but also to IDC-NST. Poor tumour characteristics were observed in the aggressive ILC subtypes, including larger tumour size, younger patient age, LN metastasis, higher proliferative activity, and more frequent ER negativity and HER2 positivity. These findings confirm the aggressive behaviour stated by previous studies [13,22,23] and are in line with previous studies on pILC [12,22,42]. pILC was previously reported to have worse overall survival than IDC-NST [13], similar to the shorter survival associated with the aggressive ILC subtype observed in our study. However, there is a debate about considering pILC as a standalone and independent prognostic variable and whether it is a variant or grade that should be considered [23,24,43]. It is worth noting that pILC has a distinct genetic profile compared to cILC. The former is more likely to have a TP53 mutation (11–42% of pILC, compared to less than 10% of cILC), as well as complex DNA copy number alterations, FER kinase expression, altered DNA methylation patterns, and mutations in both IRS2 and IGFR [44,45].
The clinical behaviour of sILC is less defined, and only a limited number of cases were studied, which showed an association between sILC and shorter survival compared to other ILC variants [12]. This study found that sILC is associated with higher grades, pleomorphism, mitosis, and ki-67 proliferation index than cILC, in addition to an association with worse outcomes. Therefore, sILC was further investigated, and this showed that the worse outcome compared to cILC was limited to grade 3 tumours. Other ILC variants, including histiocytoid and signet-ring morphologies, were reported as aggressive variants [46,47,48,49]. However, the data about these variants are scarce and were excluded from our analyses due to their limited number.
As a predominantly ER + BC subtype, ILC patients are offered ET, while the response to chemotherapy is said to be less sensitive [6]. The same survival patterns for ILC and its variants were present, as expected when endocrine-treated patients were selected, while the high-risk chemotherapy-treated ILC patients were not responsive compared to IDC-NST. Chemotherapy did not seem beneficial for improved survival outcomes in several reports [2,14,16,50].
However, in our study, aggressive ILC subtypes were associated with a worse chemotherapy response than classic variants and IDC-NST. The poor response of aggressive ILC to both ET and chemotherapies revealed in this study confirms the need for further large studies and trials investigating additional therapies tailored to these specific subtypes.
Response to neoadjuvant chemotherapy was previously reported to be higher in IDC-NST than ILC, which confirms the decreased sensitivity of ILC to chemotherapeutic agents. pCR rates ranged in previous reports between 0–11% in ILC patients while reaching up to 70% in IDC-NST patients [25,26,27,51] as the pCR rate varies widely according to the molecular subtype [52]. There is a debate on whether the low pCR rates in ILC versus IDC-NST are due to differences in ER expression, where ILC, which was ER-negative and/or HER2+, had a pCR rate of 25% [25,53]. A previous study demonstrated that the pCR rate was 4.2% in ER-positive, grade 1 or 2 ILC, 7.0% in ILC with either ER-negative or grade 3, and 17.8% in ER-negative grade 3 ILC [54]. When the variant of ILC was considered, Mamtani and colleagues [27] demonstrated a higher pCR of pILC than cILC patients (16% of pILC compared to only 1% of cILC).
Evaluation of Oncotype Dx RS in ILC histology has been tried previously, where the consistent association of ILC to low RS was revealed [50,55,56], supporting our results. Weiser et al. [57] found a role for RS in predicting ILC prognosis; however, our cohort’s survival data for Oncotype Dx are insufficient to confirm these results.
The study has limitations. First, the cohort is retrospective, and further validation in a randomised clinical trial is warranted. Second, the rarity of some ILC variants, mainly the signet ring cell variant and the histiocytoid variant, did not allow for studying their clinical behaviour and whether these tumours should be combined with cILC or the aggressive subtype.

5. Conclusions

In conclusion, ILC encompasses a mixture of variants with different clinical behaviours. pILC and high-grade sILC variants comprise an aggressive ILC subtype. Aggressive ILC variants are associated with poor prognostic characteristics, are at higher risk of recurrence and BC-related death than cILC patients, and show a poor response to chemotherapy. The poor response of the aggressive ILC group to chemotherapy requires further investigation for innovative and tailored therapies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers16101893/s1, Supplementary Figure S1: Kaplan Meier survival curves show shorter breast cancer-specific survival (A) and disease-free survival (B) associated with pleomorphic invasive lobular carcinoma (pILC) compared to classic ILC (cILC); Supplementary Table S1: Clinicopathological characteristics of grade 2 pleomorphic invasive lobular carcinoma (pILC) and grade 2 solid ILC (sILC) in comparison to classic ILC (cILC); Supplementary Table S2: Multivariate Cox Regression analysis shows prognostic variables for breast cancer-specific survival and disease-free survival.

Author Contributions

Conceptualization, S.M. and E.A.R.; Data curation, A.H.S.L. and E.A.R.; Formal analysis, S.M., N.M.A., A.H.S.L. and E.A.R.; Writing—original draft, S.M. and E.A.R.; Writing—review and editing, N.M.A., S.P., A.H.S.L., A.R.G. and E.A.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was approved by the Yorkshire and the Humber-Leeds East Research Ethics Committee (REC Reference: 19/YH/0293) under the IRAS Project ID: 266925, granted 23/08/2019. The data collected were fully anonymised. All procedures performed in the study were in accordance with the Declaration of Helsinki.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in the current study are available upon reasonable request.

Acknowledgments

SM is supported by the Egyptian Ministry of Higher Education and Scientific Research.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. García-Fernández, A.; Lain, J.M.; Chabrera, C.; García Font, M.; Fraile, M.; Barco, I.; Torras, M.; Reñe, A.; González, S.; González, C. Comparative long-term study of a large series of patients with invasive ductal carcinoma and invasive lobular carcinoma. Loco-regional recurrence, metastasis, and survival. Breast J. 2015, 21, 533–537. [Google Scholar] [CrossRef]
  2. Yang, C.; Lei, C.; Zhang, Y.; Zhang, J.; Ji, F.; Pan, W.; Zhang, L.; Gao, H.; Yang, M.; Li, J. Comparison of overall survival between invasive lobular breast carcinoma and invasive ductal breast carcinoma: A propensity score matching study based on SEER database. Front. Oncol. 2020, 10, 590643. [Google Scholar] [CrossRef]
  3. Mouabbi, J.A.; Hassan, A.; Lim, B.; Hortobagyi, G.N.; Tripathy, D.; Layman, R.M. Invasive lobular carcinoma: An understudied emergent subtype of breast cancer. Breast Cancer Res. Treat. 2022, 193, 253–264. [Google Scholar] [CrossRef]
  4. Reed, A.E.M.; Kutasovic, J.R.; Lakhani, S.R.; Simpson, P.T. Invasive lobular carcinoma of the breast: Morphology, biomarkers and ’omics. Breast Cancer Res. 2015, 17, 12. [Google Scholar] [CrossRef]
  5. Rakha, E.A.; El-Sayed, M.E.; Powe, D.G.; Green, A.R.; Habashy, H.; Grainge, M.J.; Robertson, J.F.; Blamey, R.; Gee, J.; Nicholson, R.I. Invasive lobular carcinoma of the breast: Response to hormonal therapy and outcomes. Eur. J. Cancer 2008, 44, 73–83. [Google Scholar] [CrossRef]
  6. De Schepper, M.; Vincent-Salomon, A.; Christgen, M.; Van Baelen, K.; Richard, F.; Tsuda, H.; Kurozumi, S.; Brito, M.J.; Cserni, G.; Schnitt, S. Results of a worldwide survey on the currently used histopathological diagnostic criteria for invasive lobular breast cancer. Mod. Pathol. 2022, 35, 1812–1820. [Google Scholar] [CrossRef]
  7. Mathew, A.; Rajagopal, P.S.; Villgran, V.; Sandhu, G.S.; Jankowitz, R.C.; Jacob, M.; Rosenzweig, M.; Oesterreich, S.; Brufsky, A. Distinct pattern of metastases in patients with invasive lobular carcinoma of the breast. Geburtshilfe Frauenheilkd. 2017, 77, 660–666. [Google Scholar] [CrossRef]
  8. Barbieri, E.; Caraceni, G.; Gentile, D.; Gavazzi, F.; Zerbi, A.; Tinterri, C. A rare case of duodenal metastasis from lobular breast cancer: From diagnosis to surgery. Case Rep. Oncol. 2023, 16, 391–396. [Google Scholar] [CrossRef]
  9. Duraker, N.; Hot, S.; Akan, A.; Nayır, P. A Comparison of the Clinicopathological Features, Metastasis Sites and Survival Outcomes of Invasive Lobular, Invasive Ductal and Mixed Invasive Ductal and Lobular Breast Carcinoma. Eur. J. Breast Health 2020, 16, 22–31. [Google Scholar] [CrossRef]
  10. Wong, Y.M.; Jagmohan, P.; Goh, Y.G.; Putti, T.C.; Ow, S.G.W.; Thian, Y.L.; Pillay, P. Infiltrative pattern of metastatic invasive lobular breast carcinoma in the abdomen: A pictorial review. Insights Imaging 2021, 12, 181. [Google Scholar] [CrossRef] [PubMed]
  11. Batra, H.; Mouabbi, J.A.; Ding, Q.; Sahin, A.A.; Raso, M.G. Lobular Carcinoma of the Breast: A Comprehensive Review with Translational Insights. Cancers 2023, 15, 5491. [Google Scholar] [CrossRef]
  12. Iorfida, M.; Maiorano, E.; Orvieto, E.; Maisonneuve, P.; Bottiglieri, L.; Rotmensz, N.; Montagna, E.; Dellapasqua, S.; Veronesi, P.; Galimberti, V. Invasive lobular breast cancer: Subtypes and outcome. Breast Cancer Res. Treat. 2012, 133, 713–723. [Google Scholar] [CrossRef]
  13. Jung, S.P.; Lee, S.K.; Kim, S.; Choi, M.-Y.; Bae, S.Y.; Kim, J.; Kim, M.; Kil, W.H.; Cho, E.Y.; Choe, J.-H. Invasive pleomorphic lobular carcinoma of the breast: Clinicopathologic characteristics and prognosis compared with invasive ductal carcinoma. J. Breast Cancer 2012, 15, 313. [Google Scholar] [CrossRef]
  14. Marmor, S.; Hui, J.Y.C.; Huang, J.L.; Kizy, S.; Beckwith, H.; Blaes, A.H.; Rueth, N.M.; Tuttle, T.M. Relative effectiveness of adjuvant chemotherapy for invasive lobular compared with invasive ductal carcinoma of the breast. Cancer 2017, 123, 3015–3021. [Google Scholar] [CrossRef]
  15. Cristofanilli, M.; Gonzalez-Angulo, A.; Sneige, N.; Kau, S.-W.; Broglio, K.; Theriault, R.L.; Valero, V.; Buzdar, A.U.; Kuerer, H.; Buccholz, T.A. Invasive lobular carcinoma classic type: Response to primary chemotherapy and survival outcomes. J. Clin. Oncol. 2005, 23, 41–48. [Google Scholar] [CrossRef]
  16. Truin, W.; Voogd, A.C.; Vreugdenhil, G.; van der Heiden-van der Loo, M.; Siesling, S.; Roumen, R.M. Effect of adjuvant chemotherapy in postmenopausal patients with invasive ductal versus lobular breast cancer. Ann. Oncol. 2012, 23, 2859–2865. [Google Scholar] [CrossRef]
  17. Chamalidou, C.; Fohlin, H.; Albertsson, P.; Arnesson, L.-G.; Einbeigi, Z.; Holmberg, E.; Nordenskjöld, A.; Nordenskjöld, B.; Karlsson, P.; Linderholm, B. Survival patterns of invasive lobular and invasive ductal breast cancer in a large population-based cohort with two decades of follow up. Breast 2021, 59, 294–300. [Google Scholar] [CrossRef]
  18. Flores-Díaz, D.; Arce, C.; Flores-Luna, L.; Reynoso-Noveron, N.; Lara-Medina, F.; Matus, J.A.; Bargallo-Rocha, E.; Pérez, V.; Villarreal-Garza, C.; Cabrera-Galeana, P. Impact of invasive lobular carcinoma on long-term outcomes in Mexican breast cancer patients. Breast Cancer Res. Treat. 2019, 176, 243–249. [Google Scholar] [CrossRef]
  19. Fortunato, L.; Mascaro, A.; Poccia, I.; Andrich, R.; Amini, M.; Costarelli, L.; Cortese, G.; Farina, M.; Vitelli, C. Lobular breast cancer: Same survival and local control compared with ductal cancer, but should both be treated the same way? Analysis of an institutional database over a 10-year period. Ann. Surg. Oncol. 2012, 19, 1107–1114. [Google Scholar] [CrossRef]
  20. Korhonen, T.; Kuukasjärvi, T.; Huhtala, H.; Alarmo, E.-L.; Holli, K.; Kallioniemi, A.; Pylkkänen, L. The impact of lobular and ductal breast cancer histology on the metastatic behavior and long term survival of breast cancer patients. Breast 2013, 22, 1119–1124. [Google Scholar] [CrossRef]
  21. Molland, J.; Donnellan, M.; Janu, N.; Carmalt, H.; Kennedy, C.; Gillett, D. Infiltrating lobular carcinoma—A comparison of diagnosis, management and outcome with infiltrating duct carcinoma. Breast 2004, 13, 389–396. [Google Scholar] [CrossRef] [PubMed]
  22. Haque, W.; Arms, A.; Verma, V.; Hatch, S.; Butler, E.B.; Teh, B.S. Outcomes of pleomorphic lobular carcinoma versus invasive lobular carcinoma. Breast 2019, 43, 67–73. [Google Scholar] [CrossRef]
  23. Al-Baimani, K.; Bazzarelli, A.; Clemons, M.; Robertson, S.J.; Addison, C.; Arnaout, A. Invasive pleomorphic lobular carcinoma of the breast: Pathologic, clinical, and therapeutic considerations. Clin. Breast Cancer 2015, 15, 421–425. [Google Scholar] [CrossRef] [PubMed]
  24. Rakha, E.A.; Van Deurzen, C.H.; Paish, E.C.; Macmillan, R.D.; Ellis, I.O.; Lee, A.H. Pleomorphic lobular carcinoma of the breast: Is it a prognostically significant pathological subtype independent of histological grade? Mod. Pathol. 2013, 26, 496–501. [Google Scholar] [CrossRef]
  25. Van Baelen, K.; Geukens, T.; Maetens, M.; Tjan-Heijnen, V.; Lord, C.J.; Linn, S.; Bidard, F.-C.; Richard, F.; Yang, W.; Steele, R.E. Current and future diagnostic and treatment strategies for patients with invasive lobular breast cancer. Ann. Oncol. 2022, 33, 769–785. [Google Scholar] [CrossRef] [PubMed]
  26. Petrelli, F.; Barni, S. Response to neoadjuvant chemotherapy in ductal compared to lobular carcinoma of the breast: A meta-analysis of published trials including 1764 lobular breast cancer. Breast Cancer Res. Treat. 2013, 142, 227–235. [Google Scholar] [CrossRef] [PubMed]
  27. Mamtani, A.; Grabenstetter, A.; Sevilimedu, V.; Morrow, M.; Gemignani, M.L. Do non-classic invasive lobular carcinomas derive a benefit from neoadjuvant chemotherapy? Breast Cancer Res. Treat. 2023, 197, 417–423. [Google Scholar] [CrossRef] [PubMed]
  28. Rakha, E.A.; El-Sayed, M.E.; Menon, S.; Green, A.R.; Lee, A.H.; Ellis, I.O. Histologic grading is an independent prognostic factor in invasive lobular carcinoma of the breast. Breast Cancer Res. Treat. 2008, 111, 121–127. [Google Scholar] [CrossRef] [PubMed]
  29. Allison, K.H.; Hammond, M.E.H.; Dowsett, M.; McKernin, S.E.; Carey, L.A.; Fitzgibbons, P.L.; Hayes, D.F.; Lakhani, S.R.; Chavez-MacGregor, M.; Perlmutter, J.; et al. Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Guideline Update. J. Clin. Oncol. 2020, 38, 1346–1366. [Google Scholar] [CrossRef]
  30. Dodson, A.; Parry, S.; Ibrahim, M.; Bartlett, J.M.; Pinder, S.; Dowsett, M.; Miller, K. Breast cancer biomarkers in clinical testing: Analysis of a UK national external quality assessment scheme for immunocytochemistry and in situ hybridisation database containing results from 199,300 patients. J. Pathol. Clin. Res. 2018, 4, 262–273. [Google Scholar] [CrossRef]
  31. Wolff, A.C.; Somerfield, M.R.; Dowsett, M.; Hammond, M.E.H.; Hayes, D.F.; McShane, L.M.; Saphner, T.J.; Spears, P.A.; Allison, K.H. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: ASCO-College of American Pathologists Guideline Update. J. Clin. Oncol. 2023, 41, 3867–3872. [Google Scholar] [CrossRef] [PubMed]
  32. Aleskandarany, M.A.; Green, A.R.; Ashankyty, I.; Elmouna, A.; Diez-Rodriguez, M.; Nolan, C.C.; Ellis, I.O.; Rakha, E. Impact of intratumoural heterogeneity on the assessment of Ki67 expression in breast cancer. Breast Cancer Res. Treat. 2016, 158, 287–295. [Google Scholar] [CrossRef]
  33. Sparano, J.A.; Gray, R.J.; Makower, D.F.; Pritchard, K.I.; Albain, K.S.; Hayes, D.F.; Geyer, C.E., Jr.; Dees, E.C.; Goetz, M.P.; Olson, J.A., Jr. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N. Engl. J. Med. 2018, 379, 111–121. [Google Scholar] [CrossRef] [PubMed]
  34. Rakha, E.A.; Patel, A.; Powe, D.G.; Benhasouna, A.; Green, A.R.; Lambros, M.B.; Reis-Filho, J.S.; Ellis, I.O. Clinical and Biological Significance of E-cadherin Protein Expression in Invasive Lobular Carcinoma of the Breast. Am. J. Surg. Pathol. 2010, 34, 1472–1479. [Google Scholar] [CrossRef] [PubMed]
  35. The WHO Classification of Tumours Editorial Board: Breast Tumours; WHO: Geneva, Switzerland, 2019.
  36. Lokuhetty, D.; White, V.; Watanabe, R.; Cree, I. WHO Classification of Tumours Editorial Board. Breast tumors. In WHO Classification of Tumours Series, 5th ed.; International Agency for Research on Cancer: Lyon, France, 2019; Volume 2, pp. 88–97. [Google Scholar]
  37. Rakha, E.A.; Abbas, A.; Sheeran, R. Invasive lobular carcinoma mimicking papillary carcinoma: A report of three cases. Pathobiology 2016, 83, 221–227. [Google Scholar] [CrossRef] [PubMed]
  38. Lim, S.T.; Yu, J.H.; Park, H.K.; Moon, B.I.; Ko, B.K.; Suh, Y.J. A comparison of the clinical outcomes of patients with invasive lobular carcinoma and invasive ductal carcinoma of the breast according to molecular subtype in a Korean population. World J. Surg. Oncol. 2014, 12, 56. [Google Scholar] [CrossRef] [PubMed]
  39. Moran, M.S.; Yang, Q.; Haffty, B.G. The Yale University experience of early-stage invasive lobular carcinoma (ILC) and invasive ductal carcinoma (IDC) treated with breast conservation treatment (BCT): Analysis of clinical-pathologic features, long-term outcomes, and molecular expression of COX-2, Bcl-2, and p53 as a function of histology. Breast J. 2009, 15, 571–578. [Google Scholar] [PubMed]
  40. Magnoni, F.; Corso, G.; Maisonneuve, P.; Bianchi, B.; Accardo, G.; Sangalli, C.; Massari, G.; Rotili, A.; Nicosia, L.; Pesapane, F. Comparison of long-term outcome between clinically high risk lobular versus ductal breast cancer: A propensity score matched study. Eclinicalmedicine 2024, 71, 102552. [Google Scholar] [CrossRef]
  41. Delon, C.; Brown, K.F.; Payne, N.W.S.; Kotrotsios, Y.; Vernon, S.; Shelton, J. Differences in cancer incidence by broad ethnic group in England, 2013–2017. Br. J. Cancer 2022, 126, 1765–1773. [Google Scholar] [CrossRef]
  42. Buchanan, C.L.; Flynn, L.W.; Murray, M.P.; Darvishian, F.; Cranor, M.L.; Fey, J.V.; King, T.A.; Tan, L.K.; Sclafani, L.M. Is pleomorphic lobular carcinoma really a distinct clinical entity? J. Surg. Oncol. 2008, 98, 314–317. [Google Scholar] [CrossRef]
  43. Jacobs, M.; Fan, F.; Tawfik, O. Clinicopathologic and biomarker analysis of invasive pleomorphic lobular carcinoma as compared with invasive classic lobular carcinoma: An experience in our institution and review of the literature. Ann. Diagn. Pathol. 2012, 16, 185–189. [Google Scholar] [CrossRef] [PubMed]
  44. Christgen, M.; Cserni, G.; Floris, G.; Marchio, C.; Djerroudi, L.; Kreipe, H.; Derksen, P.W.; Vincent-Salomon, A. Lobular breast cancer: Histomorphology and different concepts of a special spectrum of tumors. Cancers 2021, 13, 3695. [Google Scholar] [CrossRef]
  45. Simpson, P.; Reis-Filho, J.; Lambros, M.; Jones, C.; Steele, D.; Mackay, A.; Iravani, M.; Fenwick, K.; Dexter, T.; Jones, A.; et al. Molecular profiling pleomorphic lobular carcinomas of the breast: Evidence for a common molecular genetic pathway with classic lobular carcinomas. J. Pathol. 2008, 215, 231–244. [Google Scholar] [CrossRef]
  46. Hood, C.I.; Font, R.L.; Zimmerman, L.E. Metastatic mammary carcinoma in the eyelid with histiocytoid appearance. Cancer 1973, 31, 793–800. [Google Scholar] [CrossRef] [PubMed]
  47. Merino, M.J.; Livolsi, V.A. Signet ring carcinoma of the female breast: A clinicopathologic analysis of 24 cases. Cancer 1981, 48, 1830–1837. [Google Scholar] [CrossRef] [PubMed]
  48. Aldulaijan, F.A.; Alsahwan, A.G.; Alsulaiman, M.H.A.; Mashhour, M.M.; Alwabari, A. Histiocytoid variant of invasive lobular breast carcinoma. A case report and literature review. Ann. Med. Surg. 2021, 72, 103091. [Google Scholar] [CrossRef]
  49. Coty-Fattal, Z.; Minhas, S.; Butcher, M.; Agarwal, I.; LaBoy, C.; Blanco, L.; Novo, J. Clinicopathologic and Immunophenotypic Classification of Invasive Lobular Carcinoma with Histiocytoid Features. Int. J. Surg. Pathol. 2023, 31, 10668969231189714. [Google Scholar] [CrossRef] [PubMed]
  50. Makower, D.; Qin, J.; Lin, J.; Xue, X.; Sparano, J.A. The 21-gene recurrence score in early non-ductal breast cancer: A National Cancer Database analysis. NPJ Breast Cancer 2022, 8, 4. [Google Scholar] [CrossRef]
  51. Tsung, K.; Grobmyer, S.R.; Tu, C.; Abraham, J.; Budd, G.T.; Valente, S.A. Neoadjuvant systemic therapy in invasive lobular breast cancer: Is it indicated? Am. J. Surg. 2018, 215, 509–512. [Google Scholar] [CrossRef]
  52. Haque, W.; Verma, V.; Hatch, S.; Suzanne Klimberg, V.; Brian Butler, E.; Teh, B.S. Response rates and pathologic complete response by breast cancer molecular subtype following neoadjuvant chemotherapy. Breast Cancer Res. Treat. 2018, 170, 559–567. [Google Scholar] [CrossRef]
  53. Lips, E.H.; Mukhtar, R.A.; Yau, C.; de Ronde, J.J.; Livasy, C.; Carey, L.A.; Loo, C.E.; Vrancken-Peeters, M.-J.T.F.D.; Sonke, G.S.; Berry, D.A.; et al. Lobular histology and response to neoadjuvant chemotherapy in invasive breast cancer. Breast Cancer Res. Treat. 2012, 136, 35–43. [Google Scholar] [CrossRef] [PubMed]
  54. Loibl, S.; Volz, C.; Mau, C.; Blohmer, J.-U.; Costa, S.D.; Eidtmann, H.; Fasching, P.A.; Gerber, B.; Hanusch, C.; Jackisch, C.; et al. Response and prognosis after neoadjuvant chemotherapy in 1051 patients with infiltrating lobular breast carcinoma. Breast Cancer Res. Treat. 2014, 144, 153–162. [Google Scholar] [CrossRef] [PubMed]
  55. Wilson, P.C.; Chagpar, A.B.; Cicek, A.F.; Bossuyt, V.; Buza, N.; Mougalian, S.; Killelea, B.K.; Patel, N.; Harigopal, M. Breast cancer histopathology is predictive of low-risk Oncotype Dx recurrence score. Breast J. 2018, 24, 976–980. [Google Scholar] [CrossRef] [PubMed]
  56. Kelly, C.M.; Krishnamurthy, S.; Bianchini, G.; Litton, J.K.; Gonzalez-Angulo, A.M.; Hortobagyi, G.N.; Pusztai, L. Utility of oncotype DX risk estimates in clinically intermediate risk hormone receptor-positive, HER2-normal, grade II, lymph node-negative breast cancers. Cancer 2010, 116, 5161–5167. [Google Scholar] [CrossRef]
  57. Weiser, R.; Polychronopoulou, E.; Hatch, S.S.; Haque, W.; Ghani, H.A.; He, J.; Kuo, Y.f.; Gradishar, W.J.; Klimberg, V.S. Adjuvant chemotherapy in patients with invasive lobular carcinoma and use of the 21-gene recurrence score: A National Cancer Database analysis. Cancer 2022, 128, 1738–1747. [Google Scholar] [CrossRef]
Figure 1. Examples of non-classic variants of invasive lobular carcinoma (ILC). (A) Pleomorphic ILC (scale bar: 60 µm); (B) Solid ILC (scale bar: 60 µm); (C) Tubulo-lobular carcinoma (scale bar: 100 µm); and (D) Alveolar ILC (scale bar: 200 µm).
Figure 1. Examples of non-classic variants of invasive lobular carcinoma (ILC). (A) Pleomorphic ILC (scale bar: 60 µm); (B) Solid ILC (scale bar: 60 µm); (C) Tubulo-lobular carcinoma (scale bar: 100 µm); and (D) Alveolar ILC (scale bar: 200 µm).
Cancers 16 01893 g001
Figure 2. Kaplan–Meier survival analysis shows initial favourable outcomes of the classic invasive lobular carcinoma (cILC) subtype compared to invasive ductal carcinoma of no special type (IDC-NST) that decreased over time, with the survival curves tending to cross at 15 years: (A) breast cancer-specific survival; (B) disease-free survival.
Figure 2. Kaplan–Meier survival analysis shows initial favourable outcomes of the classic invasive lobular carcinoma (cILC) subtype compared to invasive ductal carcinoma of no special type (IDC-NST) that decreased over time, with the survival curves tending to cross at 15 years: (A) breast cancer-specific survival; (B) disease-free survival.
Cancers 16 01893 g002
Figure 3. Kaplan–Meier survival analysis shows (A,B) survival outcomes of invasive lobular carcinoma (ILC) subtypes compared to invasive ductal carcinoma of no special type (IDC-NST). (C,D) Poor survival outcomes are associated with aggressive ILC compared to cILC.
Figure 3. Kaplan–Meier survival analysis shows (A,B) survival outcomes of invasive lobular carcinoma (ILC) subtypes compared to invasive ductal carcinoma of no special type (IDC-NST). (C,D) Poor survival outcomes are associated with aggressive ILC compared to cILC.
Cancers 16 01893 g003
Figure 4. Kaplan–Meier survival analysis shows shorter survival outcomes of aggressive invasive lobular carcinoma (ILC) subtypes compared to classic ILC (cILC) in endocrine-treated only (A,B) and combined endocrine and chemotherapy-treated (C) patients.
Figure 4. Kaplan–Meier survival analysis shows shorter survival outcomes of aggressive invasive lobular carcinoma (ILC) subtypes compared to classic ILC (cILC) in endocrine-treated only (A,B) and combined endocrine and chemotherapy-treated (C) patients.
Cancers 16 01893 g004
Figure 5. Kaplan–Meier survival curves show poor survival outcomes of aggressive invasive lobular carcinoma (ILC) compared to invasive ductal carcinoma of no special type (IDC-NST) in endocrine-treated only (A) and combined endocrine and chemotherapy-treated patients (B).
Figure 5. Kaplan–Meier survival curves show poor survival outcomes of aggressive invasive lobular carcinoma (ILC) compared to invasive ductal carcinoma of no special type (IDC-NST) in endocrine-treated only (A) and combined endocrine and chemotherapy-treated patients (B).
Cancers 16 01893 g005
Figure 6. Kaplan–Meier survival curves show favourable survival outcomes of classic invasive lobular carcinoma (cILC) compared to invasive ductal carcinoma of no special type (IDC-NST) in endocrine-treated patients: (A) breast cancer-specific survival; (B) disease-free survival.
Figure 6. Kaplan–Meier survival curves show favourable survival outcomes of classic invasive lobular carcinoma (cILC) compared to invasive ductal carcinoma of no special type (IDC-NST) in endocrine-treated patients: (A) breast cancer-specific survival; (B) disease-free survival.
Cancers 16 01893 g006
Table 1. Correlation of invasive lobular carcinoma compared to invasive ductal carcinoma of no special type to clinicopathological parameters.
Table 1. Correlation of invasive lobular carcinoma compared to invasive ductal carcinoma of no special type to clinicopathological parameters.
CharacteristicsPleomorphic Invasive Lobular Carcinoma
(pILC)
N (%)
Solid Invasive Lobular Carcinoma
(sILC)
N (%)
Classic Invasive Lobular Carcinoma
(cILC)
N (%)
Invasive Ductal Carcinoma, No Special Type
(IDC-NST)
N (%)
X2
(p-Value) a
pILC vs. cILC
X2
(p-Value) b
pILC vs. IDC-NST
X2
(p-Value) c
sILC vs. cILC
X2
(p-Value) d
sILC vs. IDC-NST
X2
(p-Value) e
pILC vs. sILC
X2
(p-Value) f
cILC vs. IDC-NST
Age at diagnosis (years)
<50
≥50

27 (22)
69 (78)

6 (14)
36 (86)

95 (16)
503 (84)

1366 (30)
3109 (70)

8.6
(0.003)

0.26
(0.6)

0.07
(0.7)

5.2
(0.02)

3
(0.07)

55.1
(<0.001)
Tumour size (cm)
<2
≥2

29 (30)
67 (70)

20 (48)
22 (52)

344 (57)
254 (43)

2672 (60)
1797 (40)

24.8
(<0.001)

34.0
(<0.001)

1.6
(0.2)

2.6
(0.1)

3.9
(0.04)

1.1
(0.2)
Tumour grade
1
2
3

0
52 (54)
44 (46)

0
33 (79)
9 (21)

55 (9)
543 (91)
0

320 (7)
1659 (37)
2496 (56)

295.9
(<0.001)

15.9
(<0.001)

38.8
(<0.001)

7.9
(0.005)

7.4
(0.007)

687.4
(<0.001)
Mitotic count
1
2
3

52 (54)
28 (29)
16 (17)

26 (62)
7 (17)
9 (21)

563 (94)
35 (6)
0

1612 (36)
957 (21)
1906 (43)

156.7
(<0.001)

26.2
(<0.001)

95.5
(<0.001)

12.5
(0.002)

2.3
(0.2)

734.8
(<0.001)
Nuclear pleomorphism
1
2
3

0
0
96 (100)

0
42 (100)
0

30 (5)
568 (95)
0

14 (1)
1066 (23)
3395 (76)


522.9
(<0.001)


29.8
(<0.001)


2
(0.1)


122.1
(<0.001)


138
(<0.001)


1412.1
(<0.001)
Tubule formation
1
2
3

0
0
96 (100)

0
0
42 (100)

6 (1)
40 (7)
552 (92)

88 (2)
1120 (25)
3267 (73)

7.2
(0.007)

32.6
(<0.001)

3.2
(0.07)

14.3
(<0.001)
-
106.3
(<0.001)
Nottingham Prognostic Index
Good Prognostic Group
Moderate Prognostic Group
Poor Prognostic Group


19 (20)

52 (54)
25 (26)


15 (36)

25 (59)
2 (5)


317 (53)

239 (40)
41 (7)


1288 (29)

2453 (55)
719 (16)


54.2
(<0.001)


8.4
(0.01)


2.4
(0.1)


4.2
(0.1)


9.9
(0.007)


149.5
(<0.001)
Axillary nodal status *
Negative
Positive


51 (53)
45 (47)


31 (74)
11 (26)


411 (69)
186 (31)


2886 (65)
1580 (35)


9.1
(0.002)


5.4
(0.02)


0.5
(0.5)


1.5
(0.2)


5.2
(0.02)


4.1
(0.04)
Lymph node stage
1 (Negative)
2 (1–3 positive)
3 (>3 positive)

51 (53)
29 (30)
16 (17)

31 (74)
9 (21)
2 (5)

411 (69)
131 (22)
55 (9)

2886 (65)
1206 (27)
374 (8)

10.0
(0.007)

9.8
(0.007)

0.8
(0.3)

1.7
(0.1)

6
(0.04)

6.9
(0.03)
Lymphovascular invasion
Negative
Positive


67 (70)
29 (30)


34 (81)
8 (19)


536 (90)
62 (10)


3241 (72)
1234 (28)


28.6
(<0.001)


0.33
(0.5)


2.6
(0.1)


1.6
(0.2)


1.9
(0.1)


82.1
(<0.001)
Distant metastasis site **
Common
Uncommon


34 (97)
1 (3)


8 (89)
1 (11)


96 (91)
10 (9)


947 (98)
18 (2)


1.9
(0.1)


0.16
(0.6)


0.02
(0.8)


1.9
(0.1)


0.9
(0.3)


14.1
(<0.001)
Oestrogen receptor
Negative
Positive

13 (13)
83 (87)

0
42 (100)

12 (2)
576 (98)

1297 (30)
3103 (70)

31
(<0.001)

11.6
(<0.001)

1.7
(0.1)

17.5
(<0.001)

6.3
(0.01)

201.7
(<0.001)
Progesterone receptor
Negative
Positive

34 (37)
59 (63)

8 (20)
31 (80)

135 (25)
413 (75)

1834 (44)
2318 (56)

5.8
(0.01)

2.1
(0.1)

0.3
(0.5)

8.8
(0.003)

3
(0.07)

75.9
(<0.001)
Human epidermal growth factor receptor 2
Negative
Positive


81 (91)
8 (9)


38 (97)
1 (3)


542 (99)
8 (1)


3359 (81)
766 (19)


17.8
(<0.001)


5.3
(0.02)


0.3
(0.6)


6.6
(0.1)


2.0
(0.1)


102.9
(<0.001)
Ki67 index
Low (≤14%)
High (>14%)

28 (54)
24 (46)

7 (29)
17 (71)

220 (79)
60 (21)

744 (35)
1394 (65)

14.2
(<0.001)

8.1
(0.005)

28.5
(<0.001)

0.3
(0.5)

4.0
(0.04)

197.9
(<0.001)
Oncotype Dx recurrence score
Low
Intermediate
High


2 (22)
6 (67)
1 (1)


4 (36)
6 (55)
1 (9)


5 (11)
37 (82)
3 (7)


44 (16)
160 (56)
80 (28)


0.2
(0.6)


1.2
(0.2)


2.0
(0.1)


4.0
(0.1)


0.5
(0.7)


12.0
(0.003)
Breast surgery
Breast-conserving
Mastectomy

35 (36)
61 (64)

23 (55)
19 (45)

279 (47)
319 (53)

2354 (53)
2121 (47)

3.5
(0.06)

9.8
(0.002)

1.0
(0.3)

0.08
(0.7)

4.0
(0.04)

7.5
(0.006)
Endocrine therapy
No
Yes

24 (25)
72 (75)

4 (10)
38 (90)

161 (27)
433 (73)

1727 (39)
2711 (61)

0.19
(0.6)

7.7
(0.006)

6.3
(0.01)

15.2
(<0.001)

4.3
(0.03)

31.7
(<0.001)
Chemotherapy
No
Yes

61 (64)
35 (36)

38 (90)
4 (10)

504 (84)
94 (16)

2849 (64)
1625 (36)

23.5
(<0.001)

0.001
(0.9)

1.2
(0.2)

13
(<0.001)

10.5
(0.001)

99.9
(<0.001)
* Routine IHC for LN examination is not routinely performed, which may lead to underestimated nodal metastasis. ** Common metastatic sites include lymph nodes, lung, liver, bone, and brain. Uncommon sites include gastrointestinal, ovarian, urinary tract, endometrial, and orbital metastases. a Comparison between pILC and cILC variants; b Comparison between pILC and IDC-NST; c Comparison between sILC and cILC; d Comparison between sILC and IDC-NST; e Comparison between pILC and sILC; f Comparison between cILC and IDC-NST. Significant p-values are in bold.
Table 2. Clinicopathological characteristics of the aggressive invasive lobular carcinoma subtype (n = 105).
Table 2. Clinicopathological characteristics of the aggressive invasive lobular carcinoma subtype (n = 105).
CharacteristicsFrequency (%)
Age at diagnosis (years)
<5029 (28)
≥5076 (72)
Tumour size (cm)
<2
≥2

33 (31)
72 (69)
Tumour grade
10
252 (50)
353 (50)
Mitotic count
152 (49)
228 (27)
325 (24)
Nuclear pleomorphism
10
29 (9)
396 (91)
Tubule formation
10
20
3100 (100)
Nottingham Prognostic Index
Good Prognostic Group19 (18)
Moderate Prognostic Group60 (57)
Poor Prognostic Group26 (25)
Lymph node stage
1 (Negative)57 (54)
2 (1–3 positive)31 (30)
3 (>3 positive)17 (16)
Lymphovascular invasion
Negative74 (70)
Positive31 (30)
Oestrogen receptor
Negative13 (12)
Positive92 (88)
Progesterone receptor
Negative34 (34)
Positive67 (66)
HER2
Negative89 (92)
Positive8 (8)
Ki67 index
Low (≤14%)28 (48)
High (>14%)30 (52)
Oncotype Dx recurrence score
Low2 (20)
Intermediate6 (60)
High2 (20)
Breast surgery
Breast-conserving39 (37)
Mastectomy66 (63)
Endocrine therapy
No24 (23)
Yes81 (77)
Chemotherapy
No69 (66)
Yes36 (34)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Makhlouf, S.; Atallah, N.M.; Polotto, S.; Lee, A.H.S.; Green, A.R.; Rakha, E.A. Deciphering the Clinical Behaviour of Invasive Lobular Carcinoma of the Breast Defines an Aggressive Subtype. Cancers 2024, 16, 1893. https://doi.org/10.3390/cancers16101893

AMA Style

Makhlouf S, Atallah NM, Polotto S, Lee AHS, Green AR, Rakha EA. Deciphering the Clinical Behaviour of Invasive Lobular Carcinoma of the Breast Defines an Aggressive Subtype. Cancers. 2024; 16(10):1893. https://doi.org/10.3390/cancers16101893

Chicago/Turabian Style

Makhlouf, Shorouk, Nehal M. Atallah, Susanna Polotto, Andrew H. S. Lee, Andrew R. Green, and Emad A. Rakha. 2024. "Deciphering the Clinical Behaviour of Invasive Lobular Carcinoma of the Breast Defines an Aggressive Subtype" Cancers 16, no. 10: 1893. https://doi.org/10.3390/cancers16101893

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop