Abstract
Invasive lobular carcinoma (ILC) accounts for 10% to 15% of breast cancers in the United States, 80% of which are estrogen receptor (ER)‐positive, with an unusual metastatic pattern of spread to sites such as the serosa, meninges, and ovaries, among others. Lobular cancer presents significant challenges in detection and clinical management given its multifocality and multicentricity at presentation. Despite the unique features of ILC, it is often lumped with hormone receptor‐positive invasive ductal cancers (IDC); consequently, ILC screening, treatment, and follow‐up strategies are largely based on data from IDC. Despite both being treated as ER‐positive breast cancer, querying the Cancer Genome Atlas database shows distinctive molecular aberrations in ILC compared with IDC, such as E‐cadherin loss (66% vs. 3%), FOXA1 mutations (7% vs. 2%), and GATA3 mutations (5% vs. 20%). Moreover, compared with patients with IDC, patients with ILC are less likely to undergo breast‐conserving surgery, with lower rates of complete response following therapy as these tumors are less chemosensitive. Taken together, this suggests that ILC is biologically distinct, which may influence tumorigenesis and therapeutic strategies. Long‐term survival and clinical outcomes in patients with ILC are worse than in stage‐ and grade‐matched patients with IDC; therefore, nuanced criteria are needed to better define treatment goals and protocols tailored to ILC's unique biology. This comprehensive review highlights the histologic and clinicopathologic features that distinguish ILC from IDC, with an in‐depth discussion of ILC's molecular alterations and biomarkers, clinical trials and treatment strategies, and future targets for therapy.
Implications for Practice
The majority of invasive lobular breast cancers (ILCs) are hormone receptor (HR)‐positive and low grade. Clinically, ILC is treated similar to HR‐positive invasive ductal cancer (IDC). However, ILC differs distinctly from IDC in its clinicopathologic characteristics and molecular alterations. ILC also differs in response to systemic therapy, with studies showing ILC as less sensitive to chemotherapy. Patients with ILC have worse clinical outcomes with late recurrences. Despite these differences, clinical trials treat HR‐positive breast cancers as a single disease, and there is an unmet need for studies addressing the unique challenges faced by patients diagnosed with ILC.
Keywords: Lobular cancer, E‐cadherin, Dormancy, Late recurrence, ERBB2
Short abstract
Invasive lobular carcinoma is the second most common subtype of invasive breast cancer and presents unique challenges in detection and treatment. This review reports on the distinct morphological, molecular, and clinical features of invasive lobular carcinoma and highlights the challenges in the management of this cancer.
Introduction
Breast cancer is the most commonly diagnosed cancer among women (excluding skin cancer), with 276,480 new diagnoses in 2020 expected in the United States alone [1]. One in eight women will be diagnosed with breast cancer in their lifetime [2]. Invasive lobular carcinoma (ILC) is the second most common subtype, accounting for 10% to 15% of invasive breast cancer [3]. ILC presents unique challenges in detection and treatment, and yet it remains understudied in research. In this review, we will discuss the distinct morphological, molecular, and clinical features of ILC and highlight the challenges in the management of this cancer.
Materials and Methods
A comprehensive literature search of the PubMed database was conducted. The keywords used were “invasive lobular carcinoma,” “E‐cadherin,” “endocrine therapy resistance,” “HER2,” “FOXA1,” “FGFR,” “WNT,” and “p120 catenin.” All articles, including clinical trials, reviews, and original manuscripts, were considered. The search term “invasive lobular carcinoma” yielded 4,496 search results; thus, the search criteria was modified as follows to narrow down results: “invasive lobular carcinoma AND E‐cadherin” produces 335 results; “invasive lobular carcinoma AND p120 catenin” yields 24 results. Selected articles from the database search are included in this article. For clinical trials, further information was obtained through ClinicalTrials.gov.
Incidence, Mortality, and Clinical Features
The incidence of ILC has risen in recent years. Studies based on Surveillance, Epidemiology, and End Result (SEER) [4, 5] database found ILC rates increased from 9.6% in 1987 to 15.6% in 1999, with invasive ductal carcinoma (IDC) rates staying relatively constant. This increase in risk was associated with use of combined hormone replacement therapy (HRT) of estrogen and progestin, or progestin alone. Following the dramatic reduction in use of HRT, there was a nonsignificant downward trend in incidence of ILC for the period of 2002 to 2006 [6].
The incidence of ILC is highest in White women; however, Black women experience higher mortality from ILC. A 2017 study examined 18,295 patients with ILC from the SEER database, which included White women (n = 15,936), Black women (n = 1,451), and women of other races (American Indian/Alaskan Native and Asian/Pacific Islander, n = 908) found that the 5‐year overall survival (OS) and 5‐year breast cancer–specific survival were worst in Black patients with ILC (76.0% and 84.4%, respectively) compared with White women and women of other races (85.5% and 87.7%, p < .01; 91.1%, and 91.6%, p < .01, respectively) [7]. Other studies have reported similar 5‐year survival rates, approximately 86% for patients with ILC when not differentiating by race [8].
ILC is distinct from other breast cancer subtypes in its clinical, radiographic, and histopathology features (Table 1). Patients with ILC are generally older than other histological subtypes [8, 9, 10]. ILC has higher rates of multicentricity as well [11, 12]. The tumors can be large and are frequently bilateral, with lower rates of breast preservation [7]. ILC does not usually form a discrete palpable mass and is more difficult to detect through physical examination or imaging until at advanced stages [10, 11]. Radiographically, ILC may present as a spiculated mass or architectural distortion; however, mammography has lower sensitivity in the detection of ILC compared with IDC, resulting in potential false negatives reported to occur in 29.9% of ILCs [13]; ultrasound and MRI may be more helpful but could result in overestimation of tumor size or false positives [14, 15]. The classic subtype of ILC is composed of discohesive, small, round to ovoid cells with minimal cytoplasm and occasional cytoplasmic inclusions. Typically, these cells infiltrate the stroma as single cells or in single‐file lines, with minimal host reaction. Loss of membranous E‐cadherin expression as detected by immunohistochemistry (IHC) is seen in >90% of cases [3, 8], with concomitant loss of membranous p120 (increased cytoplasmic expression) and beta‐catenin due to disruption of the E‐cadherin complex [16]. The morphology and immunophenotype of ILC is due to inactivation of the cell–cell adhesion molecule E‐cadherin [8]. It results in the inability of tumor cells to adhere to one another [3] and leads to its unique histomorphology and mammographic presentation [10, 11]. Classic ILC is almost exclusively estrogen receptor (ER) and progesterone receptor (PR) positive and HER2 negative [8]; however, the pleomorphic variant shows higher rates of HER2 positivity [17]. In comparison with IDC, ILC is more likely to have metastases in the luminal gastrointestinal tract, cerebrospinal fluid and leptomeninges, and ovary [18, 19, 20].
Table 1.
Clinical/pathological differences | IDC | ILC |
---|---|---|
Pathology | ||
Description of growth |
|
|
Clinical features |
|
|
Detection |
|
|
Comments |
|
Abbreviations: DCIS, ductal carcinoma in situ; GI, gastrointestinal; IDC, invasive ductal cancer; ILC, invasive lobular cancer.
Histological Subtypes
Classic type ILC rarely induces a host response, does not typically destroy the architecture of breast, and grows as single‐file concentric patterns around ducts and lobular units of the breast. Patients diagnosed with classic type ILC often have associated lobular carcinoma in situ preceding diagnosis and have better overall prognosis than other ILC variant diagnoses [21, 22, 23]. However, long‐term survival is lower compared with stage‐ and grade‐matched IDC [24]. Pleomorphic lobular cancer (PLC) follows the typical ILC growth pattern but with marked nuclear pleomorphism (larger cell size with more abundant cytoplasm, more prominent nucleolus [Fig. 1], and higher mitotic rate compared with classic type ILC) [25]. Genetically, PLC shows similar genomic alterations to classic type ILC (gains in 1q and 16p, losses of 11q and 16q) along with additional genomic alterations characteristic of high‐grade IDC. PLC shows lower expression of hormone receptors (HR) and high rates of HER2 expression (9.7%–37%). Studies show PLC as a more aggressive variant with higher stage and lymph node involvement and shorter survival compared with classic type ILC [25, 26, 27, 28]. Solid variant ILC consists of discohesive lobular cells that grow in sheets throughout the breast rather than single‐file patterns, with higher grade nuclear atypia and mitotic rate compared with classic ILC [3, 21, 23]. Alveolar ILC is characterized by cells arranged in round aggregates composed of at least 20 classic type ILC cells [21, 23]. Signet ring cell variant of ILC is architecturally similar to classic ILC but contains more than 20% with signet cell morphology [22, 29]. Cells are differentiated from other variants by significant intracellular mucin displacing the nucleus within the cell. Other variants include tubulolobular carcinoma and histiocytoid variant of ILC, the latter associated with shorter survival [22, 30].
Key Molecular Alterations in ILC
Loss of E‐cadherin is the Hallmark Mutation in ILC
One of the characteristic features of ILC is the loss of E‐cadherin. The E‐cadherin gene, CDH1, is located on human chromosome 16q22.1 and codes for a 120kDa single transmembrane glycoprotein [31]. Binding of calcium to its extracellular domain regulates activity of its transmembrane domain, whereas binding of alpha, beta, gamma, and p120 catenins to its cytoplasmic tail links it to the actin cytoskeleton and sequesters catenin from participating in downstream signaling [23, 32]. The key downstream signaling pathways include Hippo, WNT, TGFβ, NFκB, and growth signaling pathways [33, 34, 35]. E‐cadherin supports cell–cell adhesion and regulates motility, and its loss leads to epithelial‐to‐mesenchymal transition. Homophilic binding of cadherins leads to contact inhibition of cell proliferation by activating growth inhibitory signals through the Hippo pathway [36]. Loss of E‐cadherin in ILC is predominantly attributed to CDH1 mutation, which in 89% of cases occurred concurrently with heterozygous deletion in chromosome 16q [37]. Analysis of invasive breast carcinoma cases in the Cancer Genome Atlas (TCGA) dataset (http://www.cbioportal.org) [38, 39, 40] revealed that 66% (107/162) of ILCs harbor mutation in CDH1 compared with only 3% (22/741) of IDCs (Fig. 2A). Over 80 different mutations have been identified throughout the coding region of CDH1, 83% of which were predicted to result in truncation of the protein [37] resulting from shallow deletion (Fig. 2B). CDH1 mutations have also been detected in matching lobular carcinoma in situ, demonstrating that this is an early event [3, 37, 41]. Although some studies have reported downregulation of CDH1 transcription due to promoter methylation [3], based on TCGA data set and later studies by Ciriello et al., methylation‐mediated silencing of Cdh1 in ILC remains controversial [37, 40].
Loss of E‐cadherin markedly increases cytoplasmic accumulation of p120 catenin, which plays a key role in tumor invasion [42]. In normal breast and in situ carcinomas, p120 catenin is primarily bound to E‐cadherin at a juxtamembrane site, where it recruits microtubules and stabilizes E‐cadherin by suppressing endocytosis. p120 also regulates the activity of Rho family GTPases, the molecular switch that is key to cell migration [43]. Using a mouse model of ILC, Schackmann et al. showed that in cytosol, p120 catenin promotes tumor growth by inducing anoikis resistance [44]. Cytosolic p120 interacts with Rho/Rock antagonist myosin phosphatase Rho‐interacting protein and relieves suppression of Rho‐Rock signaling pathway and activated RhoA and its downstream effector Rock1, leading to anoikis resistance. Consistently, human ILC samples showed hallmarks of active Rock signaling [44]. Increase in nuclear p120 was noted by van de Ven et al. in mouse ILC cells and in metastatic cells present in pleural effusion of patients with ILC [45]. Nuclear p120 accumulation relieved transcriptional repression by Kaiso on its well‐known target Wnt11, a non‐canonical Wnt signaling activator. Wnt11 in turn activates RhoA‐Rock signaling and induces anoikis resistance, linking it to E‐cadherin loss. As E‐cadherin loss and cytoplasmic localization of p120 catenin is unique to ILC, this can be a useful diagnostic tool to differentiate between ILC and IDC [16, 40, 46].
ERBB2 Mutation in ILC
ILC is characterized as a Luminal A subtype because of high ER/PR expression, low HER2 amplification, and low‐grade tumor. However, recent studies revealed that HER2 mutations are more common in ILC (2%–15%), particularly in higher grade tumors, compared with other subtypes of breast cancer [9, 17, 40, 47, 48, 49]. Deniziaut et al. reported that 6 out of 39 grade 3 HER2‐negative ILC tumors harbor at least one HER2‐activating mutation, an approximately 15% incidence rate [50]. In addition, primary ILC tumors carrying an HER2 mutation were shown to have early relapse [47]. Based on multiple studies showing higher rates of HER2 mutations in IHC‐scored HER2‐negative ILC tumors, personalizing therapy by targeting this pathway is an important consideration. Targeting HER2 mutations using tyrosine kinase inhibitors along with trastuzumab is an option [51]. Clinical studies are needed to understand when and how to target the HER2 pathway in ILC to improve clinical outcomes.
Endocrine Resistance and Fox A1 Amplification in ILC
Patients with ILC are routinely treated with endocrine therapy, but one third of patients fail to respond to treatment and develop endocrine resistance in the long term. The underlying mechanism behind acquired endocrine resistance in ILC is multifactorial. Comparative gene expression analysis of tamoxifen‐resistant ILC cell line SUM44/LCCTam revealed reduced expression of ERα and increase expression of estrogen‐related receptor γ (ERRγ) when compared with parental cell lines [52]. Orphan nuclear receptor ERRγ was shown to activate AP1‐dependent transcription in the resistant cells. Activation of AP1‐mediated transcription conferring tamoxifen resistance has been demonstrated previously in HR‐positive MCF7 and BT474 IDC cell line [53, 54]. Stires et al. reported upregulation of mitogen‐activated protein kinase (MAPK)/extracellular signal‐regulated kinase (ERK) pathway and glutamate receptor in Sum44/LCCTam cells and their inhibition‐restored tamoxifen sensitivity [55]. Using long‐term estrogen‐deprived (LTED) cell line, Du et al. demonstrated activation of lipid metabolism in MDA‐MB‐134‐VI‐LTED cells, specifically, which increased expression of sterol regulatory binding element protein‐1 (SREBP1) and its downstream target fatty acid synthase [56]. Subsequent analysis of clinical specimens showed a significant association between high SREBP‐1 expression and lack of response to the aromatase inhibitors letrozole, suggesting a role of SREBP‐1 in acquired endocrine resistance. Additional work in LTED cell lines by Sikora et al. identifies WNT4, a ligand in the WNT signaling pathway, to be a driver for estrogen‐induced growth and a potential mediator of endocrine resitance in ILC [57]. Further research into the interplay betwen E‐cadherin loss and the WNT signaling pathway might uncover novel targets to overcome endocrine resistance in ILC.
FOXA1, an ER transcription modulator, plays a key role in cancer progression and development of endocrine resistance [58]. FOXA1 is considered a pioneer factor for its ability to associate with condensed chromatin and demethylate and demarcate the site for tissue‐specific binding of transcription factors, including ER, PR, and androgen receptor [58, 59]. FOXA1 is co‐expressed with ER at high levels in endocrine‐resistant metastatic breast cancer [60]. Through comprehensive genomic analysis, Ciriello et al. observed spatially clustered mutations of FOXA1 in 7% of ILC samples (n = 127), leading to increased FOXA1 expression and activity [37]. A higher rate of FOXA1 mutations was observed in ILC compared with IDC (7% vs. 2%). In contrast, the rate of GATA3 mutation, another key regulator of ER activity, was consistently lower in ILC versus IDC (5% vs. 20%) [37], suggesting that ILC and IDC may rely on different mechanisms to regulate ER‐mediated transcription. Stires et al. showed amplification of FOXA1 in tamoxifen‐resistant SUM44PE/LCCTam cell lines, which supports the notion that FOXA1 could be critical for endocrine resistance in ILC [55]. Further studies are warranted to fully understand the implication of FOXA1 mutations in endocrine‐resistant ILC.
PI3K/AKT Signaling in ILC
Alterations in phosphoinositide 3‐kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling have been observed in numerous cancer types, including breast cancer [61, 62]. Frequent mutations in phosphatase and tensin homolog (PTEN) and PIK3CA, and low frequency of mutations in PIK3R1, AKT1, AKT2, and mTOR, have been reported in ILC [37, 63, 64]. Interestingly, Ciriello et al. noted mutually exclusive nature of PIK3CA mutation (48%) and PTEN mutation (13%) in ILC [37].
Activation of PI3K signaling was confirmed in both mouse and human ILC models by studying phosphorylation status of different intermediate proteins in this pathway. Whereas phosphorylation of AKT (at T473, T308), S6K1 (at T389), and 4EBP1 (at S65) was more prevalent in human ILCs (n = 66), mouse ILCs (n = 30) showed phosphorylation of AKT (at S473), S6 (at S235/S236), and 4EBP1 (at T37/46) [64]. Treatment of K14‐Cre;Cdh1 F/F ;Trp53 F/F mice, a model of metastatic ILC in either neoadjuvant or prolonged adjuvant setting with a mTOR inhibitor (AZD8055), showed response in primary tumors and delayed metastases. Interestingly, the therapeutic response was suggested to be in part mediated by the adaptive immune system [64]. Ongoing clinical trials with the mTOR inhibitor everolimus in early‐stage HR‐positive breast cancer may shed some light on the outcomes in patients with ILC (NCT01674140).
Fibroblast Growth Factor Signaling in ILC
The fibroblast growth factor (FGF) signaling pathway plays an essential role in cancer cell progression, angiogenesis, and survival. In approximately 7.1% of all cancers and approximately 18% of breast cancers, FGF receptor alterations have been reported [10]. Approximately 14% of patients with breast cancer have amplification of chromosome 8p11‐12, harboring FGFR1 [10, 11, 12]. Reis‐Filho et al. identified FGFR1 as the driver amplicon for 8p12‐p11.2 region in patients with ILC and demonstrated that inhibiting FGFR1 in MDA‐MB‐134 cells inhibited cell growth [14]. Overexpression of FGFR1 in breast cancer cell lines conferred tamoxifen resistance, whereas its silencing increased sensitivity to tamoxifen [15]. FGFR1 is amplified in human ILC cell lines MDA‐MB‐134 and SUM44PE, in which combined use of antiestrogen and FGFR inhibitor led to maximum cell death compared with monotherapy [24]. Using RNA‐sequencing and microarray analysis, Levine et al. identified FGFR4 overexpression in both ILC LTED and tamoxifen‐resistant cell lines. FGFR4 expression was also found to be higher in distant metastasis of patients treated with endocrine therapy [65]. In a mouse model of insertional mutagenesis, Kas et al. identified Fgfr2 as an oncogenic driver for ILC in conditional Cdh1 knockout mice, and the tumors were sensitive to FGFR inhibitor AZD4547 [25]. Eventual development of resistance to AZD4547 in these mice was attributed to activation of the canonical MAPK‐ERK pathway [26]. In other studies, FGFR mutation and amplification have been associated with disease recurrence [28, 49]. It is apparent that aberrations in the FGFR family proteins play a role in poor outcomes of patients with ILC, and further investigation into FGFR‐targeted therapy is warranted. Ongoing clinical trials with the FGFR2 inhibitor pemigatinib in advanced solid tumor malignancies harboring activating FGFR mutations and translocations may shed light on the outcomes in patients with ILC (NCT03822117).
Thus, ILC is not only a unique histological subtype with specific clinical features but also exhibits complex molecular alterations that could be potentially targeted for improved clinical outcomes (Fig. 3).
Dormant Disseminated Cancer Cells in Late Recurrence of ILC
The overall survival for patients with ILC is favorable compared with IDC within the first 5 years of diagnosis, but the outcome is worse after 5 years [66]. One reason for this is the presence of disseminated cancer cells (DCC) that remain dormant for prolonged periods and can result in delayed relapse at distant metastatic sites. Cancer cells disseminated from the primary site prior to surgery exist as minimal residual disease for a long period of time before switching to overt metastasis and aggressive growth [67]. ER‐negative breast cancers have higher rates of relapse within 5 years of primary tumor diagnosis, whereas ER‐positive breast cancers relapse at greater rates between 5 and 20 years [68]. In a comparative genomic study between ILC and IDC, Du et al. described lower rates of protein translation and metabolism in ILC compared with IDC, which are known features of tumor dormancy [69]. Additionally, Narbe et al. compared the number of circulation tumor cells (CTCs) in patients with ILC and IDC and showed that although patients with ILC had higher number of CTCs, the prognostic significance for CTCs ≥5 was weaker, suggesting a more dormant nature of these disseminated tumor cells [70]. Although in‐depth research on the specific role of the DCCs and the metastatic niche in ILC is lacking, they are believed to play a major role in the late recurrence of ILC, leading to worse patient outcomes. It is argued that targeting the dormant niche could be an effective way of delaying or preventing metastasis [71]. Understanding the dormant niche and specific molecular switch in ILC that awakens the dormant cells to proliferate into macrometastatic disease is crucial to developing novel therapeutics to treat ILC.
Genetically Engineered Mouse Models for ILC
Animal models of ILC are limited and a critical area of need. Conditional deletion of Cdh1 using either MMTV‐, K14‐, or WAP‐Cre failed to form mammary tumors [72, 73, 74]. However, dual knockout of Cdh1 and p53 in mouse mammary epithelium using either K14‐ or WAP‐Cre leads to the formation of mammary tumors resembling human pleomorphic ILC [72, 75]. The WAP‐Cre;Cdh1 F/F ;Trp53 F/F mouse is a better model, as K14‐Cre;Cdh1 F/F ;Trp53 F/F mice develop skin cancers as well. The WAP‐Cre;Cdh1 F/F ;Trp53 F/F mice develop invasive and metastatic ILC similar to human ILC, with metastatic spread to the bone, gastrointestinal tract, lymph nodes, and lung [75]. WAP‐Cre;Cdh1 F/F ;Pten F/F mice generated by Boelens et al. develop tumors that closely resemble the histological and molecular features, ER status, growth kinetics, metastatic behavior, and tumor microenvironment of classical human ILC [76]. To match the immune‐related (IR) subtype of ILC, An et al. developed the Wap‐Cre;CDH1 loxP/loxP;R26‐LSL‐Pik3ca H107R, a model that displays morhological features, immune cell infiltration, and T‐regulatory cell signaling observed in human IR‐ILC [77]. In mice with mammary gland–specific loss of E‐cadherin and expression of Cas9, the lentiviral delivery of PTEN targeted single‐guide RNA resulted in efficient induction of ILC [78]. The WAP‐Cre;Cdh1 F/F ;Cas9 mice along with guide RNA targeting specific tumor suppressors provides an opportunity to test combinations of tumor suppressor genes implicated in ILC. Although many ILC genetically engineered mouse (GEM) models have been developed over the past decade, an established model with a high rate of ER postivity that closely mirrors the unique histology and metastatic potential of human ILC is critical to study hormone therapy resistance and to validate novel drug targets.
Clinical Management of Invasive Lobular Cancer
Clinical management of invasive lobular breast cancer depends on the biology of the tumor and the stage at diagnosis.
Role of Surgery
A significant number of ILC patients undergo mastectomy. Breast‐conserving surgery (BCS) in combination with postoperative radiation is considered to be an alternative to mastectomy in women with early‐stage breast cancer. Foder et al. showed that BCS with radiation or mastectomy results in similar distant metastasis–free survival, breast cancer–specific survival, and locoregional recurrence–free survival [79]. Rates of BCS in early‐stage ILC have increased by 11.6% from 1993 to 2003 [80], with accumulating evidence suggesting no significant long‐term survival benefit for patients undergoing mastectomy [81, 82]. Approximately 17% to 65% of patients with ILC require a second intervention, and re‐excision lumpectomy is successful in achieving clear margins, particularly in those with node‐negative disease [83, 84]. The unique pathology of ILC leads to under‐recognition of the extent of the disease in the breast on routine preoperative imaging and results in a more extensive surgery in order to obtain clear margins. This is one of the reasons for the higher rates of mastectomies seen with this type of breast cancer.
Role of Neoadjuvant Therapy
Patients with clinical stage II or stage III disease are eligible to have neoadjuvant treatment, which is given to improve rates of BCS [85, 86]. However, ILC is considered to be less chemosensitive compared with IDC, with lower rates of pathologic complete response (pCR) and BCS compared to IDC. In a retrospective study by Tubiana‐Hulin et al., 860 patients received neoadjuvant chemotherapy, of whom 14% (118) had ILC [87]. About 30% of patients with ILC compared with 48% of patients with IDC underwent BCS; pCR was observed in 1% of patients with ILC versus 9% of patients with IDC (p = .002). The poor response to chemotherapy is thought to reflect the biology of ILC, being strongly ER/PR positive with a low mitotic index. Therefore, neoadjuvant chemotherapy should only be offered to patients with ILC after due consideration and a multidisciplinary discussion.
As ILC is often strongly ER/PR positive, treatment with endocrine therapy in the neoadjuvant setting could be considered. A retrospective study by Dixon et al. consisting of 61 patients with ER‐positive ILC found that neoadjuvant letrozole reduced tumor size by a mean of 66% at 3 months, with an 81% rate of successful breast conservation [88]. This further supports the concept that patients with ILC could receive neoadjuvant endocrine therapy instead of neoadjuvant chemotherapy if they desired breast conservation. The ALTernate approaches for clinical stage II or III Estrogen Receptor positive breast cancer NeoAdjuvant TrEatment (ALTERNATE trial, NCT01953588) [89] is an ongoing randomized phase III trial to determine whether neoadjuvant fulvestrant or the combination of anastrozole plus fulvestrant is better than anastrozole monotherapy for tumor shrinkage in postmenopausal patients with stage II to III invasive lobular and ductal breast cancer.
Role of Adjuvant Therapy
Systemic therapy given after definitive surgery impacts disease‐free survival (DFS) and OS in breast cancer [90]. The decision to proceed with adjuvant chemotherapy is often based on the size of the primary tumor, lymph node status, and results of genomic testing (i.e., Oncotype DX; Prosigna‐PAM50) if indicated. Oncotype DX recurrence scores for patients with ILC usually tend to be in the low or intermediate range, making the benefit of adjuvant chemotherapy small [91, 92, 93]. The application of the Oncotype DX prognostic tool in ILC needs further investigation, as very few patients with ILC were included in the trials used in the development of this tool. A retrospective analysis of patients within the Danish Breast Cancer Group database suggests that postmenopausal women with ILC and an intermediate Prosigna‐PAM50 score have 18% 10‐year risk of recurrence compared with 8% in those with low Prosigna‐PAM50 score [94]. Thus, women with intermediate/high genomic scores may have a potentially meaningful clinical benefit from chemotherapy compared with women with low scores. The effect of adjuvant chemotherapy in ILC was retrospectively studied by Truin et al. [95]. Patients were divided into two groups: one group receiving adjuvant hormonal therapy alone, and the other receiving adjuvant hormonal therapy plus adjuvant chemotherapy. The 10‐year OS rate for IDC when treated with hormonal therapy alone and combination therapy was 69% versus 74% (p < .0001); for ILC, 10‐year OS rate was 68% versus 66% (p = .45). This retrospective study suggests that adjuvant chemotherapy offered no additional benefit for patients with ILC receiving endocrine therapy, in contrast to patients with IDC.
In early‐stage HR‐positive breast cancer, 5 years of adjuvant endocrine therapy substantially reduces the risks of locoregional and distant recurrence and death from breast cancer [96, 97]. Tamoxifen is typically indicated for premenopausal women, whereas a third‐generation aromatase inhibitor is given to postmenopausal women. Investigators from the Breast International Group 1‐98 study explored the relative effectiveness of letrozole versus tamoxifen in patients with ILC compared with IDC. After a median follow‐up of 8.1 years, letrozole was associated with a significantly reduced risk of DFS event in patients with both luminal B‐like (HR = 0.34, 95% confidence interval [CI] = 0.21–0.55) and luminal A‐like ILC subtypes (HR = 0.50, 95% CI = 0.32–0.78), concluding that the magnitude of benefit from letrozole was greater in patients with ILC versus IDC [98]. CAN‐NCIC‐MA27 (NCT00066573) was a randomized phase III trial comparing anastrozole versus exemestane. There was no difference in event‐free survival between ILC and IDC; however, in the ILC group, anastrozole showed a marginal benefit (HR = 1.79, 95% CI = 0.98–3.27, p = .55), which may not have any clinical significance [99]. A current presurgery window‐of‐opportunity study (NCT02206984) is investigating which antiestrogen therapy (fulvestrant vs. anastrozole vs. tamoxifen) is most effective in ILC. Hence, current data support the important role of adjuvant/neoadjuvant endocrine therapy in HR‐positive ILC, with some data supporting the use of aromatase inhibitors over tamoxifen.
Conclusion
Given the rather uniform characteristics of ILC tumors with high HR positivity and low proliferation index, the challenge is how to identify tumors that would have a worse prognosis. One molecular signature, LobSig, that captures the unique molecular landscape of ILC along with clinicopathological features appears to provide independent prognostic information for patients with ILC, but it needs validation prospectively [100]. Identifying patients with high risk of recurrence will help in exploring novel targeted therapies and potentially immunotherapy. The ROLO study (NCT03620643), a phase II trial, is investigating the role of ROS1 inhibition with crizotinib in advanced E‐cadherin–negative, ER‐positive lobular breast cancer and diffuse gastric cancer. E‐cadherin defective cells rely on ROS1 for proper function, and ROS1 inhibition demonstrates a synthetic lethality in in vitro and in vivo studies [101].
CDK4/6 inhibitors are currently approved in combination with endocrine therapy as first‐ and second‐line treatment in advanced ER‐positive, HER2‐negative breast cancer. Multiple ongoing clinical trials are investigating the role of CDK4/6 inhibitors in neoadjuvant and adjuvant settings. PALLET (FB‐11; NCT02296801) demonstrated that the addition of palbociclib to letrozole in the neoadjuvant setting significantly improves the suppression of tumor proliferation in HR‐positive early‐stage breast cancer [102]. Palbociclib and Endocrine therapy for LObular breast cancer Preoperative Study (PELOPS, NCT02764541) is a phase II clinical trial assessing clinical utility of neoadjuvant palbociclib in women with HR‐positive, HER2‐invasive ILC. An immune‐related ILC subgroup was identified in the analysis of TCGA and RATHER consortia data sets [37, 103]. A phase II clinical trial is assessing the efficacy of carboplatin and atezolizumab in metastatic lobular breast cancer (the GELATO study, NCT03147040).
Invasive lobular carcinoma of the breast is a distinct entity with unique clinical and molecular features. Although studies show that the outcomes are largely similar to ductal cancers, the delay and difficulty in early diagnosis, inherent resistance to conventional therapies, and risk of late recurrences create significant challenges in management. With so few human ILC cell lines and established GEM models, there is a unique opportunity to develop novel models of ILC to advance ILC research. ILC‐specific diagnostic tools and treatment options are needed to improve overall patient outcomes. The lack of large national studies addressing the needs of the patients diagnosed with this histology is an unmet need.
Author Contributions
Conception/design: Bhuvaneswari Ramaswamy
Provision of study material or patients: Gary Tozbikian
Collection and/or assembly of data: Nikhil Pramod, Akanksha Nigam, Mustafa Basree, Resham Mawalkar, Saba Mehra, Gary Tozbikian, Nicole Williams, Sarmila Majumder, Bhuvaneswari Ramaswamy
Data analysis and interpretation: Nikhil Pramod, Akanksha Nigam, Sarmila Majumder, Bhuvaneswari Ramaswamy
Manuscript writing: Nikhil Pramod, Akanksha Nigam, Mustafa Basree, Resham Mawalkar, Saba Mehra, Neelam Shinde, Gary Tozbikian, Nicole Williams, Sarmila Majumder, Bhuvaneswari Ramaswamy
Final approval of manuscript: Nikhil Pramod, Akanksha Nigam, Mustafa Basree, Resham Mawalkar, Saba Mehra, Neelam Shinde, Gary Tozbikian, Nicole Williams, Sarmila Majumder, Bhuvaneswari Ramaswamy
Disclosures
Bhuvaneswari Ramaswamy: Eisai (C/A); Gary Tozbikian: Roche/Genentech (H), Lilly USA (C/A). The other authors indicated no financial relationships.
(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board
Acknowledgments
This study was funded by the Anderson Development fund (46040‐315876), the Ohio State University Comprehensive Cancer Center ‐ Arthur G. James Cancer Hospital, and the Richard J. Solove Research Institute Peotonia Fellowship.
No part of this article may be reproduced, stored, or transmitted in any form or for any means without the prior permission in writing from the copyright holder. For information on purchasing reprints contact commercialreprints@wiley.com.
Disclosures of potential conflicts of interest may be found at the end of this article.
References
- 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30. [DOI] [PubMed] [Google Scholar]
- 2. DeSantis CE, Ma J, Gaudet MM et al. Breast cancer statistics, 2019. CA Cancer J Clin 2019;69:438–451. [DOI] [PubMed] [Google Scholar]
- 3. McCart Reed AE, Kutasovic JR, Lakhani SR et al. Invasive lobular carcinoma of the breast: Morphology, biomarkers and 'omics. Breast Cancer Res 2015;17:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. SEER incidence data, 1975–2017. Avilable at https://seer.cancer.gov/data/. Accessed September 2020.
- 5. Li CI, Anderson BO, Daling JR et al. Trends in incidence rates of invasive lobular and ductal breast carcinoma. JAMA 2003;289:1421–1424. [DOI] [PubMed] [Google Scholar]
- 6. Farhat GN, Walker R, Buist DS et al. Changes in invasive breast cancer and ductal carcinoma in situ rates in relation to the decline in hormone therapy use. J Clin Oncol 2010;28:5140–5146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Yang LY, Yang LP, Zhu B. Clinicopathological characteristics and survival outcomes of invasive lobular carcinoma in different races. Oncotarget 2017;8:74287–74298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Arpino G, Bardou VJ, Clark GM et al. Infiltrating lobular carcinoma of the breast: Tumor characteristics and clinical outcome. Breast Cancer Res 2004;6:R149–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Rakha EA, El‐Sayed ME, Powe DG et al. Invasive lobular carcinoma of the breast: Response to hormonal therapy and outcomes. Eur J Cancer 2008;44:73–83. [DOI] [PubMed] [Google Scholar]
- 10. Li CI, Uribe DJ, Daling JR. Clinical characteristics of different histologic types of breast cancer. Br J Cancer 2005;93:1046–1052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. DiCostanzo D, Rosen PP, Gareen I et al. Prognosis in infiltrating lobular carcinoma. An analysis of "classical" and variant tumors. Am J Surg Pathol 1990;14:12–23. [DOI] [PubMed] [Google Scholar]
- 12. Lesser ML, Rosen PP, Kinne DW. Multicentricity and bilaterality in invasive breast carcinoma. Surgery 1982;91:234–240. [PubMed] [Google Scholar]
- 13. Porter AJ, Evans EB, Foxcroft LM et al. Mammographic and ultrasound features of invasive lobular carcinoma of the breast. J Med Imaging Radiat Oncol 2014;58:1–10. [DOI] [PubMed] [Google Scholar]
- 14. Reis‐Filho JS, Simpson PT, Turner NC, et al. FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clin Cancer Res 2006;12(22):6652–6662. [DOI] [PubMed] [Google Scholar]
- 15. Le Gal M, Ollivier L, Asselain B et al. Mammographic features of 455 invasive lobular carcinomas. Radiology 1992;185:705–708. [DOI] [PubMed] [Google Scholar]
- 16. Dabbs DJ, Bhargava R, Chivukula M. Lobular versus ductal breast neoplasms: The diagnostic utility of p120 catenin. Am J Surg Pathol 2007;31:427–437. [DOI] [PubMed] [Google Scholar]
- 17. Lien HC, Chen YL, Juang YL et al. Frequent alterations of HER2 through mutation, amplification, or overexpression in pleomorphic lobular carcinoma of the breast. Breast Cancer Res Treat 2015;150:447–455. [DOI] [PubMed] [Google Scholar]
- 18. Borst MJ, Ingold JA. Metastatic patterns of invasive lobular versus invasive ductal carcinoma of the breast. Surgery 1993;114:637–641; discussion 641–642. [PubMed] [Google Scholar]
- 19. McLemore EC, Pockaj BA, Reynolds C et al. Breast cancer: Presentation and intervention in women with gastrointestinal metastasis and carcinomatosis. Ann Surg Oncol 2005;12:886–894. [DOI] [PubMed] [Google Scholar]
- 20. Parekh RB, Tse AG, Dwek RA et al. Tissue‐specific N‐glycosylation, site‐specific oligosaccharide patterns and lentil lectin recognition of rat Thy‐1. EMBO J 1987;6:1233–1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Boukhechba M, Kadiri H, El Khannoussi B. Invasive lobular carcinoma of the breast with extracellular mucin: Case report of a new variant of lobular carcinoma of the breast. Case Rep Pathol 2018;2018:5362951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Makki J. Diversity of breast carcinoma: Histological subtypes and clinical relevance. Clin Med Insights Pathol 2015;8:23–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Thomas M, Kelly ED, Abraham J et al. Invasive lobular breast cancer: A review of pathogenesis, diagnosis, management, and future directions of early stage disease. Semin Oncol 2019;46:121–132. [DOI] [PubMed] [Google Scholar]
- 24. Engstrøm MJ, Opdahl S, Vatten LJ et al. Invasive lobular breast cancer: The prognostic impact of histopathological grade, E‐cadherin and molecular subtypes. Histopathology 2015;66:409–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Vargas AC, Lakhani SR, Simpson PT. Pleomorphic lobular carcinoma of the breast: Molecular pathology and clinical impact. Future Oncol 2009;5:233–243. [DOI] [PubMed] [Google Scholar]
- 26. Narendra S, Jenkins SM, Khoor A et al. Clinical outcome in pleomorphic lobular carcinoma: A case‐control study with comparison to classic invasive lobular carcinoma. Ann Diagn Pathol 2015;19:64–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Rakha EA, van Deurzen CH, Paish EC, et al. Pleomorphic lobular carcinoma of the breast: Is it a prognostically significant pathological subtype independent of histological grade? Mod Pathol 2013;26:496–501. [DOI] [PubMed] [Google Scholar]
- 28. Simpson PT, Reis‐Filho JS, Lambros MB 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. [DOI] [PubMed] [Google Scholar]
- 29. Eltorky M, Hall JC, Osborne PT et al. Signet‐ring cell variant of invasive lobular carcinoma of the breast. A clinicopathologic study of 11 cases. Arch Pathol Lab Med 1994;118:245–248. [PubMed] [Google Scholar]
- 30. Li P, Zheng J, Zhang T et al. Histiocytoid breast carcinoma: A case report showing immunohistochemical profiles. Int J Clin Exp Pathol 2013;6:2609–2614. [PMC free article] [PubMed] [Google Scholar]
- 31. Berx G, Staes K, van Hengel J et al. Cloning and characterization of the human invasion suppressor gene E‐cadherin (CDH1). Genomics 1995;26:281–289. [DOI] [PubMed] [Google Scholar]
- 32. Pećina‐Slaus N. Tumor suppressor gene E‐cadherin and its role in normal and malignant cells. Cancer Cell Int 2003;3:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Kim NG, Koh E, Chen X et al. E‐cadherin mediates contact inhibition of proliferation through Hippo signaling‐pathway components. Proc Natl Acad Sci USA 2011;108:11930–11935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Padua D, Massagué J. Roles of TGFbeta in metastasis. Cell Res 2009;19:89–102. [DOI] [PubMed] [Google Scholar]
- 35. Qian X, Karpova T, Sheppard AM et al. E‐cadherin‐mediated adhesion inhibits ligand‐dependent activation of diverse receptor tyrosine kinases. EMBO J 2004;23:1739–1748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Kim NG, Gumbiner BM. Adhesion to fibronectin regulates Hippo signaling via the FAK‐Src‐PI3K pathway. J Cell Biol 2015;210:503–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Ciriello G, Gatza ML, Beck AH et al. Comprehensive molecular portraits of invasive lobular breast cancer. Cell 2015;163:506–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Cerami E, Gao J, Dogrusoz U et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Gao J, Aksoy BA, Dogrusoz U et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1. [DOI] [PMC free article] [PubMed]
- 40. Cancer Genome Atlas Network . Comprehensive molecular portraits of human breast tumours. Nature 2012;490:61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Vos CB, Cleton‐Jansen AM, Berx G et al. E‐cadherin inactivation in lobular carcinoma in situ of the breast: An early event in tumorigenesis. Br J Cancer 1997;76:1131–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Sarrió D, Pérez‐Mies B, Hardisson D et al. Cytoplasmic localization of p120ctn and E‐cadherin loss characterize lobular breast carcinoma from preinvasive to metastatic lesions. Oncogene 2004;23:3272–3283. [DOI] [PubMed] [Google Scholar]
- 43. Kourtidis A, Ngok SP, Anastasiadis PZ. p120 catenin: An essential regulator of cadherin stability, adhesion‐induced signaling, and cancer progression. Prog Mol Biol Transl Sci 2013;116:409–432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Schackmann RC, van Amersfoort M, Haarhuis JH et al. Cytosolic p120‐catenin regulates growth of metastatic lobular carcinoma through Rock1‐mediated anoikis resistance. J Clin Invest 2011;121:3176–3188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. van de Ven RA, Tenhagen M, Meuleman W et al. Nuclear p120‐catenin regulates the anoikis resistance of mouse lobular breast cancer cells through Kaiso‐dependent Wnt11 expression. Dis Model Mech 2015;8:373–384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Turashvili G, Bouchal J, Burkadze G et al. Differentiation of tumours of ductal and lobular origin: I. Proteomics of invasive ductal and lobular breast carcinomas. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2005;149:57–62. [DOI] [PubMed] [Google Scholar]
- 47. Cao L, Basudan A, Sikora MJ et al. Frequent amplifications of ESR1, ERBB2 and MDM4 in primary invasive lobular breast carcinoma. Cancer Lett 2019;461:21–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Christgen M, Bartels S, Radner M et al. ERBB2 mutation frequency in lobular breast cancer with pleomorphic histology or high‐risk characteristics by molecular expression profiling. Genes Chromosomes Cancer 2019;58:175–185. [DOI] [PubMed] [Google Scholar]
- 49. Ross JS, Wang K, Sheehan CE et al. Relapsed classic E‐cadherin (CDH1)‐mutated invasive lobular breast cancer shows a high frequency of HER2 (ERBB2) gene mutations. Clin Cancer Res 2013;19:2668–2676. [DOI] [PubMed] [Google Scholar]
- 50. Deniziaut G, Tille JC, Bidard FC et al. ERBB2 mutations associated with solid variant of high‐grade invasive lobular breast carcinomas. Oncotarget 2016;7:73337–73346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Grellety T, Soubeyran I, Robert J et al. A clinical case of invasive lobular breast carcinoma with ERBB2 and CDH1 mutations presenting a dramatic response to anti‐HER2‐directed therapy. Ann Oncol 2016;27:199–200. [DOI] [PubMed] [Google Scholar]
- 52. Riggins RB, Lan JP, Zhu Y et al. ERRgamma mediates tamoxifen resistance in novel models of invasive lobular breast cancer. Cancer Res 2008;68:8908–8917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Schiff R, Reddy P, Ahotupa M et al. Oxidative stress and AP‐1 activity in tamoxifen‐resistant breast tumors in vivo. J Natl Cancer Inst 2000;92:1926–1934. [DOI] [PubMed] [Google Scholar]
- 54. Zhou Y, Yau C, Gray JW et al. Enhanced NF kappa B and AP‐1 transcriptional activity associated with antiestrogen resistant breast cancer. BMC Cancer 2007;7:59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Stires H, Heckler MM, Fu X et al. Integrated molecular analysis of Tamoxifen‐resistant invasive lobular breast cancer cells identifies MAPK and GRM/mGluR signaling as therapeutic vulnerabilities. Mol Cell Endocrinol 2018;471:105–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Du T, Sikora MJ, Levine KM et al. Key regulators of lipid metabolism drive endocrine resistance in invasive lobular breast cancer. Breast Cancer Res 2018;20:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Sikora MJ, Jacobsen BM, Levine K et al. WNT4 mediates estrogen receptor signaling and endocrine resistance in invasive lobular carcinoma cell lines. Breast Cancer Res 2016;18:92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Fu X, Jeselsohn R, Pereira R et al. FOXA1 overexpression mediates endocrine resistance by altering the ER transcriptome and IL‐8 expression in ER‐positive breast cancer. Proc Natl Acad Sci USA 2016;113:E6600–E6609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Glont SE, Chernukhin I, Carroll JS. Comprehensive genomic analysis reveals that the pioneering function of FOXA1 is independent of hormonal signaling. Cell Rep 2019;26:2558–2565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Ross‐Innes CS, Stark R, Teschendorff AE et al. Differential oestrogen receptor binding is associated with clinical outcome in breast cancer. Nature 2012;481:389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Guerrero‐Zotano A, Mayer IA, Arteaga CL. PI3K/AKT/mTOR: Role in breast cancer progression, drug resistance, and treatment. Cancer Metastasis Rev 2016;35:515–524. [DOI] [PubMed] [Google Scholar]
- 62. Janku F, Yap TA, Meric‐Bernstam F. Targeting the PI3K pathway in cancer: Are we making headway? Nat Rev Clin Oncol 2018;15:273–291. [DOI] [PubMed] [Google Scholar]
- 63. Feng Y, Spezia M, Huang S et al. Breast cancer development and progression: Risk factors, cancer stem cells, signaling pathways, genomics, and molecular pathogenesis. Genes Dis 2018;5:77–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Klarenbeek S, Doornebal CW, Kas SM et al. Response of metastatic mouse invasive lobular carcinoma to mTOR inhibition is partly mediated by the adaptive immune system. Oncoimmunology 2020;9:1724049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Levine KM, Priedigkeit N, Basudan A et al. FGFR4 overexpression and hotspot mutations in metastatic ER+ breast cancer are enriched in the lobular subtype. NPJ Breast Cancer 2019;5:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Chen Z, Yang J, Li S et al. Invasive lobular carcinoma of the breast: A special histological type compared with invasive ductal carcinoma. PLoS One 2017;12:e0182397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Montagner M, Sahai E. In vitro models of breast cancer metastatic dormancy. Front Cell Dev Biol 2020;8:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Pan H, Gray R, Braybrooke J et al. 20‐year risks of breast‐cancer recurrence after stopping endocrine therapy at 5 years. N Engl J Med 2017;377:1836–1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Du T, Zhu L, Levine KM et al. Invasive lobular and ductal breast carcinoma differ in immune response, protein translation efficiency and metabolism. Sci Rep 2018;8:7205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Narbe U, Bendahl PO, Aaltonen K et al. The distribution of circulating tumor cells is different in metastatic lobular compared to ductal carcinoma of the breast‐long‐term prognostic significance. Cells 2020;9:1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Ghajar CM. Metastasis prevention by targeting the dormant niche. Nat Rev Cancer 2015;15:238–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Derksen PW, Liu X, Saridin F et al. Somatic inactivation of E‐cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell 2006;10:437–449. [DOI] [PubMed] [Google Scholar]
- 73. Boussadia O, Kutsch S, Hierholzer A et al. E‐cadherin is a survival factor for the lactating mouse mammary gland. Mech Dev 2002;115:53–62. [DOI] [PubMed] [Google Scholar]
- 74. Kotb AM, Hierholzer A, Kemler R. Replacement of E‐cadherin by N‐cadherin in the mammary gland leads to fibrocystic changes and tumor formation. Breast Cancer Res 2011;13:R104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Derksen PW, Braumuller TM, van der Burg E et al. Mammary‐specific inactivation of E‐cadherin and p53 impairs functional gland development and leads to pleomorphic invasive lobular carcinoma in mice. Dis Model Mech 2011;4:347–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Boelens MC, Nethe M, Klarenbeek S, et al. PTEN loss in E‐cadherin‐deficient mouse mammary epithelial cells rescues apoptosis and results in development of classical invasive lobular carcinoma. Cell Rep 2016;16:2087–2101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. An Y, Adams JR, Hollern DP et al. Cdh1 and Pik3ca mutations cooperate to induce immune‐related invasive lobular carcinoma of the breast. Cell Rep 2018;25:702–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Annunziato S, Kas SM, Nethe M et al. Modeling invasive lobular breast carcinoma by CRISPR/Cas9‐mediated somatic genome editing of the mammary gland. Genes Dev 2016;30:1470–1480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Fodor J, Major T, Toth J et al. Comparison of mastectomy with breast‐conserving surgery in invasive lobular carcinoma: 15‐year results. Rep Pract Oncol Radiother 2011;16:227–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Wasif N, Maggard MA, Ko CY et al. Invasive lobular vs. ductal breast cancer: A stage‐matched comparison of outcomes. Ann Surg Oncol 2010;17:1862–1869. [DOI] [PubMed] [Google Scholar]
- 81. Wang K, Zhu GQ, Shi Y et al. Long‐term survival differences between T1‐2 invasive lobular breast cancer and corresponding ductal carcinoma after breast‐conserving surgery: A propensity‐scored matched longitudinal cohort study. Clin Breast Cancer 2019;19:e101–e115. [DOI] [PubMed] [Google Scholar]
- 82. Mamtani A, King TA. Lobular breast cancer: Different disease, different algorithms? Surg Oncol Clin N Am 2018;27:81–94. [DOI] [PubMed] [Google Scholar]
- 83. Piper ML, Wong J, Fahrner‐Scott K et al. Success rates of re‐excision after positive margins for invasive lobular carcinoma of the breast. NPJ Breast Cancer 2019;5:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Christgen M, Steinemann D, Kühnle E et al. Lobular breast cancer: Clinical, molecular and morphological characteristics. Pathol Res Pract 2016;212:583–597. [DOI] [PubMed] [Google Scholar]
- 85. Tiezzi DG, Andrade JM, Marana HR et al. Breast conserving surgery after neoadjuvant therapy for large primary breast cancer. Eur J Surg Oncol 2008;34:863–867. [DOI] [PubMed] [Google Scholar]
- 86. Untch M, Konecny GE, Paepke S et al. Current and future role of neoadjuvant therapy for breast cancer. Breast 2014;23:526–537. [DOI] [PubMed] [Google Scholar]
- 87. Tubiana‐Hulin M, Stevens D, Lasry S et al. Response to neoadjuvant chemotherapy in lobular and ductal breast carcinomas: A retrospective study on 860 patients from one institution. Ann Oncol 2006;17:1228–1233. [DOI] [PubMed] [Google Scholar]
- 88. Dixon JM, Renshaw L, Dixon J et al. Invasive lobular carcinoma: Response to neoadjuvant letrozole therapy. Breast Cancer Res Treat 2011;130:871–877. [DOI] [PubMed] [Google Scholar]
- 89.Fulvestrant and/or anastrozole in treating postmenopausal patients with stage ii‐iii breast cancer undergoing surgery. Available at https://www.clinicaltrials.gov/ct2/show/NCT01953588. Accessed September 2020.
- 90. Early Breast Cancer Trialists' Collaborative Group . Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15‐year survival: An overview of the randomised trials. Lancet 2005;365:1687–1717. [DOI] [PubMed] [Google Scholar]
- 91. Conlon N, Ross DS, Howard J et al. Is there a role for oncotype Dx testing in invasive lobular carcinoma? Breast J 2015;21:514–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Kizy S, Huang JL, Marmor S et al. Impact of the 21‐gene recurrence score on outcome in patients with invasive lobular carcinoma of the breast. Breast Cancer Res Treat 2017;165:757–763. [DOI] [PubMed] [Google Scholar]
- 93. Tsai ML, Lillemoe TJ, Finkelstein MJ et al. Utility of oncotype DX risk assessment in patients with invasive lobular carcinoma. Clin Breast Cancer 2016;16:45–50. [DOI] [PubMed] [Google Scholar]
- 94. Lænkholm AV, Jensen MB, Eriksen JO et al. Population‐based study of Prosigna‐PAM50 and outcome among postmenopausal women with estrogen receptor‐positive and HER2‐negative operable invasive lobular or ductal breast cancer. Clin Breast Cancer 2020;20:e423–e432. [DOI] [PubMed] [Google Scholar]
- 95. Truin W, Voogd AC, Vreugdenhil G et al. Effect of adjuvant chemotherapy in postmenopausal patients with invasive ductal versus lobular breast cancer. Ann Oncol 2012;23:2859–2865. [DOI] [PubMed] [Google Scholar]
- 96. Early Breast Cancer Trialists' Collaborative Group . Aromatase inhibitors versus tamoxifen in early breast cancer: Patient‐level meta‐analysis of the randomised trials. Lancet 2015;386:1341–1352. [DOI] [PubMed] [Google Scholar]
- 97. Early Breast Cancer Trialists' Collaborative Group , Davies C, Godwin J et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: Patient‐level meta‐analysis of randomised trials. Lancet 2011;378:771–784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Metzger Filho O, Giobbie‐Hurder A, Mallon E et al. Relative effectiveness of letrozole compared with tamoxifen for patients with lobular carcinoma in the BIG 1‐98 trial. J Clin Oncol 2015;33:2772–2779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Strasser‐Weippl K, Sudan G, Ramjeesingh R et al. Outcomes in women with invasive ductal or invasive lobular early stage breast cancer treated with anastrozole or exemestane in CCTG (NCIC CTG) MA.27. Eur J Cancer 2018;90:19–25. [DOI] [PubMed] [Google Scholar]
- 100. McCart Reed AE, Lal S, Kutasovic JR et al. LobSig is a multigene predictor of outcome in invasive lobular carcinoma. NPJ Breast Cancer 2019;5:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Bajrami I, Marlow R, van de Ven M et al. E‐cadherin/ROS1 inhibitor synthetic lethality in breast cancer. Cancer Discov 2018;8:498–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Johnston S, Puhalla S, Wheatley D et al. Randomized phase II study evaluating palbociclib in addition to letrozole as neoadjuvant therapy in estrogen receptor‐positive early breast cancer: PALLET trial. J Clin Oncol 2019;37:178–189. [DOI] [PubMed] [Google Scholar]
- 103. Michaut M, Chin SF, Majewski I et al. Integration of genomic, transcriptomic and proteomic data identifies two biologically distinct subtypes of invasive lobular breast cancer. Sci Rep 2016;6:18517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Mersin H, Yildirim E, Gülben K et al. Is invasive lobular carcinoma different from invasive ductal carcinoma? Eur J Surg Oncol 2003;29:390–395. [DOI] [PubMed] [Google Scholar]
- 105.Invasive lobular carcinoma (ILC). Available at https://www.breastcancer.org/symptoms/types/ilc. Accessed September 2020.
- 106. Moran MS, Yang Q, Haffty BG. 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. [DOI] [PubMed] [Google Scholar]
- 107.Invasive ductal carcinoma (IDC). Available at https://www.breastcancer.org/symptoms/types/idc. Accessed September 2020.
- 108.Invasive breast cancer (IDC/ILC). Available at https://www.cancer.org/cancer/breast-cancer/understanding-a-breast-cancer-diagnosis/types-of-breast-cancer/invasive-breast-cancer.html. Accessed September 2020.
- 109. Bombonati A, Sgroi DC The molecular pathology of breast cancer progression. J Pathol 2011;223:307–317. [DOI] [PMC free article] [PubMed] [Google Scholar]