Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management (2024)

Article Navigation

Volume 163 Issue 4 April 2022

Article Contents

  • Abstract

  • Epidemiology of DCIS

  • Histological Grades of DCIS

  • Molecular Mechanisms/Characteristics of DCIS Progression to IDC

  • Diagnosis and Treatments for DCIS

  • Conclusion

  • Acknowledgments

  • Financial Support

  • Conflict of Interest Statement

  • Data Availability

  • References

  • < Previous
  • Next >

Journal Article

,

Shelby Lynn Hophan

Department of Obstetrics and Gynecology, Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine

,

Chicago, IL 60611

,

USA

Search for other works by this author on:

Oxford Academic

,

Olena Odnokoz

Department of Pharmacology and Chemical Biology, Winship Cancer Center, Emory University School of Medicine

,

Atlanta, GA 30322

,

USA

Search for other works by this author on:

Oxford Academic

,

Huiping Liu

Department of Pharmacology, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine

,

Chicago, IL 60611

,

USA

Search for other works by this author on:

Oxford Academic

,

Yuan Luo

Department of Preventive Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine

,

Chicago, IL 60611

,

USA

Search for other works by this author on:

Oxford Academic

,

Seema Khan

Department of Surgery, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine

,

Chicago, IL 60611

,

USA

Search for other works by this author on:

Oxford Academic

,

William Gradishar

Department of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine

,

Chicago, IL 60611

,

USA

Search for other works by this author on:

Oxford Academic

,

Zhuan Zhou

Department of Surgery, UT Southwestern Medical Center

,

Dallas, TX 75390

,

USA

Search for other works by this author on:

Oxford Academic

,

Sunil Badve

Department of Pathology, Emory University School of Medicine

,

Atlanta, GA 30322

,

USA

Search for other works by this author on:

Oxford Academic

,

Mylin A Torres

Department of Hematology and Oncology, Winship Cancer Center, Emory University School of Medicine

,

Atlanta, GA 30322

,

USA

Search for other works by this author on:

Oxford Academic

Yong Wan

Department of Pharmacology and Chemical Biology, Winship Cancer Center, Emory University School of Medicine

,

Atlanta, GA 30322

,

USA

Department of Hematology and Oncology, Winship Cancer Center, Emory University School of Medicine

,

Atlanta, GA 30322

,

USA

Correspondence: Yong Wan, Department of Pharmacology and Chemical Biology, Winship Cancer Institute, Department of Hematology and Oncology, Emory University School of Medicine, 1365 Clifton Road, NE, Suite C 5090. Atlanta, GA 30322,

USA

. Email: yong.wan@emory.edu.

Search for other works by this author on:

Oxford Academic

Shelby Lynn Hophan and Olena Odnokoz share co-first authorship and have an equal contribution in preparation of this manuscript.

Author Notes

Endocrinology, Volume 163, Issue 4, April 2022, bqac027, https://doi.org/10.1210/endocr/bqac027

Published:

04 March 2022

Article history

Received:

21 October 2021

Editorial decision:

01 March 2022

Published:

04 March 2022

Corrected and typeset:

25 March 2022

  • PDF
  • Split View
  • Views
    • Article contents
    • Figures & tables
    • Video
    • Audio
    • Supplementary Data
  • Cite

    Cite

    Shelby Lynn Hophan, Olena Odnokoz, Huiping Liu, Yuan Luo, Seema Khan, William Gradishar, Zhuan Zhou, Sunil Badve, Mylin A Torres, Yong Wan, Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management, Endocrinology, Volume 163, Issue 4, April 2022, bqac027, https://doi.org/10.1210/endocr/bqac027

    Close

Search

Close

Search

Advanced Search

Search Menu

Abstract

Ductal carcinoma in situ (DCIS) makes up a majority of noninvasive breast cancer cases. DCIS is a neoplastic proliferation of epithelial cells within the ductal structure of the breast. Currently, there is little known about the progression of DCIS to invasive ductal carcinoma (IDC), or the molecular etiology behind each DCIS lesion or grade. The DCIS lesions can be heterogeneous in morphology, genetics, cellular biology, and clinical behavior, posing challenges to our understanding of the molecular mechanisms by which approximately half of all DCIS lesions progress to an invasive status. New strategies that pinpoint molecular mechanisms are necessary to overcome this gap in understanding, which is a barrier to more targeted therapy. In this review, we will discuss the etiological factors associated with DCIS, as well as the complexity of each nuclear grade lesion. Moreover, we will discuss the possible molecular features that lead to progression of DCIS to IDC. We will highlight current therapeutic management and areas for improvement.

ductal carcinoma in situ, breast cancer, invasive breast cancer, invasive ductal carcinoma

Breast cancer is the most common cancer in women in several developed and developing countries; more than half of these cases are in developed countries. In the United States, the rate of breast cancer–related deaths is second only to lung cancer. According to the American Cancer Society, in 2022 an estimated number of 287 850 new cases of invasive breast cancer (IBC) are expected to be diagnosed in women in the United States. Moreover, 51 400 women are estimated to be diagnosed with ductal carcinoma in situ (DCIS) (1).

DCIS is stage 0 of breast cancer and a nonobligatory precursor to invasive ductal carcinoma (IDC) (Fig. 1). It is a noninvasive form of breast cancer, which is characterized by a neoplastic proliferation of epithelial cells that are surrounded by myoepithelial cells. In DCIS cases, the myoepithelial cells are confined by an intact basem*nt membrane, which separates these cells from the breast stroma, and prevents the neoplastic cells from metastasizing. DCIS cells have a morphology similar to tumor cells but are still confined by the ductal structure. When detected early, or treated appropriately, this type of breast cancer may be prevented from progressing into a more invasive cancer.

Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management (6)

Figure 1.

Stages of breast cancer progression.

Open in new tabDownload slide

In fact, the intact basem*nt membrane is the key distinction between DCIS and IDC. DCIS cells can break through the membrane and invade surrounding breast tissue, progressing to IDC (2). IDC is the most common type of invasive breast cancer and accounts for approximately 80% of all invasive breast cancer cases. Although DCIS is noninvasive, affected women are at higher risk of developing IDC later in their lifetime. Currently, the standard treatment for DCIS is surgical intervention in the form of mastectomy or breast-conserving surgical resection followed by radiation therapy. However, the probability of recurrence within the same breast tissue is nearly 30% within 15 years after diagnosis (3). In most cases, DCIS is considered to be either over- or undertreated; thus, more time and effort must be devoted to fully understand the complexity of such disease to enable more efficient treatment measures.

Epidemiology of DCIS

DCIS makes up a relatively high proportion of newly diagnosed noninvasive breast cancer cases within North America. In screened populations, DCIS is typically detected as a small cluster of calcifications, but the area can be large, spanning several centimeters, or even occupying more than one quadrant of the breast. The widespread implementation of mammography screening has resulted in a dramatic increase of the incident rate of DCIS during the past few decades; however, these screening-detected DCIS lesions tend to be associated with a lower grade and smaller size (4). In fact, Surveillance, Epidemiology, and End Results (SEER) program data suggest that there was a 500% increase in DCIS cases from 1993 to 2003 among women older than 50 years, and a decline since 2003 (5), which could be explained by the decrease in use of postmenopausal hormonal therapies (6). The 20-year risk of breast cancer–related mortality is 3.3% among women with DCIS (7).

The development of DCIS lesions and IDC have similar risk factors, suggesting a common etiology for both diseases. Both carcinoma lesions have identical etiological relationships, including increasing age, family history, late age of first birth, late onset of menopause, elevated body mass index in postmenopausal women, and high breast density (8). Detailed studies of DCIS and existing IDC shows intratumoral genetic heterogeneity, cellular biology, and behavior (9).

Histological Grades of DCIS

Multiple systems have been used to identify histological grade of DCIS. The nuclear grade is an important part of overall tumor grade, which evaluates nuclear features of malignant cells. The presence or absence of comedonecrosis, or a buildup of dead (necrotic) cells within the tumor, is another key pathological component. There are 3 grades of DCIS: low grade (Grade I), intermediate grade (Grade 2), and high grade (Grade 3) (Table 1, Fig. 2); where increasing grades possess features ranging from normal or atypical ductal hyperplasia to pleomorphic carcinoma cells with irregular and large nuclei (Table 1). The low-grade DCIS resembles atypical ductal hyperplasia cells but can look more like normal breast cells. The intermediate-grade DCIS closely resembles a lower grade DCIS lesion but grows faster than normal breast cells. The high-grade DCIS tends to grow at a faster rate and looks abnormal. Patients with high-grade lesions have a higher risk of IDC and recurrence. Comedonecrosis is usually associated with high-grade DCIS.

Table 1.

Open in new tab

DCIS classification and Van Nuys Prognostic Index scoring

Grade ER/PR/HER2 Status Nuclear grade VNPI Scoring
Low grade DCIS (Grade 1)ER: Positive (+)Small size nuclei with a diameter of 1-1.5× the size of RBCFeature Score: 1
PR: Positive (+)Nuclei are round monotonousSize: ≤ 15 mm
HER2: Negative (-) Nucleoli are absent or indistinctMargin: ≥ 10 mm
Age: > 60 years
Nuclear grade 1 or 2
Without necrosis
Overall score: 4-6
Intermediate grade DCIS (Grade 2)ER: Positive (+)Nuclei size is between Grade 1 and 3, with a diameter of 1.5-2.5× the size of RBCFeature Score: 2
PR: Positive (+)Nuclei are moderate pleomorphic lacking monotonySize: 16-40 mm
HER2: Negative (-)Nucleoli are occasionalMargin: 1-9 mm
Age: 40-60 years
Nuclear grade 1 or 2
With necrosis
Overall score: 7-9
High grade DCIS (Grade 3)ER: Negative (-)Large pleomorphic nuclei with a diameter of > 2.5× the size of RBCFeature Score: 3
PR: Negative (-)Nucleoli are prominent, often multipleSize: > 40 mm
HER2: Positive (+)Margin: < 1 mm
Age: < 40 years
Nuclear grade 3
With or without necrosis
Overall score: 10-12
Grade ER/PR/HER2 Status Nuclear grade VNPI Scoring
Low grade DCIS (Grade 1)ER: Positive (+)Small size nuclei with a diameter of 1-1.5× the size of RBCFeature Score: 1
PR: Positive (+)Nuclei are round monotonousSize: ≤ 15 mm
HER2: Negative (-) Nucleoli are absent or indistinctMargin: ≥ 10 mm
Age: > 60 years
Nuclear grade 1 or 2
Without necrosis
Overall score: 4-6
Intermediate grade DCIS (Grade 2)ER: Positive (+)Nuclei size is between Grade 1 and 3, with a diameter of 1.5-2.5× the size of RBCFeature Score: 2
PR: Positive (+)Nuclei are moderate pleomorphic lacking monotonySize: 16-40 mm
HER2: Negative (-)Nucleoli are occasionalMargin: 1-9 mm
Age: 40-60 years
Nuclear grade 1 or 2
With necrosis
Overall score: 7-9
High grade DCIS (Grade 3)ER: Negative (-)Large pleomorphic nuclei with a diameter of > 2.5× the size of RBCFeature Score: 3
PR: Negative (-)Nucleoli are prominent, often multipleSize: > 40 mm
HER2: Positive (+)Margin: < 1 mm
Age: < 40 years
Nuclear grade 3
With or without necrosis
Overall score: 10-12

Ductal carcinoma in situ (DCIS) lesions can be classified based on receptor status, cell morphology, and VNPI scoring (based on size, margin, histology, and age of the individual). Most lesions can be labeled as intermediate grade (Grade II) if the cell morphology analysis falls between Grade I and III. The VNPI score of each lesion determines the proper excision approach when it comes to a treatment plan. Each score is based on an algorithm that predicts local recurrence in conservatively treated patients with DCIS. A VNPI score of 4-6 accounts for 33% of patients, with a 97% local recurrence-free survival, and wide-local excision (WLE) would be the treatment plan. A VNPI score of 7-9 accounts for 58% of patients, who have a 73% local recurrence-free survival, and the appropriate treatment would be WLE with radiotherapy (RT). A VNPI score of 10-12 accounts for 11% of patients, who have a 34% local recurrence-free survival rate, and the appropriate treatment would be a full mastectomy.

Abbreviations: DCIS, ductal carcinoma in situ; ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; PR, progesterone receptor; RBC, red blood cell; VNPI, Van Nuys Prognostic Index.

Table 1.

Open in new tab

DCIS classification and Van Nuys Prognostic Index scoring

Grade ER/PR/HER2 Status Nuclear grade VNPI Scoring
Low grade DCIS (Grade 1)ER: Positive (+)Small size nuclei with a diameter of 1-1.5× the size of RBCFeature Score: 1
PR: Positive (+)Nuclei are round monotonousSize: ≤ 15 mm
HER2: Negative (-) Nucleoli are absent or indistinctMargin: ≥ 10 mm
Age: > 60 years
Nuclear grade 1 or 2
Without necrosis
Overall score: 4-6
Intermediate grade DCIS (Grade 2)ER: Positive (+)Nuclei size is between Grade 1 and 3, with a diameter of 1.5-2.5× the size of RBCFeature Score: 2
PR: Positive (+)Nuclei are moderate pleomorphic lacking monotonySize: 16-40 mm
HER2: Negative (-)Nucleoli are occasionalMargin: 1-9 mm
Age: 40-60 years
Nuclear grade 1 or 2
With necrosis
Overall score: 7-9
High grade DCIS (Grade 3)ER: Negative (-)Large pleomorphic nuclei with a diameter of > 2.5× the size of RBCFeature Score: 3
PR: Negative (-)Nucleoli are prominent, often multipleSize: > 40 mm
HER2: Positive (+)Margin: < 1 mm
Age: < 40 years
Nuclear grade 3
With or without necrosis
Overall score: 10-12
Grade ER/PR/HER2 Status Nuclear grade VNPI Scoring
Low grade DCIS (Grade 1)ER: Positive (+)Small size nuclei with a diameter of 1-1.5× the size of RBCFeature Score: 1
PR: Positive (+)Nuclei are round monotonousSize: ≤ 15 mm
HER2: Negative (-) Nucleoli are absent or indistinctMargin: ≥ 10 mm
Age: > 60 years
Nuclear grade 1 or 2
Without necrosis
Overall score: 4-6
Intermediate grade DCIS (Grade 2)ER: Positive (+)Nuclei size is between Grade 1 and 3, with a diameter of 1.5-2.5× the size of RBCFeature Score: 2
PR: Positive (+)Nuclei are moderate pleomorphic lacking monotonySize: 16-40 mm
HER2: Negative (-)Nucleoli are occasionalMargin: 1-9 mm
Age: 40-60 years
Nuclear grade 1 or 2
With necrosis
Overall score: 7-9
High grade DCIS (Grade 3)ER: Negative (-)Large pleomorphic nuclei with a diameter of > 2.5× the size of RBCFeature Score: 3
PR: Negative (-)Nucleoli are prominent, often multipleSize: > 40 mm
HER2: Positive (+)Margin: < 1 mm
Age: < 40 years
Nuclear grade 3
With or without necrosis
Overall score: 10-12

Ductal carcinoma in situ (DCIS) lesions can be classified based on receptor status, cell morphology, and VNPI scoring (based on size, margin, histology, and age of the individual). Most lesions can be labeled as intermediate grade (Grade II) if the cell morphology analysis falls between Grade I and III. The VNPI score of each lesion determines the proper excision approach when it comes to a treatment plan. Each score is based on an algorithm that predicts local recurrence in conservatively treated patients with DCIS. A VNPI score of 4-6 accounts for 33% of patients, with a 97% local recurrence-free survival, and wide-local excision (WLE) would be the treatment plan. A VNPI score of 7-9 accounts for 58% of patients, who have a 73% local recurrence-free survival, and the appropriate treatment would be WLE with radiotherapy (RT). A VNPI score of 10-12 accounts for 11% of patients, who have a 34% local recurrence-free survival rate, and the appropriate treatment would be a full mastectomy.

Abbreviations: DCIS, ductal carcinoma in situ; ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; PR, progesterone receptor; RBC, red blood cell; VNPI, Van Nuys Prognostic Index.

Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management (7)

Figure 2.

Grading and characteristics of DCIS. Ductal carcinoma in situ (DCIS) lesions are classified based on their hormone receptor status, morphology, and comedo status. The DCIS grades are Grade I (low grade), Grade II (intermediate grade), and Grade III (high grade). Grade I and II lesions are less likely to develop into invasive ductal carcinoma (IDC); however, Grade III lesions are at higher risk of breaking through the basem*nt membrane into surrounding breast tissue. Comedonecrosis, or buildup of dead cells inside the tumor, is often present in Grade III DCIS lesions. The microenvironment surrounding the DCIS cells, including myoepithelial cells, fibroblasts, and lymphocytes, contributes to progression of the lesions.

Open in new tabDownload slide

Some DCIS lesions can have microinvasions (MI) (Fig. 2) or be known as DCIS-MI (8). DCIS-MI is a rare diagnosis, comprising 10% to 20% of DCIS cases and approximately 1% of all breast cancer cases. The American Joint Committee on Cancer (AJCC) staging system defined MI as cancer cells breaching through the wall of the duct and entering the adjacent stroma with invasive focus not more than 1 millimeter in greatest dimension (10). Compared with pure DCIS, DCIS-MI have both a noninvasive DCIS component and a small component of invasive disease. Although the invasive component is relatively small, prognosis in DCIS-MI is more similar to IDC than pure DCIS (11). DCIS-MI is the first step of invasion into the nearby stroma, or tissue past the basem*nt membrane, which further illustrates the importance of the complexity of this disease.

Interestingly, most substantial cases of DCIS show multiple grades within the same lesions (8). This knowledge might help fully understand the evolution of this carcinoma, but most importantly, what causes the progression of DCIS to become more invasive. It’s important to understand that each lesion can consist of one or more nuclear grades, since that information would direct the treatment plan.

Most DCIS lesions have an intertwining makeup of several different nuclear grades, each aided by multiple cells within the microenvironment (Fig. 2). Despite the prevalence of DCIS, there is no uniform accepted classification system for diagnosing this disease. There has been a growing opinion as to the importance of grade identification over its phenotypic morphology (8).

Emergence of diversity during breast cancer evolution could help explain the reasoning behind multiple histologic nuclear grade lesions (Fig. 3). It is believed that DCIS influences the progression to IDC. There are 3 main theories behind progression of DCIS to IDC, understood through the idea of evolution: Independent Evolution, Evolutionary Bottleneck, and Multiclonal Invasion (12). Both Evolutionary Bottleneck and Multiclonal Invasion are a direct lineage from a single clonal ancestry cell. Allred and colleagues analyzed 120 cases of pure DCIS to understand the nuclear grade makeup, as well as evaluate the biological diversity among different grades of DCIS to further understand its role within breast cancer evolution. According to the study, 46% of cases showed the presence of multiple histologic nuclear grades, primarily between Grade 1 and Grade 2, and 33% showed diversity of several important biomarkers (13).

Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management (8)

Figure 3.

Evolutionary models of DCIS progression. Independent evolution method allows the theory that DCIS (in situ) and invasion cells evolve independently of each other. These cells originate from 2 different normal cells within the breast environment. Evolutionary Bottleneck proposes that evolution of 3 subpopulations have a single clonal ancestral cell, from which a single cell is selected to become invasive. Multiclonal Invasion is similar to Evolutionary Bottleneck; however, all 3 clonal cells escape in situ and migrate into adjacent tissues to become invasive.

Open in new tabDownload slide

Molecular Mechanisms/Characteristics of DCIS Progression to IDC

The molecular characteristics of DCIS and mechanisms of its progression to IDC are still poorly investigated. The heterogeneity of DCIS lesions and variation in clinicopathological characteristics, such as estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2/ERBB2) status, create challenges in identifying unique markers that is associated with high risk of progression to IDC.

Nonetheless, previous studies identified specific genomic alterations for different grades of DCIS lesions and for different stages of breast cancer progression from ductal hyperplasia to IDC (14). For example, amplification of FGFR1 and MYC are more frequent compared to DCIS (14-16). The variation within these genomic features between DCIS and IDC could be related to the tumor microenvironment (17), cancer stem-like cell (CSC) populations, myoepithelial differentiation, acquisition of mesenchymal phenotype, changes in expression of cancer-related genes, and some epigenetic changes that can contribute to breast cancer progression (18). It was shown that DCIS cells and tumors contain high cancer stem-like cell subpopulations (ALDH1 + and CD44+/CD49f+/CD24-) with increased self-renewal, treatment resistance, and tumor development capabilities (19).

The recent study using in vitro 3D culture model of DCIS showed that reduction of Rap1Gap, a negative regulator of the small GTPase Rap1, resulted in ERK/MAPK activation, cytoskeletal reorganization, epithelial-mesenchymal transition (EMT) and invasion (20). Ectopic expression of Rap1Gap reversed the invasive phenotype. Immunohistochemistry (IHC) analysis of DCIS human samples compared with normal breast tissue and IDCs shows an increased Rap1Gap expression levels (20). This study suggested that Rap1Gap can function as a switch from DCIS to IDC.

ER, PR, and HER2

Most DCIS lesions are positive for ER and PR, while also being HER2-negative (21-23). These lesions can be closely classified as being Grade 1 or 2 (Table 1). Comparison analysis of DCIS and IDC lesions have not shown significant differences in the expression of ER and PR (22). The HER2 is amplified in 20% of IDCs, associated with increased proliferation abnormalities, which promotes cell motility that contributes to tumor cells metastasizing (22). Regarding DCIS lesions, HER2 is overexpressed in higher grades, such as Grade 3 (Table 1) (23, 24). Interestingly, lesions with HER2 amplification were found with higher frequency in DCIS than in IDC tumors (22).

The St. Gallen system can be used to classify DCIS into surrogate molecular subtypes (25). Five surrogate molecular subtypes were used to classify 381 DCIS cases: Luminal A (ER + and/or PR+, HER2−, Ki67 < 14%), Luminal B/HER2− (ER and/or PR+, HER2−, Ki67 ≥ 14%), Luminal B/HER2+ (ER+ and/or PR+, HER2+), HER2+/ER− (nonluminal) (ER−, PR−, HER2+), and Triple negative (ductal) (ER−, PR−, HER2−) (26). Identifying these subtypes within DCIS cases further confirms that these lesions are precursors of the different subtypes of IDC carcinomas. The ability to correlate these subtypes raises the possibility of preventing DCIS lesions from progressing, similar to that of IDC.

Tumor Suppressor Genes

Surprisingly, the genetic comparison between DCIS and IDC shows identical genetic abnormalities when it comes to loss of tumor suppressors and amplifications. Like most cancer derived cells, DCIS cells show a faster proliferation rate than most cells surrounding it, which contributes to their positive growth imbalance. The progressive growth of DCIS comes from the alterations of normal growth-regulating mechanisms and microenvironment.

The tumor suppressor gene TP53 is located on chromosome 17p13 and encodes 53-kDa nuclear phosphoprotein, which regulates a cell cycle and a programed cell death (27). Loss of tumor suppressor gene TP53 is thought to be an early driver of breast cancer and to occur before the development of IDC, in particularly in DCIS of nuclear grade 2 or 3 (23, 28, 29). TP53 mutation was not found in normal breast epithelium, hyperplasia areas, or DCIS of nuclear grade 1 (29).

In normal breast tissue, TP63, a member of the tumor suppressor p53 family of transcriptional factors, is associated with differentiated myoepithelial-specific genes and is essential for development and maintenance of epithelial stem cells. In cancer, loss of TP63 expression promotes migration, invasion, and distant metastasis. DCIS cells showed a significant decrease in TP63 + population and impaired differentiation of myoepithelial cells (30).

Myoepithelial Differentiation

Myoepithelial cells play essential roles in normal mammary gland development by contributing to the differentiation, proliferation, and polarity of luminal epithelial cells, branching and elongation of ducts, and milk production (31-33). The layer of myoepithelial cells and the basem*nt membrane outline the duct lumen, form a structural barrier, and serve as a limiting factor of DCIS to IDC progression (30). The intact continuous myoepithelial cell layer and basem*nt membrane inhibit penetration by malignant cells into surrounding tissue and progression to invasive disease. Immunohistochemical analysis of myoepithelial markers including smooth muscle actin (SMA), calponin, p63, smooth muscle myosin heavy chain (SMMHC), cytokeratins (CKs) 5/6, P-cadherin, and CD10 can be used to detect the presence or absence of an intact myoepithelial cell layer and differentiate between DCIS and IDC (30, 34).

P-cadherin is a cell adhesion molecule within myoepithelial cells that helps with intercellular connection, cellular migration, and growth (35). P-cadherin is frequently overexpressed in high-grade IDC and is an enhancer of migration, which can be correlated with tumor aggressiveness (36). When comparing P-cadherin levels across DCIS and IDC cases, the majority of lesions were positive for P-cadherin in the luminal cells of the DCIS (35).

Microenvironment

In DCIS, the proliferative epithelial cells are enclosed in a continuous myoepithelial basem*nt membrane along with enrichment of immune cells and fibroblasts (Fig. 2) (37). The progression of DCIS to IDC happens when the continuity of myoepithelial basem*nt membrane is disrupted, and the tumor cells invade into the stroma. The primarily collagenous stroma of a normal duct contains fibroblasts and immune cells.

T cells are rarely detected inside the duct (38). In DCIS, an intact myoepithelial cell layer and basem*nt membrane serve as a barrier between immune system in stroma and the intraductal tumor cells. During transition from DCIS to IDC, the status of the immune microenvironment changes to become more immunosuppressive (38, 39). In DCIS, there is an activated immune environment surrounding the ductal structure, with enrichment of cytotoxic CD8 + T cells (38, 39). Compared with DCIS, IDCs contained fewer activated MKi67+CD8+ and GZMB+CD8+ T cells (38). In IDC, there is a suppressed immune environment, with enrichment of Treg cells (38, 39). DCIS and IDC also have significant differences in expression of several immune checkpoint proteins which may contribute to immune escape during breast cancer progression (38, 39). In particular, DCIS contained more TIGIT+ T cells, whereas IDCs had higher expression of PD-L1 and CTLA4 (38, 39).

Besides promoting DCIS invasion, fibroblasts can help aid the rapid development of most breast cancer lesions. Specifically, breast cancer–associated fibroblasts (CAFs) release growth factors, angiogenic factors, and proteases into the tumor microenvironment that support tumor growth, angiogenesis, metastasis, and therapy resistance (40).

In IDC, the fibroblasts transform into CAFs, which express PDGFRα/β, αSMA, FAP, and FSP1 (41). The CAFs assist tumor cells invading into the surrounding stroma. Fibroblasts produce proteases, like matrix metalloproteinases (MMPs) that degrade the extracellular matrix, which results in the stromal reorganization and releasing of growth factors (42).

Several studies have been conducted to understand how fibroblasts are capable of contributing to DCIS invasion. Sung et al showed that co-culture with human mammary fibroblasts induced invasion of MCF10DCIS cells through collagen reorganization (43). Using a 3D model, Hu and colleagues co-cultured fibroblasts with MCF10DCIS cells within a recombinant basem*nt membrane (44). From this study, it was shown that there was an increased invasive branching within the Matrigel and higher levels of MMP-9 and MMP-14 with the presence of fibroblasts (44). MMP-9 promotes cancer cell invasion and disease progression whereas MMP-14 activates other MMP family members to promote cancer (45). Using the mammary architecture and microenvironment engineering 3D model, Osuala and colleagues demonstrated that CAF-derived IL-6 contributed to invasion of MCF10DCIS cells (46). Another study identified that CAF-derived chemokine CXCL1 induced DCIS invasion in the MMTV-PyMT mouse model (47). Moreover, Brummer et al conducted a study using a mammary intraductal injection DCIS model, demonstrating that CCL2/CCR2 chemokine signaling promoted invasion of DCIS cells through fibroblast-dependent mechanisms (48).

Diagnosis and Treatments for DCIS

Since the implementation of mammography screening, there has been a dramatic increase of the incident rate of DCIS. Mammograms, an x-ray imaging of the breast, are the most efficient way to detect breast cancer cases early on its progressive state. Mammographic screening accounts for majority of diagnosed DCIS cases. DCIS presents on a mammography as microcalcifications, or bright white clusters, that have irregular sizes and shapes (49). Further evaluation with core breast needle biopsy is essential for determining the diagnosis and grade of DCIS.

There are no clear guidelines to predict which DCIS cases would progress into IDC and require aggressive treatment as opposed to cases that would remain indolent during a women’s lifetime. As a result, many women with low-risk DCIS are overtreated with surgery (lumpectomy or mastectomy), radiation, and hormone therapy (50). Between 1991 and 2010, approximately 70% of DCIS cases were treated with breast-conserving surgery with or without radiation, and approximately 25% of DCIS patients underwent mastectomy (50). Only a very few cases of mastectomy are followed by breast reconstruction. The appropriate treatment plan option depends on a variety of clinical-pathological factors like age and the extent of the disease (8). Most breast-conserving surgical cases are associated with minor excision of the ductal structure affected.

In order to identify the most suitable treatment for every single DCIS, the Van Nuys Prognostic Index (VNPI) was established based on characterizing the most important predictive factors and combining them in an algorithm (Table 1). VNPI scores can be associated with predicting the 10-year local recurrence-free survival in treated patients with DCIS, the percent of those that would remain cancer-free after excision, and which excision is more appropriate for the given lesions (23). Those with a low score (VNPI 4-6) account for 33% of patients, have a 97% local recurrence-free survival, and wide-local excision (WLE) would be the treatment plan. Those with an intermediate score (VNPI 7-9) account for 57% of patients, have a 73% local recurrence-free survival, and the appropriate treatment would be WLE with radiotherapy (RT). Those with a high score (VNPI 10-12) account for 11% of patients, have a 34% local recurrence-free survival, and the appropriate treatment would be a full mastectomy. RT after DCIS reduced the risk of local recurrence by 50% but did not reduce the risk of dying of breast cancer (51). Based on lesion size, margin width, histological features, and patient’s age, the recommended treatment could be breast-conserving surgery alone, lumpectomy followed by adjuvant radiotherapy, or mastectomy (Table 1) (52). Other scoring methods have been used to determine both DCIS recurrence and to predict benefits of RT. OncotypeDX DCIS scoring was the first clinically validated genomic test to determine a 10-year risk of local recurrence using a 12-gene assay and was shown to complement traditional clinical or pathologic factors (53, 54). Another scoring method developed is DCISionRT (PreludeDX), which analyzes the expression of specific markers and provides information associated with the benefits of RT (54). With this scoring method, physicians can determine if a patient who has undergone breast-conserving surgery would benefit overall from RT, therefore limiting unnecessary radiational exposure (55).

There has been very little progress to identify innovative targeted treatments for DCIS, simply because of its intertwined subtypes, with different biological potential (56). Each subtype of DCIS is composed of different biological potential; thus, the only successful targeted treatment for DCIS is tamoxifen, which is used for Grade I or II ER-positive lesions. For 21 years, tamoxifen has been known for treating IDC. Essentially, tamoxifen binds to ER on the cancer cells and prevents estrogen from binding in return prohibiting cell proliferation. Tamoxifen is used for estrogen and progesterone receptor-positive diseases, consistent with the biologic mechanism of action of the drug (57).

To confirm the significant benefits of tamoxifen, Allred and colleagues used a clinical trial to evaluate ER and PR relationships in DCIS after lumpectomy and radiation, followed by adjuvant tamoxifen (58). This study was undertaken to evaluate the relationship between adjuvant tamoxifen and the receptor status among DCIS lesions. With prolonged follow-up, the results showed that the adjuvant tamoxifen had a significant reduction of ipsilateral breast cancer in patients with ER-positive DCIS lesions; similarly, tamoxifen reduced contralateral breast cancer in patients with ER-positive and ER-negative DCIS lesions. However, in ER-negative only DCIS lesions, there was no ipsilateral benefit with tamoxifen. This emphasized that tamoxifen must bind to functional ER to be effective on preexisting tumor cells (57, 58).

A similar trial was conducted to demonstrate the impact and effect when combining the use of tamoxifen and lumpectomy with radiation. The results showed an additional 37% reduction in relative risk of local recurrence and a decrease in contralateral breast cancer of comparable magnitude (59). The RTOG 9804 trial showed that RT significantly decreases ipsilateral recurrence in the good-risk subset of DCIS patients (60). These studies potentially offer patients and physicians an additional option that is not only therapeutic but more efficient.

Wesseling and colleagues published a meta-analysis summary of the current knowledge and prognostic factors for invasive disease after a diagnosis of DCIS. Most of the factors pointed to a higher relative risk of subsequent IDC for patients with DCIS, but the effects were generally small. Six prognostic factors had a statistically significant pooled estimate (61). The 6 factors were race, premenopausal status, detection by palpation, involved surgical margin, high histologic grade, and high p16 expression, the majority of which can be biologically explained. A recent study used immunofluorescence multiplexing and single cell analysis to study co-expression of breast cancer associated cellular biomarkers in limited formalin-fixed, paraffin-embedded DCIS tissue microarrays (62). Using the developed multi-step comprehensive analysis, the investigators demonstrated that high HER2 and low ER and PR in DCIS samples were associated with breast cancer events. In contrast, Ki67 and COX2 did not show significant correlation with breast cancer events. A large cohort study is needed for further validation of this prognostic signature (62).

Conclusion

Why study DCIS? The core reason for studying this disease is to understand its etiological factors. Some patients diagnosed with DCIS have the possibility of progressing to IDC if left untreated or treated incorrectly. Currently, the most used treatment for DCIS is a lumpectomy followed by RT. The treatment for DCIS patients could be improved based on comprehension of which lesions have the potential to become invasive. The problem is that all lesions lack different capacities for progression under a microscope. Since only 20% to 50% of DCIS cases progress to IDC if left untreated and since the patients with DCIS after surgical resection with or without radiation have an overall good prognosis, DCIS is often overlooked. The primary focus is on understanding the invasive disease and its treatment strategies, rather than on DCIS. Thus, there is a lot unknown about DCIS. This unknown aspect leads to the misperception of whether this disease can be determined as invasive.

To fully grasp the progression and explanation of the mechanisms of DCIS, one must uncover the molecular pathways involved and the risk factors associated with each lesion on DCIS that allows invasive properties to evolve. After a patient has DCIS lesions, it takes approximately 10 years for these lesions to achieve an IDC diagnosis. Because of this, some scientists and doctors are not convinced of the urgency to further investigate DCIS lesions or search for better treatment strategies. If the gap of knowledge was breached, or a better comprehension of DCIS lesions on a molecular level was understood, then more manageable measures would be taken that could nearly eliminate the current mortality rate associated with this diagnosis.

Breast cancer makes up an entire field of researchers, professors, and doctors, who share the same goal to help the 300 000 women affected yearly by this cancer. Breast cancer research is a multibillion-dollar industry. However, most of these funds support research in invasive breast cancer rather than DCIS. There is little to no research conducted to elucidate the progression of DCIS to IDC, or the mechanism to explain why certain lesions progress beyond the basem*nt membrane into its surrounding tissues. Members of the National Institutes of Health DCIS conference proposed that the word carcinoma should be removed from the term ductal carcinoma in situ because DCIS is noninvasive and has a favorable prognosis. The term indolent lesions of epithelial origin (IDLE) has been proposed for use instead of DCIS (54). However, experimental studies of human and mouse DCIS lesions are showing the opposite: carcinoma precursor cells exist in these lesions, and the aggressive phenotype of breast cancer is predetermined early at the premalignant stage (2).

By identifying the mechanism that inhibits the progression of DCIS to IDC, a major unknown about DCIS would become known. This discovery alone will broaden the understanding of breast cancer, as well as develop a more concrete explanation of why this carcinoma might be the initiator of IDC. This would create a big impact within the field and change the outlook of DCIS.

Abbreviations

    Abbreviations

  • CAF

    cancer-associated fibroblast

  • DCIS

    ductal carcinoma in situ

  • EMT

    epithelial-mesenchymal transition

  • ER

    estrogen receptor

  • HER2

    human epidermal growth factor receptor 2

  • IDC

    invasive ductal carcinoma

  • MI

    microinvasive (microinvasions)

  • MMP

    matrix metalloproteinase

  • PR

    progesterone receptor

  • RT

    radiotherapy

  • VNPI

    Van Nuys Prognostic Index

  • WLE

    wide-local excision

Acknowledgments

This work was supported by Emory startup and the Northwestern University Zell scholar fund.

Financial Support

Emory startup. NIH grant: R01CA202948.

Conflict of Interest Statement

The authors declare that there are no conflicts of interest.

Data Availability

Data sharing is not applicable to this article as no new data have been included in this review.

References

1.

American Cancer Society.

Cancer Facts & Figures 2022.

Atlanta

:

American Cancer Society

;

2022

.

Google Scholar

OpenURL Placeholder Text

2.

Kim

SY

,

Jung

SH

,

Kim

MS

, et al.

Genomic differences between pure ductal carcinoma in situ and synchronous ductal carcinoma in situ with invasive breast cancer

.

Oncotarget.

2015

;

6

(

10

):

7597

-

7607

.

Google Scholar

OpenURL Placeholder Text

3.

Wapnir

IL

,

Dignam

JJ

,

Fisher

B

, et al.

Long-term outcomes of invasive ipsilateral breast tumor recurrences after lumpectomy in NSABP B-17 and B-24 randomized clinical trials for DCIS

.

J Natl Cancer Inst.

2011

;

103

(

6

):

478

-

488

.

Google Scholar

OpenURL Placeholder Text

4.

Fadare

O

,

Clement

NF

,

Ghofrani

M

.

High and intermediate grade ductal carcinoma in-situ of the breast: a comparison of pathologic features in core biopsies and excisions and an evaluation of core biopsy features that may predict a close or positive margin in the excision

.

Diagn Pathol.

2009

;

4

:

26

.

Google Scholar

OpenURL Placeholder Text

5.

Kerlikowske

K

.

Epidemiology of ductal carcinoma in situ

.

JNCI Monographs.

2010

;

2010

(

41

):

139

-

141

.

Google Scholar

OpenURL Placeholder Text

6.

Kerlikowske

K

,

Miglioretti

DL

,

Buist

DSM

,

Walker

R

,

Carney

PA

.

Declines in invasive breast cancer and use of postmenopausal hormone therapy in a screening mammography population

.

J Natl Cancer Inst.

2007

;

99

(

17

):

1335

-

1339

.

7.

DeChant

CA

,

Thomas

SM

,

Rosenberger

LH

, et al.

Ductal carcinoma in situ in the elderly: what is the ideal treatment plan?

J Unexplored Med Data.

2019

;4:2.

Google Scholar

OpenURL Placeholder Text

8.

Bane

A

.

Ductal carcinoma in situ: what the pathologist needs to know and why

.

Int J Breast Cancer.

2013

;

2013

:

914053

.

Google Scholar

OpenURL Placeholder Text

9.

Doebar

SC

,

Krol

NM

,

van Marion

R

, et al.

Progression of ductal carcinoma in situ to invasive breast cancer: comparative genomic sequencing

.

Virchows Arch.

2019

;

474

(

2

):

247

-

251

.

Google Scholar

OpenURL Placeholder Text

10.

Edge

SB

,

Compton

CC

.

The American Joint Committee on Cancer: the 7th edition of the AJCC cancer staging manual and the future of TNM

.

Ann Surg Oncol.

2010

;

17

(

6

):

1471

-

1474

.

Google Scholar

OpenURL Placeholder Text

11.

Champion

CD

,

Ren

Y

,

Thomas

SM

, et al.

DCIS with microinvasion: is it in situ or invasive disease?

Ann Surg Oncol.

2019

;

26

(

10

):

3124

-

3132

.

Google Scholar

OpenURL Placeholder Text

12.

Casasent

AK

,

Edgerton

M

,

Navin

NE

.

Genome evolution in ductal carcinoma in situ: invasion of the clones

.

J Pathol.

2017

;

241

(

2

):

208

-

218

.

Google Scholar

OpenURL Placeholder Text

13.

Allred

DC

,

Wu

Y

,

Mao

S

, et al.

Ductal carcinoma in situ and the emergence of diversity during breast cancer evolution

.

Clin Cancer Res.

2008

;

14

(

2

):

370

-

378

.

Google Scholar

OpenURL Placeholder Text

14.

Lesurf

R

,

Aure

MR

,

Mork

HH

, et al.

Molecular features of subtype-specific progression from ductal carcinoma in situ to invasive breast cancer

.

Cell Rep.

2016

;

16

(

4

):

1166

-

1179

.

Google Scholar

OpenURL Placeholder Text

15.

Jang

MH

,

Kim

EJ

,

Choi

Y

, et al.

FGFR1 is amplified during the progression of in situ to invasive breast carcinoma

.

Breast Cancer Res.

2012

;

14

(

4

):R115.

Google Scholar

OpenURL Placeholder Text

16.

Robanus-Maandag

EC

,

Bosch

CAJ

,

Kristel

PM

, et al.

Association of C-MYC amplification with progression from the in situ to the invasive stage in C-MYC-amplified breast carcinomas

.

J Pathol.

2003

;

201

(

1

):

75

-

82

.

Google Scholar

OpenURL Placeholder Text

17.

Lee

S

,

Stewart

S

,

Nagtegaal

I

, et al.

Differentially expressed genes regulating the progression of ductal carcinoma in situ to invasive breast cancer

.

Cancer Res.

2012

;

72

(

17

):

4574

-

4586

.

Google Scholar

OpenURL Placeholder Text

18.

Fleischer

T

,

Frigessi

A

,

Johnson

KC

, et al.

Genome-wide DNA methylation profiles in progression to

.

Genome Biol.

2014

;

15

(

8

):

435

.

Google Scholar

OpenURL Placeholder Text

19.

Duru

N

,

Gernapudi

R

,

Lo

PK

, et al.

Characterization of the CD49f+/CD44+/CD24- single-cell derived stem cell population in basal-like DCIS cells

.

Oncotarget.

2016

;

7

(

30

):

47511

-

47525

.

Google Scholar

OpenURL Placeholder Text

20.

Shah

S

,

Brock

EJ

,

Jackson

RM

, et al.

Downregulation of Rap1Gap: a switch from DCIS to invasive breast carcinoma via ERK/MAPK activation

.

Neoplasia.

2018

;

20

(

9

):

951

-

963

.

Google Scholar

OpenURL Placeholder Text

21.

Dobrescu

A

,

Chang

M

,

Kirtani

V

,

Turi

GK

,

Hennawy

R

,

Hindenburg

AA

.

Study of estrogen receptor and progesterone receptor expression in breast ductal carcinoma in situ by immunohistochemical staining in ER/PgR-negative invasive breast cancer

.

ISRN Oncol.

2011

;

2011

:

673790

.

Google Scholar

OpenURL Placeholder Text

22.

Zhang

Y

,

Wu

Y

,

Huo

LJ

,

Yan

SS

,

Wang

DD

,

Gao

HY

.

Molecular alterations differentiate microinvasive carcinoma from ductal carcinoma in situ and invasive breast carcinoma: retrospective analysis of a large single-center series

.

Int J Clin Exp Pathol.

2021

;

14

(

8

):

892

-

901

.

Google Scholar

OpenURL Placeholder Text

23.

Gorringe

KL

,

Fox

SB

.

Ductal carcinoma in situ biology, biomarkers, and diagnosis

.

Front Oncol.

2017

;

7

:

248

.

Google Scholar

OpenURL Placeholder Text

24.

Yin

J

,

Gascard

P

,

Last

B

,

Singh

M

,

Tlsty

T

,

Tjoe

J

.

Abstract P5-18-04: HER2 overexpression in ductal carcinoma in situ: a biomarker for risk stratification and therapeutic implication

.

Cancer Res.

2019

;

79

(

4 Supplement

):

P5-18-04

-

P15-18-04

.

Google Scholar

OpenURL Placeholder Text

25.

Goldhirsch

A

,

Wood

WC

,

Coates

AS

, et al.

Strategies for subtypes--dealing with the diversity of breast cancer: highlights of the St. Gallen international expert consensus on the primary therapy of early breast cancer 2011

.

Ann Oncol.

2011

;

22

(

8

):

1736

-

1747

.

Google Scholar

OpenURL Placeholder Text

26.

Zhou

W

,

Jirström

K

,

Amini

R-M

, et al.

Molecular subtypes in ductal carcinoma in situ ofthe breast and their relation to prognosis: a population-based cohort study

.

BMC Cancer.

2013

;13:512. doi:10.1186/1471-2407-13-512

Google Scholar

OpenURL Placeholder Text

27.

Martin

J

,

Dufour

JF

.

Tumor suppressor and hepatocellular carcinoma

.

World J Gastroenterol.

2008

;

14

(

11

):

1720

-

1733

.

Google Scholar

OpenURL Placeholder Text

28.

Abba

MC

,

Gong

T

,

Lu

Y

, et al.

A molecular portrait of high-grade ductal carcinoma in situ

.

Cancer Res.

2015

;

75

(

18

):

3980

-

3990

.

Google Scholar

OpenURL Placeholder Text

29.

Done

SJ

,

Eskandarian

S

,

Bull

S

,

Redston

M

,

Andrulis

IL

.

p53 missense mutations in microdissected high-grade ductal carcinoma in situ of the breast

.

J Natl Cancer Inst

.

2001

;

93

(

9

):

700

-

704

.

Google Scholar

OpenURL Placeholder Text

30.

Ding

L

,

Su

Y

,

Fassl

A

, et al.

Perturbed myoepithelial cell differentiation in BRCA mutation carriers and in ductal carcinoma in situ

.

Nat Commun.

2019

;

10

(

1

):

4182

.

Google Scholar

OpenURL Placeholder Text

31.

Deugnier

M-A

,

Teulière

J

,

Faraldo

MM

,

Thiery

JP

,

Glukhova

MA

.

The importance of being a myoepithelial cell

.

Breast Cancer Res.

2002

;

4

(

6

):224-230. doi: 10.1186/bcr459

Google Scholar

OpenURL Placeholder Text

32.

Moumen

M

,

Chiche

A

,

Cagnet

S

, et al.

The mammary myoepithelial cell

.

Int J Dev Biol.

2011

;

55

(

7-9

):

763

-

771

.

Google Scholar

OpenURL Placeholder Text

33.

Adriance

MC

,

Inman

JL

,

Petersen

OW

,

Bissell

MJ

.

Myoepithelial cells: good fences make good neighbors

.

Breast Cancer Res.

2005

;

7

(

5

):

190

-

197

.

Google Scholar

OpenURL Placeholder Text

34.

Russell

TD

,

Jindal

S

,

Agunbiade

S

, et al.

Myoepithelial cell differentiation markers in ductal carcinoma in situ progression

.

Am J Pathol.

2015

;

185

(

11

):

3076

-

3089

.

Google Scholar

OpenURL Placeholder Text

35.

Kanjanapradit

K

,

Wangsawibul

S

.

P-cadherin and CD10 expression to distinguish between ductal carcinoma in situ and invasive ductal carcinoma of the breast

.

J Health Sci Med Res.

2019

.

Google Scholar

OpenURL Placeholder Text

36.

Paredes

J

,

Correia

A

,

Ribeiro

A

,

Albergaria

A

,

Milanezi

F

,

Schmitt

FC

.

P-cadherin expression in breast cancer: a review

.

Breast Cancer Res

.

2007

;

9

(

5

):

214

.

37.

Nelson

AC

,

Machado

HL

,

Schwertfeger

KL

.

Breaking through to the other side: microenvironment contributions to DCIS initiation and progression

.

J Mammary Gland Biol Neoplasia.

2018

;

23

(

4

):

207

-

221

.

Google Scholar

OpenURL Placeholder Text

38.

Gil Del Alcazar

CR

,

Huh

SJ

,

Ekram

MB

, et al.

Immune escape in breast cancer during in situ to invasive carcinoma transition

.

Cancer Discov.

2017

;

7

(

10

):

1098

-

1115

.

Google Scholar

OpenURL Placeholder Text

39.

Hendry

S

,

Pang

JB

,

Byrne

DJ

, et al.

Relationship of the breast ductal carcinoma in situ immune microenvironment with clinicopathological and genetic features

.

Clin Cancer Res.

2017

;

23

(

17

):

5210

-

5217

.

Google Scholar

OpenURL Placeholder Text

40.

Tao

L

,

Huang

G

,

Song

H

,

Chen

Y

,

Chen

L

.

Cancer associated fibroblasts: an essential role in the tumor microenvironment

.

Oncol Lett.

2017

;

14

(

3

):

2611

-

2620

.

Google Scholar

OpenURL Placeholder Text

41.

Cichon

MA

,

Degnim

AC

,

Visscher

DW

,

Radisky

DC

.

Microenvironmental influences that drive progression from benign breast disease to invasive breast cancer

.

J Mammary Gland Biol Neoplasia.

2010

;

15

(

4

):

389

-

397

.

Google Scholar

OpenURL Placeholder Text

42.

Inman

JL

,

Robertson

C

,

Mott

JD

,

Bissell

MJ

.

Mammary gland development: cell fate specification, stem cells and the microenvironment

.

Development.

2015

;

142

(

6

):

1028

-

1042

.

Google Scholar

OpenURL Placeholder Text

43.

Sung

KE

,

Yang

N

,

Pehlke

C

, et al.

Transition to invasion in breast cancer: a microfluidic in vitro model enables examination of spatial and temporal effects

.

Integr Biol.

2011

;

3

(

4

):

439

-

450

.

Google Scholar

OpenURL Placeholder Text

44.

Hu

M

,

Peluffo

G

,

Chen

H

,

Gelman

R

,

Schnitt

S

,

Polyak

K

.

Role of COX-2 in epithelial-stromal cell interactions and progression of ductal carcinoma in situ of the breast

.

Proc Natl Acad Sci USA.

2009

;

106

(

9

):

3372

-

3377

.

Google Scholar

OpenURL Placeholder Text

45.

Shirian

J

,

Arkadash

V

,

Cohen

I

, et al.

Converting a broad matrix metalloproteinase family inhibitor into a specific inhibitor of MMP ‐9 and MMP ‐14

.

FEBS Lett.

2018

;

592

(

7

):

1122

-

1134

.

Google Scholar

OpenURL Placeholder Text

46.

Osuala

KO

,

Sameni

M

,

Shah

S

, et al.

Il-6 signaling between ductal carcinoma in situ cells and carcinoma-associated fibroblasts mediates tumor cell growth and migration

.

BMC Cancer.

2015

;

15

:

584

.

Google Scholar

OpenURL Placeholder Text

47.

Bernard

S

,

Myers

M

,

Fang

WB

, et al.

CXCL1 derived from mammary fibroblasts promotes progression of mammary lesions to invasive carcinoma through CXCR2 dependent mechanisms

.

J Mammary Gland Biol Neoplasia.

2018

;

23

(

4

):

249

-

267

.

Google Scholar

OpenURL Placeholder Text

48.

Brummer

G

,

Acevedo

DS

,

Hu

Q

, et al.

Chemokine signaling facilitates early-stage breast cancer survival and invasion through fibroblast-dependent mechanisms

.

Mol Cancer Res.

2018

;

16

(

2

):

296

-

308

.

Google Scholar

OpenURL Placeholder Text

49.

Shao

YZ

,

Liu

LZ

,

Bie

MJ

, et al.

Characterizing the clustered microcalcifications on mammograms to predict the pathological classification and grading: a mathematical modeling approach

.

J Digit Imaging.

2011

;

24

(

5

):

764

-

771

.

Google Scholar

OpenURL Placeholder Text

50.

Hwang

ES

,

Hyslop

T

,

Lynch

T

, et al.

The COMET (Comparison of Operative versus Monitoring and Endocrine Therapy) trial: a phase III randomised controlled clinical trial for low-risk ductal carcinoma in situ (DCIS)

.

BMJ Open.

2019

;

9

(

3

):

e026797

.

Google Scholar

OpenURL Placeholder Text

51.

Narod

SA

,

Sopik

V

.

Is invasion a necessary step for metastases in breast cancer?

Breast Cancer Res Treat.

2018

;

169

(

1

):

9

-

23

.

Google Scholar

OpenURL Placeholder Text

52.

Mariotti

C

, and

Raffaeli

E

.

The surgical treatment of DCIS: from local excision to conservative breast surgery and conservative mastectomies

. In:

Mariotti

C

, ed.

Ductal Carcinoma in Situ of the Breast

.

Springer

;

2018

.

Google Scholar

OpenURL Placeholder Text

53.

Solin

LJ

,

Gray

R

,

Baehner

FL

, et al.

A multigene expression assay to predict local recurrence risk for ductal carcinoma in situ of the breast

.

J Natl Cancer Inst.

2013

;

105

(

10

):

701

-

710

.

Google Scholar

OpenURL Placeholder Text

54.

Badve

SS

,

Gokmen-Polar

Y

.

Ductal carcinoma in situ of breast: update 2019

.

Pathology.

2019

;

51

(

6

):

563

-

569

.

Google Scholar

OpenURL Placeholder Text

55.

Shah

C

,

Bremer

T

,

Cox

C

, et al.

The clinical utility of DCISionRT((R)) on radiation therapy decision making in patients with ductal carcinoma in situ following breast-conserving surgery

.

Ann Surg Oncol.

2021

;

28

(

11

):

5974

-

5984

.

Google Scholar

OpenURL Placeholder Text

56.

Siziopikou

KP

.

Ductal carcinoma in situ of the breast: current concepts and future directions

.

Arch Pathol Lab Med.

2013

;

137

(

4

):

462

-

466

.

Google Scholar

OpenURL Placeholder Text

57.

Cuzick

J

,

Sestak

I

,

Pinder

SE

, et al.

Effect of tamoxifen and radiotherapy in women with locally excised ductal carcinoma in situ: long-term results from the UK/ANZ DCIS trial

.

Lancet Oncol.

2011

;

12

(

1

):

21

-

29

.

Google Scholar

OpenURL Placeholder Text

58.

Allred

DC

,

Anderson

SJ

,

Paik

S

, et al.

Adjuvant tamoxifen reduces subsequent breast cancer in women with estrogen receptor-positive ductal carcinoma in situ: a study based on NSABP protocol B-24

.

J Clin Oncol.

2012

;

30

(

12

):

1268

-

1273

.

Google Scholar

OpenURL Placeholder Text

59.

Fisher

B

,

Dignam

J

,

Wolmark

N

, et al.

Tamoxifen in treatment of intraductal breast cancer: National Surgical Adjuvant Breast and Bowel Project B-24 randomised controlled trial

.

Lancet

1999

;

353

(

9169

):

1993

-

2000

.

Google Scholar

OpenURL Placeholder Text

60.

McCormick

B

,

Winter

K

,

Hudis

C

, et al.

RTOG 9804: a prospective randomized trial for good-risk ductal carcinoma in situ comparing radiotherapy with observation

.

J Clin Oncol.

2015

;

33

(

7

):

709

-

715

.

Google Scholar

OpenURL Placeholder Text

61.

Perron

M

.

Overtreatment of DCIS: new meta-analysis Identifies Prog

.

Oncol Times.

2019

;

41

(

13

):

1912

-

1922

.

Google Scholar

OpenURL Placeholder Text

62.

Badve

SS

,

Cho

S

,

Gokmen-Polar

Y

, et al.

Multi-protein spatial signatures in ductal carcinoma in situ (DCIS) of breast

.

Br J Cancer.

2021

;

124

(

6

):

1150

-

1159

.

Google Scholar

OpenURL Placeholder Text

Author notes

Shelby Lynn Hophan and Olena Odnokoz share co-first authorship and have an equal contribution in preparation of this manuscript.

© The Author(s) 2022. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Download all slides

Advertisem*nt

Citations

Views

2,025

Altmetric

More metrics information

Metrics

Total Views 2,025

1,324 Pageviews

701 PDF Downloads

Since 3/1/2022

Month: Total Views:
March 2022 41
April 2022 130
May 2022 103
June 2022 128
July 2022 118
August 2022 123
September 2022 19
October 2022 45
November 2022 17
December 2022 7
January 2023 36
February 2023 21
March 2023 18
April 2023 80
May 2023 84
June 2023 72
July 2023 101
August 2023 58
September 2023 94
October 2023 87
November 2023 77
December 2023 64
January 2024 79
February 2024 85
March 2024 67
April 2024 124
May 2024 70
June 2024 64
July 2024 13

Citations

Powered by Dimensions

6 Web of Science

Altmetrics

×

Email alerts

Article activity alert

Advance article alerts

New issue alert

Receive exclusive offers and updates from Oxford Academic

Citing articles via

Google Scholar

  • Latest

  • Most Read

  • Most Cited

Beta-hydroxybutyrate promotes basal insulin secretion while decreasing glucagon secretion in mouse and human islets
Vitamin D Receptor Regulates Liver Regeneration After Partial Hepatectomy in Male Mice
Reduced nephrin tyrosine phosphorylation enhances insulin secretion and increases glucose tolerance with age
Gut-Specific Neprilysin Deletion Protects Against Fat-Induced Insulin Secretory Dysfunction In Male Mice
Characterization of m6A Modifiers and RNA Modifications in Uterine Fibroids

More from Oxford Academic

Clinical Medicine

Endocrinology and Diabetes

Medicine and Health

Books

Journals

Advertisem*nt

Ductal Carcinoma In Situ of Breast: From Molecular Etiology to Therapeutic Management (2024)
Top Articles
DAX am Wochenende traden | DAX Weekend Trading erklärt
Dax Weekend Trading: den Dax über das Wochenende handeln
Swissport Timecard
Msc Open House Fall 2023
Food Universe Near Me Circular
Buff Streams .Io
Camila Cabello Wikifeet
Pollen Levels Richmond
Savannah Rae Demers Fanfix
Colossians 2 Amplified
Netlearning Login Rwjbh
Generation Zero beginner’s guide: six indispensable tips to help you survive the robot revolution
Hangar 67
Entegra Forum
Shahala Middle School Shahala Middle School Student Handbook
Browse | Obituaries | Enid News and Eagle
Power Outage Hales Corners
Lufkin Isd Calendar
Promotional Code For Spades Royale
FREE Printable Pets Animal Playdough Mats
Chi Trib Weather
Free 120 Step 2 Correlation
H. P. Lovecraft - Deutsche Lovecraft Gesellschaft
Tbom Genesis Retail Phone Number
Mercedes E-Klasse Rembekrachtigers voorraad | Onderdelenlijn.nl
Rochester Ny Missed Connections
Perry County Mugshots Busted
Express-Reisepass beantragen - hamburg.de
Roomba I3 Sealing Problem With Clean Base
Creator League Standings
Chrissy Laboy Daughter
Durrell: The Alexandria Quartet - The Modern Novel
Www.lookmovie.og
6030 Topsail Rd, Lady Lake, FL 32159 - MLS G5087027 - Coldwell Banker
Slim Thug’s Wealth and Wellness: A Journey Beyond Music
M3Gan Showtimes Near Cinemark North Hills And Xd
Central Valley growers, undocumented farmworkers condemn Trump's 'emergency'
Credit Bureau Contact Information
Glassbox Eyecare
Hourly Pay At Dick's Sporting Goods
Enterprise Car Sales Jacksonville Used Cars
Autozone Cercano
Texas State Final Grades
Did You Hear About Worksheet Answers Page 211
Sam's Club Gas Price Hilliard
Burkes Outlet Credit Card Sign In
Photogeek Goddess
University Of Michigan Paging System
Puppiwi World : Age, Height, Family, Relationship Status, Net Worth, Wiki, and More Including Exclusive Insights! WikistarFact
Portmanteau Structure Built With Cans
Saryn Prime Build 2023
Evil Dead Rise Showtimes Near Regal Destiny Usa
Latest Posts
Article information

Author: Ms. Lucile Johns

Last Updated:

Views: 5267

Rating: 4 / 5 (61 voted)

Reviews: 84% of readers found this page helpful

Author information

Name: Ms. Lucile Johns

Birthday: 1999-11-16

Address: Suite 237 56046 Walsh Coves, West Enid, VT 46557

Phone: +59115435987187

Job: Education Supervisor

Hobby: Genealogy, Stone skipping, Skydiving, Nordic skating, Couponing, Coloring, Gardening

Introduction: My name is Ms. Lucile Johns, I am a successful, friendly, friendly, homely, adventurous, handsome, delightful person who loves writing and wants to share my knowledge and understanding with you.