1
|
Van Bockstal MR, Wesseling J, Lips EH, Smidt M, Galant C, van Deurzen CHM. Systematic assessment of HER2 status in ductal carcinoma in situ of the breast: a perspective on the potential clinical relevance. Breast Cancer Res 2024; 26:125. [PMID: 39192322 DOI: 10.1186/s13058-024-01875-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024] Open
Abstract
In many countries, hormone receptor status assessment of ductal carcinoma in situ (DCIS) is routinely performed, as hormone receptor-positive DCIS patients are eligible for adjuvant anti-hormonal treatment, aiming to reduce the ipsilateral and contralateral breast cancer risk. Although HER2 gene amplification and its associated HER2 protein overexpression constitute a major prognostic and predictive marker in invasive breast carcinoma, its use in the diagnosis and treatment of DCIS is less straightforward. HER2 immunohistochemistry is not routinely performed yet, as the role of HER2-positivity in DCIS biology is unclear. Nonetheless, recent data challenge this practice. Here, we discuss the value of routine HER2 assessment for DCIS. HER2-positivity correlates strongly with DCIS grade: around four in five HER2-positive DCIS show high grade atypia. As morphological DCIS grading is prone to interobserver variability, HER2 immunohistochemistry could render grading more robust. Several studies showed an association between HER2-positive DCIS and ipsilateral recurrence risk, albeit currently unclear whether this is for overall, in situ or invasive recurrence. HER2-positive DCIS tends to be larger, with a higher risk of involved surgical margins. HER2-positive DCIS patients benefit more from adjuvant radiotherapy: it substantially decreases the local recurrence risk after lumpectomy, without impact on overall survival. HER2-positivity in pure biopsy-diagnosed DCIS is associated with increased upstaging to invasive carcinoma after surgery. HER2 immunohistochemistry on preoperative biopsies might therefore provide useful information to surgeons, favoring wider excisions. The time seems right to consider DCIS subtype-dependent treatment, comprising appropriate local treatment for HER2-positive DCIS patients and de-escalation for hormone receptor-positive, HER2-negative DCIS patients.
Collapse
MESH Headings
- Humans
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Female
- Breast Neoplasms/pathology
- Breast Neoplasms/metabolism
- Breast Neoplasms/genetics
- Breast Neoplasms/therapy
- Breast Neoplasms/mortality
- Breast Neoplasms/diagnosis
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Carcinoma, Intraductal, Noninfiltrating/therapy
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Biomarkers, Tumor/metabolism
- Prognosis
- Immunohistochemistry
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/genetics
- Neoplasm Grading
- Clinical Relevance
Collapse
Affiliation(s)
- Mieke R Van Bockstal
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.
- Pôle de Morphologie (MORF), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| | - Jelle Wesseling
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
- Department of Pathology, The Netherlands Cancer Institute - Antoni van Leeuwenhoek, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
- Department of Pathology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Ester H Lips
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX, Amsterdam, The Netherlands
| | - Marjolein Smidt
- Department of Surgery, Maastricht University Medical Center, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Christine Galant
- Department of Pathology, Cliniques universitaires Saint-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
- Pôle de Morphologie (MORF), Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Avenue Hippocrate 10, 1200, Brussels, Belgium
| | - Carolien H M van Deurzen
- Department of Pathology, Erasmus MC Cancer Institute Rotterdam, Doctor Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Delaloge S, Khan SA, Wesseling J, Whelan T. Ductal carcinoma in situ of the breast: finding the balance between overtreatment and undertreatment. Lancet 2024; 403:2734-2746. [PMID: 38735296 DOI: 10.1016/s0140-6736(24)00425-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 01/10/2024] [Accepted: 02/29/2024] [Indexed: 05/14/2024]
Abstract
Ductal carcinoma in situ (DCIS) accounts for 15-25% of all breast cancer diagnoses. Its prognosis is excellent overall, the main risk being the occurrence of local breast events, as most cases of DCIS do not progress to invasive cancer. Systematic screening has greatly increased the incidence of this non-obligate precursor of invasion, lending urgency to the need to identify DCIS that is prone to invasive progression and distinguish it from non-invasion-prone DCIS, as the latter can be overdiagnosed and therefore overtreated. Treatment strategies, including surgery, radiotherapy, and optional endocrine therapy, decrease the risk of local events, but have no effect on survival outcomes. Active surveillance is being evaluated as a possible new option for low-risk DCIS. Considerable efforts to decipher the biology of DCIS have led to a better understanding of the factors that determine its variable natural history. Given this variability, shared decision making regarding optimal, personalised treatment strategies is the most appropriate course of action. Well designed, risk-based de-escalation studies remain a major need in this field.
Collapse
Affiliation(s)
- Suzette Delaloge
- Department of Cancer Medicine, Interception Programme, Gustave Roussy, Villejuif, France.
| | - Seema Ahsan Khan
- Department of Surgery, Northwestern University, Chicago, IL, USA
| | - Jelle Wesseling
- Divisions of Molecular Pathology & Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands; Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | - Timothy Whelan
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
3
|
Hamaoka T, Bando H, Okazaki M, Iguchi-Manaka A, Hara H. Two Cases of Distant Metastasis After Mastectomy for Breast Ductal Carcinoma In Situ. Cureus 2024; 16:e59655. [PMID: 38836147 PMCID: PMC11147741 DOI: 10.7759/cureus.59655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 06/06/2024] Open
Abstract
While the prognosis for ductal carcinoma in situ (DCIS) of the breast is generally excellent, distant metastasis after appropriate local treatment is extremely rare. We experienced two cases of distant metastasis after mastectomy for breast ductal carcinoma in situ. In both cases, the surgical margins were negative, the sentinel nodes were negative for metastasis. The first case was a 67-year-old woman who developed lung metastases four years after mastectomy for high-grade DCIS. The second case was a 34-year-old woman with intermediate-grade DCIS who developed intraductal recurrence localized to the nipple two years after the initial nipple-sparing mastectomy and multiple lung and liver metastases six months later. Both cases developed distant metastases despite appropriate local treatment, without preceding or concurrent invasive local recurrence. Although the probability of distant recurrence is low, it is important to inform patients about the risk of recurrence.
Collapse
Affiliation(s)
- Takeru Hamaoka
- Department of Breast and Endocrine Surgery, University of Tsukuba Hospital, Ibaraki, JPN
| | - Hiroko Bando
- Institute of Medicine, Breast and Endocrine Surgery, University of Tsukuba, Ibaraki, JPN
| | - Mai Okazaki
- Department of Breast and Endocrine Surgery, University of Tsukuba Hospital, Ibaraki, JPN
| | - Akiko Iguchi-Manaka
- Institute of Medicine, Breast and Endocrine Surgery, University of Tsukuba, Ibaraki, JPN
| | - Hisato Hara
- Institute of Medicine, Breast and Endocrine Surgery, University of Tsukuba, Ibaraki, JPN
| |
Collapse
|
4
|
Lobo CL, Shetty A, M M, Dubey A, El-Zahaby SA. Non-systemic Approaches for Ductal Carcinoma In Situ: Exploring the Potential of Ultra-flexible Combisomes as a Novel Drug Delivery Strategy-a Review. AAPS PharmSciTech 2023; 24:119. [PMID: 37173545 DOI: 10.1208/s12249-023-02574-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Ductal carcinoma in situ (DCIS) is currently treated through breast-conserving surgery (lumpectomy), radiation therapy, breast-removing surgery (mastectomy), and hormone therapy to prevent further progression into invasive breast cancer and recurrence. Discrepancies concerning the prognosis of DCIS have sparked controversy about adequate treatment. Considering the severe medical and psychological consequences of mastectomy, developing a treatment approach that arrests the progression of DCIS to the invasive stage without affecting the non-cancerous cells is of utmost importance. In the current review, the problems associated with the diagnosis and management of DCIS have been thoroughly discussed. A summary of the route of administration and drug delivery systems to manage DCIS was also provoked. Innovative ultra-flexible combisomes were also proposed for the effective management of DCIS. Prevention is essential in managing the risk of DCIS and reducing the risk of progression to invasive breast cancer. While prevention is vital, it is not always possible to prevent DCIS, and in some cases, treatment may be necessary. Hence, this review recommends that ultra-flexible combisomes administered as a topical gel provide a non-systemic approach for managing DCIS and thus significantly minimize the side effects and costs associated with existing therapies.
Collapse
Affiliation(s)
- Cynthia Lizzie Lobo
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Amitha Shetty
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Manohar M
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Deralakatte, Mangalore, 575018, India.
| | - Sally A El-Zahaby
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, King Salman International University, South Sinai, Egypt
| |
Collapse
|
5
|
Akrida I, Mulita F. The clinical significance of HER2 expression in DCIS. Med Oncol 2022; 40:16. [PMID: 36352293 DOI: 10.1007/s12032-022-01876-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/15/2022] [Indexed: 11/11/2022]
Abstract
HER2 is an established prognostic and predictive marker for patients with invasive breast cancer. The clinical and biological significance of HER2 overexpression in patients with ductal carcinoma in situ (DCIS) remains poorly defined. DCIS is a heterogeneous disease and some patients with DCIS will not progress to invasive breast cancer. However, clinically significant recurrence rates have been reported after breast-conserving surgery for DCIS and approximately half of these cases will be life-threatening invasive recurrences. Since the incidence of DCIS is rising due to the widespread use of screening mammography, there is robust interest in selecting high-risk DCIS patients that may benefit from adjuvant therapies. Molecular prognostic and predictive models in early invasive breast cancer help clinicians identify patients that will benefit from chemotherapy. Molecular subtyping and profiling could also be useful in treating DCIS patients. According to current practice guidelines, HER2 testing is not recommended in DCIS patients. Nevertheless, evidence suggests that HER2-positive DCIS cases may be associated with adverse clinicopathological parameters and increased recurrence rates. This review summarizes the existing body of evidence linking HER2 expression and ipsilateral breast cancer recurrence in DCIS. HER2, as well as its correlation with other clinicopathological markers might be a useful prognostic and predictive marker, helping clinical decision-making in DCIS patients.
Collapse
Affiliation(s)
- Ioanna Akrida
- Department of General Surgery, University General Hospital of Patras, Rion, Greece.
- Department of Anatomy-Histology-Embryology, University of Patras Medical School, 26504, Rion, Greece.
| | - Francesk Mulita
- Department of General Surgery, University General Hospital of Patras, Rion, Greece
| |
Collapse
|
6
|
Xu F, Cao L, Xu C, Cai G, Cai R, Qi W, Wang S, Shen K, Chai W, Chen J. A Novel Nomogram for Predicting Prognosis and Tailoring Local Therapy Decision for Ductal Carcinoma In Situ after Breast Conserving Surgery. J Clin Med 2022; 11:jcm11175188. [PMID: 36079116 PMCID: PMC9456583 DOI: 10.3390/jcm11175188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose: We sought to explore the role of nomogram-combined biomarkers, mammographic microcalcification and inflammatory hematologic markers in guiding local therapy decisions in ductal carcinoma in situ (DCIS) subgroups with different ipsilateral breast tumour recurrence (IBTR) risk. Methods: Between January 2009 and December 2018, consecutive patients with DCIS and breast conserving surgery (BCS) were enrolled and randomly assigned to a training cohort (n = 181) and internally validation cohort (n = 78). Multivariate analyses were performed to identify predictors of IBTR. Model performance was evaluated by the concordance index (C-index) and calibration plot. The time-to-event curves were calculated by the Kaplan−Meier methods and compared by the log-rank test. Results: In total, 259 patients were enrolled and 182 of them received whole breast irradiation (WBI). After a median follow-up of 51.02 months, 23 IBTR events occurred in the whole cohort. By multivariate analyses of training cohort, presence of microinvasion, Ki67 index >14%, mammographic-clustered fine linear microcalcifications and neutrophil/lymphocyte ratio before BCS (preop-NLR), >1.1 remained independent risk factors of IBTR to develop a nomogram. The C-indexes of the nomogram were 0.87 and 0.86 in the training and internal validation set, respectively. Calibration plots illustrated good agreement between the predictions and actual observations for 5-year IBTR. Cut-off values of nomogram point were identified as 53 and 115 points, which divided all patients into low-, intermediate- and high-risk groups. Significant differences in IBTR existed between low-, intermediate- and high-risk subgroups (p < 0.01). For the whole cohort and ER-positive tumours, the benefit of WBI was found only in the intermediate-risk subgroup, but not in those with low or high risk. Fourteen out of 23 IBTRs occurred outside the original quadrant and all occurred in the high-risk group. Conclusions: The novel nomogram demonstrated potential to separate the risk of IBTR and locations of IBTR. For the whole cohort and ER-positive tumours, the benefit of WBI was restricted to an intermediate-risk subgroup.
Collapse
Affiliation(s)
- Feifei Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Lu Cao
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Cheng Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Gang Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Rong Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Weixiang Qi
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Shubei Wang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Weimin Chai
- Department of Radiology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
| | - Jiayi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 20025, China
- Correspondence: ; Tel.: +86-021-64370045; Fax: +86-021-64156886
| |
Collapse
|
7
|
Matić IZ, Grujić M, Kolundžija B, Damjanović A, Tomašević Z, Đorđić Crnogorac M, Džodić R, Filipović Lješković I, Ždrale Z, Erić-Nikolić A, Juranić Z. White blood cell subsets in HER2-positive breast cancer patients treated with trastuzumab in relation to clinical outcome. Pathol Res Pract 2021; 224:153543. [PMID: 34273805 DOI: 10.1016/j.prp.2021.153543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 11/27/2022]
Abstract
To examine whether HER2+ breast cancer patients who have decreased immune effector cells could respond well to trastuzumab, we evaluated the alterations in circulating immune system cell subsets: CD16+ and/or CD56+ lymphocytes, lymphocytes and granulocytes in these patients before and after treatment with trastuzumab-based regimens in relation to clinical response to therapy. The study involved 55 patients with HER2+ breast cancer before and 2 months after the initiation of the therapy. Progressive disease was confirmed in nine out of 55 patients (non-responders), while other patients achieved complete or partial response, or stable disease (responders). Control group consisted of up to 52 healthy individuals. Significantly lower percentages of total lymphocytes, CD16+, CD56+, and CD16+CD56+ lymphocytes as well as higher percentage of granulocytes and a higher ratio of granulocyte to lymphocyte percentages were found in patients before therapy and 2 months after the initiation of the therapy, compared with those in healthy individuals. Responder subgroup showed significantly lower percentages of CD16+, CD56+, and CD16+CD56+ lymphocytes before therapy, compared with those in healthy controls. Two months after the initiation of the therapy, the percentages of immune cell subsets remained significantly lower in responders in comparison with those in the healthy donors, while a significantly decreased percentages of CD56+ and CD16+CD56+ lymphocytes were observed in non-responders, in comparison with those in healthy controls. Our study demonstrated that HER2+ breast cancer patients who have decreased percentages of CD16+, CD56+, and CD16+CD56+ lymphocytes may achieve response to trastuzumab-containing treatment.
Collapse
Affiliation(s)
- Ivana Z Matić
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia.
| | - Milica Grujić
- Institute of Rheumatology, Resavska 69, 11000 Belgrade, Serbia
| | - Branka Kolundžija
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Ana Damjanović
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Zorica Tomašević
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | | | - Radan Džodić
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia; School of Medicine, University of Belgrade, dr Subotića 8, 11000 Belgrade, Serbia
| | | | - Zdravko Ždrale
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | | | - Zorica Juranić
- Institute of Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| |
Collapse
|
8
|
Lewis GD, Haque W, Farach A, Hatch SS, Butler EB, Niravath PA, Schwartz MR, Bonefas E, Teh BS. The impact of HER2-directed targeted therapy on HER2-positive DCIS of the breast. ACTA ACUST UNITED AC 2021; 26:179-187. [PMID: 34211767 DOI: 10.5603/rpor.a2021.0026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 01/30/2021] [Indexed: 12/19/2022]
Abstract
Background In invasive breast cancer, HER2 is a well-established negative prognostic factor. However, its significance on the prognosis of ductal carcinoma in situ (DCIS) of the breast is unclear. As a result, the impact of HER2-directed therapy on HER2-positive DCIS is unknown and is currently the subject of ongoing clinical trials. In this study, we aim to determine the possible impact of HER 2-directed targeted therapy on survival outcomes for HER2-positive DCIS patients. Materials and methods The National Cancer Data Base (NCDB) was used to retrieve patients with biopsy-proven DCIS diagnosed from 2004-2015. Patients were divided into two groups based on the adjuvant therapy they received: systemic HER2-directed targeted therapy or no systemic therapy. Statistics included multivariable logistic regression to determine factors predictive of receiving systemic therapy, Kaplan-Meier analysis to evaluate overall survival (OS), and Cox proportional hazards modeling to determine variables associated with OS. Results Altogether, 1927 patients met inclusion criteria; 430 (22.3%) received HER2-directed targeted therapy; 1497 (77.7%) did not. Patients who received HER2-directed targeted therapy had a higher 5-year OS compared to patients that did not (97.7% vs. 95.8%, p = 0.043). This survival benefit remained on multivariable analysis. Factors associated with worse OS on multivariable analysis included Charlson-Deyo Comorbidity Score ≥ 2 and no receipt of hormonal therapy. Conclusion In this large study evaluating HER2-positive DCIS patients, the receipt of HER2-directed targeted therapy was associated with an improvement in OS. The results of currently ongoing clinical trials are needed to confirm this finding.
Collapse
Affiliation(s)
- Gary D Lewis
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Waqar Haque
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Andrew Farach
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Sandra S Hatch
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas, United States
| | - E Brian Butler
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Polly A Niravath
- Department of Clinical Medicine in Oncology, Houston Methodist Hospital, Houston, Texas, United States
| | - Mary R Schwartz
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas, United States
| | | | - Bin S Teh
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, United States
| |
Collapse
|
9
|
Xu FF, Zheng SF, Xu C, Cai G, Wang SB, Qi WX, Wang CF, Chen JY, Lu C. Prognostic and predictive significance of tumor infiltrating lymphocytes for ductal carcinoma in situ. Oncoimmunology 2021; 10:1875637. [PMID: 33796401 PMCID: PMC7993193 DOI: 10.1080/2162402x.2021.1875637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
This study aims to identify the density of TILs in ductal carcinoma in situ (DCIS) in terms of prognostic significance with recurrence and the benefit of whole breast irradiation (WBI). The clinicopathological data of DCIS patients from Jan 2009 to Dec 2016 who received breast-conserving surgery (BCS) were retrospectively reviewed. Cox regression analysis was used to confirm independent prognostic factors of ipsilateral breast tumor recurrence (IBTR). Kaplan–Meier method was utilized to analyze IBTR and values of WBI. Touching-tumor-infiltrating lymphocytes (TILs) were defined by TILs touching or within one lymphocyte cell thickness from the malignant ducts’ basement membrane. In total, 129 patients were enrolled in this analysis with 98 patients who received WBI. After a median follow-up of 53.0 months, there were 16 IBTR events with five invasive IBTRs. Univariate and multivariate analyses showed that touching-TILs >5 were an independent prognostic factor for higher IBTR (HR = 6.17, 95%CI 1.95–19.56, p < .01). The whole cohort was classified into two subgroups: dense group (>5 touching-TILs per duct) and sparse group (≤5 touching-TILs per duct). Dense touching-TILs were associated with unfavorable biologic characteristics. The 5-y rate of IBTR between dense and sparse group was 29.0% versus 4.5% (p < .01). For the sparse group, WBI significantly reduced the rate of 5-y-IBTR risk from 13.2% to 1.7% (p = .02), but there was no benefit of WBI in the dense group. Touching-TILs density was heterogeneous in patients with DCIS. Sparse touching-TILs were associated with better prognosis and benefit from WBI. Dense touching-TILs not only were associated with a higher risk of IBTR but also lack of benefit from WBI.
Collapse
Affiliation(s)
- Fei-Fei Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sai-Fang Zheng
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cheng Xu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Gang Cai
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shu-Bei Wang
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei-Xiang Qi
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chao-Fu Wang
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jia-Yi Chen
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cao Lu
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Basen-Engquist K, Brown P, Coletta AM, Savage M, Maresso KC, Hawk E. Lifestyle and Cancer Prevention. ABELOFF'S CLINICAL ONCOLOGY 2020:337-374.e12. [DOI: 10.1016/b978-0-323-47674-4.00022-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
11
|
Estrogen Receptor-positive Ductal Carcinoma In Situ Frequently Overexpresses HER2 Protein Without Gene Amplification. Am J Surg Pathol 2019; 43:1221-1228. [DOI: 10.1097/pas.0000000000001300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
12
|
Miligy IM, Toss MS, Gorringe KL, Lee AHS, Ellis IO, Green AR, Rakha EA. The clinical and biological significance of HER2 over-expression in breast ductal carcinoma in situ: a large study from a single institution. Br J Cancer 2019; 120:1075-1082. [PMID: 31065110 PMCID: PMC6738110 DOI: 10.1038/s41416-019-0436-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Previous studies have reported up to 50% of ductal carcinoma in situ (DCIS), is HER2 positive, but the frequency of HER2-positive invasive breast cancer (IBC) is lower. The aim of this study is to characterise HER2 status in DCIS and assess its prognostic value. METHODS HER2 status was evaluated in a large series of DCIS (n = 868), including pure DCIS and DCIS associated with IBC, prepared as tissue microarrays (TMAs). HER2 status was assessed using immunohistochemistry (IHC) and chromogenic in situ hybridisation (CISH). RESULTS In pure DCIS, HER2 protein was over-expressed in 9% of DCIS (3+), whereas 15% were HER2 equivocal (2+). Using CISH, the final HER2 status was positive in 20%. In mixed DCIS, HER2 amplification of the DCIS component was detected in 15% with amplification in the invasive component of only 12%. HER2-positive DCIS was associated with features of aggressiveness (p < 0.0001) and more frequent local recurrence (p = 0.03). On multivariate analysis, combined HER2+/Ki67+ profile was an independent predictor of local recurrence (p = 0.006). CONCLUSIONS The frequency of HER2 positivity in DCIS is comparable to IBC- and HER2-positive DCIS is associated with features of poor prognosis. The majority of HER2 over-expression in DCIS is driven by gene amplification.
Collapse
Affiliation(s)
- Islam M Miligy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK.,Histopathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK.,Histopathology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, University of Melbourne, Parkville, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Andrew H S Lee
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK
| | - Ian O Ellis
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, Nottingham City Hospital, The University of Nottingham, Nottingham, UK. .,Histopathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| |
Collapse
|
13
|
Griguolo G, Pascual T, Dieci MV, Guarneri V, Prat A. Interaction of host immunity with HER2-targeted treatment and tumor heterogeneity in HER2-positive breast cancer. J Immunother Cancer 2019; 7:90. [PMID: 30922362 PMCID: PMC6439986 DOI: 10.1186/s40425-019-0548-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/27/2019] [Indexed: 12/18/2022] Open
Abstract
Growing evidence suggests a clear role of the host immune system in HER2+ breast cancer. In addition, HER2+ breast cancer is generally considered more immunogenic than hormone receptor-positive (HR+)/HER2-, and specific molecular HER2+ subgroups (e.g. HER2-enriched disease) are more immunogenic than others (e.g. Luminal A or B). From a clinical perspective, the immune system plays a relevant prognostic role in HER2+ breast cancer and contributes to the therapeutic effects of trastuzumab. However, as more HER2-targeted agents become available, a better understanding of the role played by the immune system in modulating therapy response to different agents will be needed. Furthermore, the recent introduction in oncology of immune checkpoint inhibitors capable of unleashing anti-tumor immune response opens new possibilities for therapeutic combinations in HER2+ breast cancer. Here, we review the current pre-clinical and clinical data on the interplay between the immune system and HER2+ breast cancer, focusing on different HER2-targeted treatments and the biological heterogeneity that exists within HER2+ disease. Finally, we discuss new therapeutic approaches exploiting the immune system to increase activity or revert resistance to HER2-targeted agents.
Collapse
Affiliation(s)
- Gaia Griguolo
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Tomás Pascual
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona, Spain.
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain.
| |
Collapse
|
14
|
Han MS, Khan SA. Clinical Trials for Ductal Carcinoma In Situ of the Breast. J Mammary Gland Biol Neoplasia 2018; 23:293-301. [PMID: 30206737 DOI: 10.1007/s10911-018-9413-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 09/07/2018] [Indexed: 12/26/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) of the breast is a non-obligatory precursor to invasive breast carcinoma, with a variable natural history and biological potential for progression to invasive disease. Over the past 30 years, clinical trials have applied the therapeutic principles used for invasive carcinoma to treat DCIS (surgery, with or without breast radiotherapy, and post-operative endocrine therapy), with excellent survival outcomes, and in-breast recurrence rates that range from 0.5 to 1% annually. However, half of such recurrences are again in-situ lesions, and intensive therapy is likely not necessary for all patients. Current clinical research is focused on a better characterization of the potential of individual lesions to progress to invasive disease, and to identify women who would do well with lesser treatment. Three ongoing trials in the United States and Europe randomize women to active surveillance (with or without endocrine therapy) versus usual treatment with surgery and radiotherapy. The use of pre-operative endocrine therapy has been evaluated in a recently completed trial of letrozole use in postmenopausal women with DCIS; and in on-going trials of tamoxifen, used either orally, or as a 4-hydroxytamoxifen gel formulation for application to the breast skin. This review summaries the major past and current clinical trials of DCIS, and the likely trajectories of DCIS management in the near future.
Collapse
Affiliation(s)
- Michelle S Han
- Department of Surgery, Northwestern University, 250 E. Superior Street, Suite 4-420, Chicago, IL, 60611, USA
| | - Seema A Khan
- Prentice Women's Hospital, Northwestern University, 250 E. Superior Street, Suite 4-420, Chicago, IL, 60611, USA.
| |
Collapse
|
15
|
Abstract
Ductal carcinoma in situ has been stable in incidence for a decade and has an excellent prognosis. Breast conservation therapy is safe and effective for most patients. Adjuvant whole breast radiation therapy is recommended to reduce the risk of local recurrence. Accelerated partial breast irradiation is a promising alternative to decrease toxicity and improve cosmetic results. Adjuvant hormonal therapy can reduce local recurrence, but should be used cautiously. Future directions in management include developing predictive tools for guidance for use of adjuvant therapy and selecting low-risk patients with ductal carcinoma in situ in whom surgery may be safely omitted.
Collapse
Affiliation(s)
- FangMeng Fu
- Fujian Medical University Union Hospital, 29 Xinquan Rd, DongJieKou SangQuan, Gulou Qu, Fuzhou Shi, Fujian Sheng 350001, China
| | - Richard C Gilmore
- Johns Hopkins Hospital, The Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD 21287, USA
| | - Lisa K Jacobs
- Johns Hopkins Hospital, The Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD 21287, USA.
| |
Collapse
|
16
|
Bajikar SS, Wang CC, Borten MA, Pereira EJ, Atkins KA, Janes KA. Tumor-Suppressor Inactivation of GDF11 Occurs by Precursor Sequestration in Triple-Negative Breast Cancer. Dev Cell 2017; 43:418-435.e13. [PMID: 29161592 DOI: 10.1016/j.devcel.2017.10.027] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/18/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous carcinoma in which various tumor-suppressor genes are lost by mutation, deletion, or silencing. Here we report a tumor-suppressive mode of action for growth-differentiation factor 11 (GDF11) and an unusual mechanism of its inactivation in TNBC. GDF11 promotes an epithelial, anti-invasive phenotype in 3D triple-negative cultures and intraductal xenografts by sustaining expression of E-cadherin and inhibitor of differentiation 2 (ID2). Surprisingly, clinical TNBCs retain the GDF11 locus and expression of the protein itself. GDF11 bioactivity is instead lost because of deficiencies in its convertase, proprotein convertase subtilisin/kexin type 5 (PCSK5), causing inactive GDF11 precursor to accumulate intracellularly. PCSK5 reconstitution mobilizes the latent TNBC reservoir of GDF11 in vitro and suppresses triple-negative mammary cancer metastasis to the lung of syngeneic hosts. Intracellular GDF11 retention adds to the concept of tumor-suppressor inactivation and reveals a cell-biological vulnerability for TNBCs lacking therapeutically actionable mutations.
Collapse
Affiliation(s)
- Sameer S Bajikar
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Chun-Chao Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA; Institute of Molecular Medicine & Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Michael A Borten
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Elizabeth J Pereira
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kristen A Atkins
- Department of Pathology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
17
|
Current treatment trends and the need for better predictive tools in the management of ductal carcinoma in situ of the breast. Cancer Treat Rev 2017; 55:163-172. [PMID: 28402908 DOI: 10.1016/j.ctrv.2017.03.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/21/2017] [Accepted: 03/23/2017] [Indexed: 12/14/2022]
Abstract
Ductal carcinoma in situ (DCIS) of the breast represents a group of heterogeneous non-invasive lesions the incidence of which has risen dramatically since the advent of mammography screening. In this review we summarise current treatment trends and up-to-date results from clinical trials studying surgery and adjuvant therapy alternatives, including the recent consensus on excision margin width and its role in decision-making for post-excision radiotherapy. The main challenge in the clinical management of DCIS continues to be the tailoring of treatment to individual risk, in order to avoid the over-treatment of low-risk lesions or under-treatment of DCIS with higher risk of recurring or progressing into invasion. While studies estimate that only about 40% of DCIS would become invasive if untreated, heterogeneity and complex natural history have prevented adequate identification of these higher-risk lesions. Here we discuss attempts to develop prognostic tools for the risk stratification of DCIS lesions and their limitations. Early results of a UK-wide audit of DCIS management (the Sloane Project) have also demonstrated a lack of consistency in treatment. In this review we offer up-to-date perspectives on current treatment and prediction of DCIS, highlighting the pressing clinical need for better prognostic indices. Tools integrating both clinical and histopathological factors together with molecular biomarkers may hold potential for adequate stratification of DCIS according to risk. This could help develop standardised practices for optimal management of patients with DCIS, improving clinical outcomes while providing only the amount of therapy required for each individual patient.
Collapse
|
18
|
Parham DM, Pinder SE. The impact of human epidermal growth factor receptor 2 neoadjuvant monoclonal antibody (trastuzumab) therapy in ductal carcinoma in situ of the breast. Histopathology 2017; 70:1009-1011. [PMID: 27943389 DOI: 10.1111/his.13145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- David M Parham
- Department of Pathology, Royal Bournemouth Hospital, Dorset, UK
| | - Sarah E Pinder
- Division of Cancer Studies, Kings College London, London, UK
| |
Collapse
|
19
|
Krishnamurthy A, Soundara V, Ramshankar V. Preventive and Risk Reduction Strategies for Women at High Risk of Developing Breast Cancer: a Review. Asian Pac J Cancer Prev 2017; 17:895-904. [PMID: 27039715 DOI: 10.7314/apjcp.2016.17.3.895] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Breast cancer is the most commonly diagnosed invasive cancer among women. Many factors, both genetic and non-genetic, determine a woman's risk of developing breast cancer and several breast cancer risk prediction models have been proposed. It is vitally important to risk stratify patients as there are now effective preventive strategies available. All women need to be counseled regarding healthy lifestyle recommendations to decrease breast cancer risk. As such, management of these women requires healthcare professionals to be familiar with additional risk factors so that timely recommendations can be made on surveillance/risk-reducing strategies. Breast cancer risk reduction strategies can be better understood by encouraging the women at risk to participate in clinical trials to test new strategies for decreasing the risk. This article reviews the advances in the identification of women at high risk of developing breast cancer and also reviews the strategies available for breast cancer prevention.
Collapse
|
20
|
Horimoto Y, Hayashi T, Arakawa A. Pathology of healing: what else might we look at? Cancer Med 2016; 5:3586-3587. [PMID: 27781408 PMCID: PMC5224836 DOI: 10.1002/cam4.952] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/22/2016] [Accepted: 09/23/2016] [Indexed: 01/22/2023] Open
Abstract
Several aspects of the article by Morita et al. (Cancer Medicine 5:1607‐18, 2016), examining the spontaneous healing phenomenon with reference to tumor infiltrating lymphocytes (TILs), require clarification. The concept of “healing”, which can perhaps be more accurately termed “regressive change”, remains controversial due to a lack of concrete evidence. Since regressive change is characterized by fibrosis and lymphocytes, a cancer nest that appears to lack a distinct basement membrane, surrounded only by lymphocytes, as in Morita et al's Figure 2F, should be meticulously examined because the appearance may correspond to a tumor having just completed the process of invasion. In our experience, a layer of myoepithelial cells in such foci is often difficult to detect even with immunohistochemistry. Thus, we suggest evaluating the viability of cancer cells within the nest by employing several markers, such as Ki67 and apoptotic markers, to judge whether the tumor is intraductal. It might also be useful to compare cases with versus without regressive change to elucidate the biology of such tumors. For these reasons, a tumor, floating within a pool of TILs and lacking obvious fibrous bands, might be an interesting material to examine in future studies.
Collapse
Affiliation(s)
- Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Pathology and Oncology, Juntendo University School of Medicine, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| | - Atsushi Arakawa
- Department of Human Pathology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Molecular classification of non-invasive breast lesions for personalised therapy and chemoprevention. Oncotarget 2016; 6:43244-54. [PMID: 26657114 PMCID: PMC4791229 DOI: 10.18632/oncotarget.6525] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 11/27/2015] [Indexed: 12/29/2022] Open
Abstract
Breast cancer screening has led to a dramatic increase in the detection of pre-invasive breast lesions. While mastectomy is almost guaranteed to treat the disease, more conservative approaches could be as effective if patients can be stratified based on risk of co-existing or recurrent invasive disease.Here we use a range of biomarkers to interrogate and classify purely non-invasive lesions (PNL) and those with co-existing invasive breast cancer (CEIN). Apart from Ductal Carcinoma In Situ (DCIS), relative homogeneity is observed. DCIS contained a greater spread of molecular subtypes. Interestingly, high expression of p-mTOR was observed in all PNL with lower expression in DCIS and invasive carcinoma while the opposite expression pattern was observed for TOP2A.Comparing PNL with CEIN, we have identified p53 and Ki67 as predictors of CEIN with a combined PPV and NPV of 90.48% and 43.3% respectively. Furthermore, HER2 expression showed the best concordance between DCIS and its invasive counterpart.We propose that these biomarkers can be used to improve the management of patients with pre-invasive breast lesions following further validation and clinical trials. p53 and Ki67 could be used to stratify patients into low and high-risk groups for co-existing disease. Knowledge of expression of more actionable targets such as HER2 or TOP2A can be used to design chemoprevention or neo-adjuvant strategies. Increased knowledge of the molecular profile of pre-invasive lesions can only serve to enhance our understanding of the disease and, in the era of personalised medicine, bring us closer to improving breast cancer care.
Collapse
|
22
|
Abstract
Ductal carcinoma in situ (DCIS) traditionally has been managed through various combinations of surgery, radiation, and endocrine therapy. However, concern for under- or over-treatment of DCIS has led many surgeons to question historically standardized approaches and instead begin to tailor treatment based on individual prognostic indicators. Recent and ongoing clinical trials have investigated the potential for active surveillance in DCIS, the possibility of eliminating radiation therapy (RT), and ways in which adjuvant systemic therapy may be refined. This review will summarize the current trends in the treatment of DCIS, as well as highlight the most pertinent clinical trials that are shaping management today.
Collapse
Affiliation(s)
- Katrina B Mitchell
- Breast Surgical Oncology, MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA.
| | - Henry Kuerer
- Breast Surgical Oncology, MD Anderson Cancer Center, 1400 Pressler Street, Unit 1484, Houston, TX, 77030, USA.
| |
Collapse
|
23
|
Morita M, Yamaguchi R, Tanaka M, Tse GM, Yamaguchi M, Kanomata N, Naito Y, Akiba J, Hattori S, Minami S, Eguchi S, Yano H. CD8(+) tumor-infiltrating lymphocytes contribute to spontaneous "healing" in HER2-positive ductal carcinoma in situ. Cancer Med 2016; 5:1607-18. [PMID: 27061242 PMCID: PMC4944888 DOI: 10.1002/cam4.715] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/29/2016] [Accepted: 03/01/2016] [Indexed: 11/18/2022] Open
Abstract
We evaluated the associations between tumor‐infiltrating lymphocytes (TIL) including CD8‐positive [+] lymphocytes in ductal carcinoma in situ (DCIS) and histopathologic factors, particularly spontaneous “healing” and immunohistochemical (IHC)‐based subtypes, to clarify the effects of host immune response to cancer cells proliferation during early carcinogenesis for the breast cancer. This cohort enrolled 82 DCIS patients. We examined the relationships between clinicopathologic factors including age, DCIS architecture, Van Nuys classification, grade, comedo necrosis, apocrine features, TIL, CD8+ lymphocytes, healing, estrogen receptor and HER2 positivity, and IHC‐based subtypes [luminal, luminal‐HER2, HER2‐positive, triple negative (TN)]. The results were analyzed by univariate and multivariate analyses. High numbers of TIL (high‐TIL) and healing were seen in 30.5% and 39.0% of the cohort, respectively. The distributions of luminal, luminal‐HER2, HER2 and TN subtypes were 73.2%, 9.8%, 13.4%, and 3.6%, respectively. High Van Nuys grading, high‐grade, comedo necrosis, apocrine features, high‐TIL, high CD8+ lymphocytes and healing were significantly associated with HER2‐positive (luminal‐HER2, HER2), and TN subtypes. High‐TIL was significantly associated with high‐grade, comedo necrosis, apocrine features, healing, high CD8+ lymphocytes and HER2 and TN subtypes. Healing was significantly correlated with high CD8+ lymphocytes, high‐grade, comedo necrosis, apocrine features, and HER2‐positive and TN subtypes. Logistic regression analysis revealed a strong association between healing and TIL (odds ratio: 11.72, P = 0.024). High CD8+ lymphocytes was also significantly associated with healing (odds ratio: 9.26, P = 0.009). The results of this study suggested that the spontaneous healing phenomenon might be induced by CD8+ high‐TIL associated with high‐grade, comedo necrosis, apocrine features and HER2‐positive DCIS.
Collapse
Affiliation(s)
- Michi Morita
- Department of Pathology and Laboratory Medicine, Kurume University Medical Center, Kurume, Fukuoka, Japan.,Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan.,Department of Surgery, Japan Community Health Care Organization Kurume General Hospital, Kurume, Fukuoka, Japan
| | - Rin Yamaguchi
- Department of Pathology and Laboratory Medicine, Kurume University Medical Center, Kurume, Fukuoka, Japan.,Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Maki Tanaka
- Department of Surgery, Japan Community Health Care Organization Kurume General Hospital, Kurume, Fukuoka, Japan
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Miki Yamaguchi
- Department of Surgery, Japan Community Health Care Organization Kurume General Hospital, Kurume, Fukuoka, Japan
| | - Naoki Kanomata
- Department of Pathology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Yoshiki Naito
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Satoshi Hattori
- Department of Biostatistics Center, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Shigeki Minami
- Department of Surgery, Nagasaki Harbor Medical Center City Hospital, Nagasaki, Japan
| | - Susumu Eguchi
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Hirohisa Yano
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| |
Collapse
|
24
|
Systematic Review of the Side Effects Associated With Anti-HER2-Targeted Therapies Used in the Treatment of Breast Cancer, on Behalf of the EORTC Quality of Life Group. Target Oncol 2015; 11:277-92. [DOI: 10.1007/s11523-015-0409-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
25
|
Maresso KC, Tsai KY, Brown PH, Szabo E, Lippman S, Hawk ET. Molecular cancer prevention: Current status and future directions. CA Cancer J Clin 2015; 65:345-83. [PMID: 26284997 PMCID: PMC4820069 DOI: 10.3322/caac.21287] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 05/26/2015] [Accepted: 05/28/2015] [Indexed: 12/20/2022] Open
Abstract
The heterogeneity and complexity of advanced cancers strongly support the rationale for an enhanced focus on molecular prevention as a priority strategy to reduce the burden of cancer. Molecular prevention encompasses traditional chemopreventive agents as well as vaccinations and therapeutic approaches to cancer-predisposing conditions. Despite challenges to the field, we now have refined insights into cancer etiology and early pathogenesis; successful risk assessment and new risk models; agents with broad preventive efficacy (eg, aspirin) in common chronic diseases, including cancer; and a successful track record of more than 10 agents approved by the US Food and Drug Administration for the treatment of precancerous lesions or cancer risk reduction. The development of molecular preventive agents does not differ significantly from the development of therapies for advanced cancers, yet it has unique challenges and special considerations given that it most often involves healthy or asymptomatic individuals. Agents, biomarkers, cohorts, overall design, and endpoints are key determinants of molecular preventive trials, as with therapeutic trials, although distinctions exist for each within the preventive setting. Progress in the development and evolution of molecular preventive agents has been steadier in some organ systems, such as breast and skin, than in others. In order for molecular prevention to be fully realized as an effective strategy, several challenges to the field must be addressed. Here, the authors provide a brief overview of the context for and special considerations of molecular prevention along with a discussion of the results from major randomized controlled trials.
Collapse
Affiliation(s)
- Karen Colbert Maresso
- Program Manager, Division of Cancer Prevention & Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kenneth Y Tsai
- Assistant Professor, Department of Dermatology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Powel H Brown
- Chair, Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Eva Szabo
- Chair, Lung and Upper Aerodigestive Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Scott Lippman
- Director, Moores Cancer Center, University of California, San Diego, San Diego, CA
| | - Ernest T Hawk
- Vice President and Division Head, Boone Pickens Distinguished Chair for Early Prevention of Cancer, Division of Cancer Prevention & Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
26
|
Kumar BR, Padmanabhan N, Bose G, Paneer V. A Case Report of Adenoid Cystic Carcinoma of Breast- So Close Yet So Far from Triple Negative Breast Cancer. J Clin Diagn Res 2015; 9:XD01-XD03. [PMID: 26393191 DOI: 10.7860/jcdr/2015/14283.6164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/04/2015] [Indexed: 02/02/2023]
Abstract
Adenoid cystic carcinoma (ACC) of breast is a rare tumour with a low malignant potential. Though negative for oestrogen (ER), progesterone (PR) and human epidermal growth factor receptor 2 (Her2/neu), it is different from triple negative breast cancer (TNBC); ACC has an indolent course with a good prognosis.We present a case of a 40 year old premenopausal female initially diagnosed with ductal carcinoma in situ (DCIS) on core needle biopsy. She underwent breast-conserving surgery (BCS) and her final histopathological diagnosis was ACC. She subsequently underwent adjuvant external beam radiotherapy. The patient is on follow-up for more than a year with no recurrence till date.
Collapse
Affiliation(s)
- Barath Raj Kumar
- Registrar, Department of Surgical Oncology, Apollo Speciality Hospital , Chennai, India
| | - Naveen Padmanabhan
- Registrar, Department of Surgical Oncology, Apollo Speciality Hospital , Chennai, India
| | - Gajasaraj Bose
- Registrar, Department of Surgical Oncology, Apollo Speciality Hospital , Chennai, India
| | - Venkat Paneer
- Consultant, Department of Surgical Oncology, Apollo Speciality Hospital , Chennai, India
| |
Collapse
|
27
|
Borgquist S, Zhou W, Jirström K, Amini RM, Sollie T, Sørlie T, Blomqvist C, Butt S, Wärnberg F. The prognostic role of HER2 expression in ductal breast carcinoma in situ (DCIS); a population-based cohort study. BMC Cancer 2015; 15:468. [PMID: 26062614 PMCID: PMC4464713 DOI: 10.1186/s12885-015-1479-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/02/2015] [Indexed: 12/04/2022] Open
Abstract
Background HER2 is a well-established prognostic and predictive factor in invasive breast cancer. The role of HER2 in ductal breast carcinoma in situ (DCIS) is debated and recent data have suggested that HER2 is mainly related to in situ recurrences. Our aim was to study HER2 as a prognostic factor in a large population based cohort of DCIS with long-term follow-up. Methods All 458 patients diagnosed with a primary DCIS 1986–2004 in two Swedish counties were included. Silver-enhanced in situ hybridisation (SISH) was used for detection of HER2 gene amplification and protein expression was assessed by immunohistochemistry (IHC) in tissue microarrays. HER2 positivity was defined as amplified HER2 gene and/or HER2 3+ by IHC. HER2 status in relation to new ipsilateral events (IBE) and Invasive Breast Cancer Recurrences, local or distant (IBCR) was assessed by Kaplan-Meier survival analyses and Cox proportional hazards regression models. Results Primary DCIS was screening-detected in 75.5 % of cases. Breast conserving surgery (BCS) was performed in 78.6 % of whom 44.0 % received postoperative radiotherapy. No patients received adjuvant endocrine- or chemotherapy. The majority of DCIS could be HER2 classified (N = 420 (91.7 %)); 132 HER2 positive (31 %) and 288 HER2 negative (69 %)). HER2 positivity was related to large tumor size (P = 0.002), high grade (P < 0.001) and ER- and PR negativity (P < 0.001 for both). During follow-up (mean 184 months), 106 IBCRs and 105 IBEs were identified among all 458 cases corresponding to 54 in situ and 51 invasive recurrences. Eighteen women died from breast cancer and another 114 had died from other causes. The risk of IBCR was statistically significantly lower subsequent to a HER2 positive DCIS compared to a HER2 negative DCIS, (Log-Rank P = 0.03, (HR) 0.60 (95 % CI 0.38–0.94)). Remarkably, the curves did not separate until after 10 years. In ER-stratified analyses, HER2 positive DCIS was associated with lower risk of IBCR among women with ER negative DCIS (Log-Rank P = 0.003), but not for women with ER positive DCIS. Conclusions Improved prognostic tools for DCIS patients are warranted to tailor adjuvant therapy. Here, we demonstrate that HER2 positive disease in the primary DCIS is associated with lower risk of recurrent invasive breast cancer.
Collapse
Affiliation(s)
- Signe Borgquist
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village Building 404:B3, Scheelevägen 2, SE-223 81, Lund, Sweden.
| | - Wenjing Zhou
- Department of Surgical Science, Uppsala University, Uppsala, SE-75105, Sweden.
| | - Karin Jirström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village Building 404:B3, Scheelevägen 2, SE-223 81, Lund, Sweden.
| | - Rose-Marie Amini
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| | - Thomas Sollie
- Department of Pathology, Örebro University, Örebro, Sweden.
| | - Therese Sørlie
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Montebello, 0310, Oslo, Norway.
| | - Carl Blomqvist
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
| | - Salma Butt
- Department of Surgery, Clinical Sciences, Lund University, Malmö, Sweden.
| | - Fredrik Wärnberg
- Department of Surgical Science, Uppsala University, Uppsala, SE-75105, Sweden.
| |
Collapse
|
28
|
|
29
|
Molecular phenotypes of DCIS predict overall and invasive recurrence. Ann Oncol 2015; 26:1019-1025. [DOI: 10.1093/annonc/mdv062] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 02/03/2015] [Indexed: 11/14/2022] Open
|
30
|
Curigliano G, Disalvatore D, Esposito A, Pruneri G, Lazzeroni M, Guerrieri-Gonzaga A, Luini A, Orecchia R, Goldhirsch A, Rotmensz N, Bonanni B, Viale G. Risk of subsequentin situ and invasive breast cancer in human epidermal growth factor receptor 2-positive ductal carcinomain situ. Ann Oncol 2015; 26:682-687. [DOI: 10.1093/annonc/mdv013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Serrano D, Lazzeroni M, Bonanni B. Cancer chemoprevention: Much has been done, but there is still much to do. State of the art and possible new approaches. Mol Oncol 2014; 9:1008-17. [PMID: 25556583 DOI: 10.1016/j.molonc.2014.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 12/14/2022] Open
Abstract
Over the past three decades great efforts have been made in search of cancer chemoprevention strategies. The increase in knowledge of the long process from normal to cancer cell has enabled interventions in terms of lifestyle modifications, natural compounds or drugs to block or reverse the process. Great successes have been achieved, especially for breast and colorectal cancer. However, these strategies have yet to find clinical application on a large scale. In this article we identify the achievements, the pitfalls and the next steps to be taken to improve the efficacy and applicability of chemoprevention strategies. Among the crucial key points to be implemented are educational activities for physicians to appropriately disseminate the aim and indeed the culture of chemoprevention. It is essential to improve the risk-benefit balance, seeking the minimal active doses, intermittent schedules, a better characterization of the risk categories via a more personalized intervention based on individual characteristics, and ensure the containment of costs of public and private health prevention programs.
Collapse
Affiliation(s)
- Davide Serrano
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Matteo Lazzeroni
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy.
| |
Collapse
|
32
|
Estévez LG, Suarez-Gauthier A, García E, Miró C, Calvo I, Fernández-Abad M, Herrero M, Marcos M, Márquez C, Lopez Ríos F, Perea S, Hidalgo M. Molecular effects of lapatinib in patients with HER2 positive ductal carcinoma in situ. Breast Cancer Res 2014; 16:R76. [PMID: 25186428 PMCID: PMC4448559 DOI: 10.1186/bcr3695] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 07/01/2014] [Indexed: 02/02/2023] Open
Abstract
Introduction Human epidermal growth factor receptor 2 (HER2) amplification is frequent in ductal carcinoma in situ (DCIS) of the breast and is associated with poorly differentiated tumors and adverse prognosis features. This study aimed to determine the molecular effects of the HER2 inhibitor lapatinib in patients with HER2 positive DCIS. Methods Patients with HER2 positive DCIS received 1,500 mg daily of lapatinib for four consecutive weeks prior to surgical resection. Magnetic resonance imaging (MRI) was used to determine changes in tumor volume. The molecular effects of lapatinib on HER2 signaling (PI3K/AKT and RAS/MAPK pathways), cell proliferation (Ki67 and p27) and apoptosis (TUNEL) were determined in pre and post-lapatinib treatment samples. Results A total of 20 patients were included. Lapatinib was well tolerated with only minor and transient side effects. The agent effectively modulated HER2 signaling decreasing significantly pHER2 and pERK1 expression, together with a decrease in tumor size evaluated by MRI. There was no evidence of changes in Ki67. Conclusions Four weeks of neoadjuvant lapatinib in patients with HER2-positive DCIS resulted in inhibition of HER2 and RAS/MAPK signaling pathway. Trial registration 2008-004492-21 (Registered June 25th 2008). Electronic supplementary material The online version of this article (doi:10.1186/bcr3695) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura G Estévez
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Ana Suarez-Gauthier
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Elena García
- Pathology Department, Hospital Universitario Fundación Alcorcón, Madrid, 28922, Spain.
| | - Cristina Miró
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Isabel Calvo
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - María Fernández-Abad
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Mercedes Herrero
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Manuel Marcos
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Cristina Márquez
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Fernando Lopez Ríos
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Sofía Perea
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain.
| | - Manuel Hidalgo
- Breast Cancer Program, Centro Integral Oncológico Clara Campal, C/Oña 10, Madrid, 28050, Spain. .,Clinical Research Program, Spanish National Cancer Research Center, Madrid, 28029, Spain.
| |
Collapse
|
33
|
Kim JH, Choi DH, Park W, Ahn SD, Kim SS, Ha SW, Kim K, Kim YB, Yea JW, Kang MK, Shin KH, Kim DW, Lee JH, Suh CO. Influence of boost radiotherapy in patients with ductal carcinoma in situ breast cancer: a multicenter, retrospective study in Korea (KROG 11-04). Breast Cancer Res Treat 2014; 146:341-345. [PMID: 24939061 DOI: 10.1007/s10549-014-3025-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/02/2014] [Indexed: 10/25/2022]
Abstract
To estimate the effect of boost radiotherapy on local recurrence-free survival (LRFS) in patients with ductal carcinoma in situ (DCIS) breast cancer. We included patients from nine institutions who met the following criteria: having Tis, age 18 years or older, having breast conserving surgery (BCS) and radiotherapy within 12 weeks after surgery. From 1995 through 2006, 728 patients were analyzed retrospectively by the Korean Radiation Oncology Group. All patients received whole-breast radiation therapy (WBRT) after BCS. 232 patients (31.9 %) also received boost radiation therapy (RT) (median 10 Gy). After median follow-up of 82 months, 5-year LRFS was 98.4 % and 10-year LRFS was 95.8 % for all patients. There was no statistically significant difference of LRFS between the boost and no-boost groups. Nineteen (2.6 %) patients had ipsilateral breast recurrences, including 12 of invasive recurrence and 7 DCIS. The presence of the HER2 receptor was associated with more invasive recurrences. Nine (1.2 %) patients developed contralateral breast cancer, including six invasive breast cancer and three DCIS. In the multivariate analysis, only the margin status was a significant prognostic factor for LRFS. Boost RT was not associated with further improvement of local control in DCIS after BCS and WBRT. HER2 receptor-positive patients may need further treatment with the anti-HER2 agents.
Collapse
Affiliation(s)
- Jin Hee Kim
- Department of Radiation Oncology, Dongsan Medical Center, Keimyung University School of Medicine, 56 Dalseong-Ro, Jung-Gu, Daegu, Korea,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Identifying a highly-aggressive DCIS subgroup by studying intra-individual DCIS heterogeneity among invasive breast cancer patients. PLoS One 2014; 9:e100488. [PMID: 24978026 PMCID: PMC4076240 DOI: 10.1371/journal.pone.0100488] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 05/24/2014] [Indexed: 12/21/2022] Open
Abstract
The heterogeneity among multiple ductal carcinoma in situ (DCIS) lesions within the same patient also diagnosed with invasive ductal carcinoma (IDC) has not been well evaluated, leaving research implications of intra-individual DCIS heterogeneity yet to be explored. In this study formalin-fixed paraffin embedded sections from 36 patients concurrently diagnosed with DCIS and IDC were evaluated by immunohistochemistry. Ten DCIS lesions from each patient were then randomly selected and scored. Our results showed that expression of PR, HER2, Ki-67, and p16 varied significantly within DCIS lesions from a single patient (P<0.05 for PR; P<1×10−8 for HER2, Ki-67 and p16). In addition, seventy-two percent of the individuals had heterogeneous expression of at least 2/6 markers. Importantly, by evaluating the expression of promising DCIS risk biomarkers (Ki-67, p53 and p16) among different DCIS subgroups classified by comparing DCIS molecular subtypes with those of adjacent normal terminal duct lobular units (TDLU) and IDC, our results suggest the existence of a highly-aggressive DCIS subgroup, which had the same molecular subtype as the adjacent IDC but not the same subtype as the adjacent normal TDLU. By using a systematic approach, our results clearly demonstrate that intra-individual heterogeneity in DCIS is very common in patients concurrently diagnosed with IDC. Our novel findings of a DCIS subpopulation with aggressive characteristics will provide a new paradigm for mechanistic studies of breast tumor progression and also have broad implications for prevention research as heterogeneous pre-invasive lesions are present in many other cancer types.
Collapse
|
35
|
Abstract
Preventing breast cancer is an effective strategy for reducing breast cancer deaths. The purpose of chemoprevention (also termed preventive therapy) is to reduce cancer incidence by use of natural, synthetic, or biological agents. The efficacy of tamoxifen, raloxifene, and exemestane as preventive therapy against estrogen-receptor (ER)-positive breast cancer is well established for women at increased risk for breast cancer. However, because breast cancer is a heterogeneous disease, distinct preventive approaches may be required for effective prevention of each subtype. Current research is, therefore, focused on identifying alternative mechanisms by which biologically active compounds can reduce the risk of all breast cancer subtypes including ER-negative breast cancer. Promising agents are currently being developed for prevention of HER2-positive and triple-negative breast cancer (TNBC) and include inhibitors of the ErbB family receptors, COX-2 inhibitors, metformin, retinoids, statins, poly(ADP-ribose) polymerase inhibitors, and natural compounds. This review focuses on recent progress in research to develop more effective preventive agents, in particular for prevention of ER-negative breast cancer.
Collapse
|
36
|
Carraro DM, Elias EV, Andrade VP. Ductal carcinoma in situ of the breast: morphological and molecular features implicated in progression. Biosci Rep 2014; 34:e00090. [PMID: 27919043 PMCID: PMC3894794 DOI: 10.1042/bsr20130077] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/29/2013] [Accepted: 11/20/2013] [Indexed: 12/21/2022] Open
Abstract
The spread of mammographic screening programmes around the world, including in developing countries, has substantially contributed to the diagnosis of small non-palpable lesions, which has increased the detection rate of DCIS (ductal carcinoma in situ). DCIS is heterogeneous in several ways, such as its clinical presentation, morphology and genomic profile. Excellent outcomes have been reported; however, many questions remain unanswered. For example, which patients groups are overtreated and could instead benefit from minimal intervention and which patient groups require a more traditional multidisciplinary approach. The development of a comprehensive integrated analysis that includes the radiological, morphological and genetic aspects of DCIS is necessary to answer these questions. This review focuses on discussing the significant findings about the morphological and molecular features of DCIS and its progression that have helped to uncover the biological and genetic heterogeneity of this disease. The knowledge gained in recent years might allow the development of tailored clinical management for women with DCIS in the future.
Collapse
Affiliation(s)
- Dirce M Carraro
- Laboratory of Genomics and Molecular Biology, International Center of Research, A.C. Camargo Cancer Center, São Paulo, SP 01509-900, Brazil
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo, SP 01509-900, Brazil
| | - Eliana V Elias
- Laboratory of Genomics and Molecular Biology, International Center of Research, A.C. Camargo Cancer Center, São Paulo, SP 01509-900, Brazil
| | - Victor P Andrade
- Department of Anatomical Pathology, A.C. Camargo Cancer Center, São Paulo, S.P. 01509-900, Brazil
| |
Collapse
|
37
|
Bevers TB, Brown PH, Maresso KC, Hawk ET. Cancer Prevention, Screening, and Early Detection. ABELOFF'S CLINICAL ONCOLOGY 2014:322-359.e12. [DOI: 10.1016/b978-1-4557-2865-7.00023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
38
|
Farnie G, Johnson RL, Williams KE, Clarke RB, Bundred NJ. Lapatinib inhibits stem/progenitor proliferation in preclinical in vitro models of ductal carcinoma in situ (DCIS). Cell Cycle 2013; 13:418-25. [PMID: 24247151 DOI: 10.4161/cc.27201] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Breast-conserving surgery for ductal carcinoma in situ (DCIS) is often combined with irradiation, reducing recurrence rates to 20% within 10 years; however, there is no change in overall survival. Evidence in the invasive breast indicates that breast cancer stem cells (CSCs) are radiotherapy-resistant and are capable of re-initiating a tumor recurrence; hence, targeting CSCs in high risk DCIS patient may improve survival. HER2 is overexpressed in 20% of DCIS and is known to be highly active in breast CSCs; we therefore investigated the effect of Lapatinib on DCIS CSC activity using 2 in vitro culture systems. Two DCIS cell lines DCIS.com (HER2 normal) and SUM225 (HER2 overexpressed) as well as DCIS cells from patient samples (n = 18) were cultured as mammospheres to assess CSC activity and in differentiated 3D-matrigel culture to determine effects within the non-CSCs. Mammosphere formation was reduced regardless of HER2 status, although this was more marked within the HER2-positive samples. When grown as differentiated DCIS acini in 3D-matrigel culture, Lapatinib only reduced acini size in the HER2-positive samples via decreased proliferation. Further investigation revealed lapatinib did not reduce self-renewal activity in the CSC population, but their proliferation was decreased regardless of HER2 status. In conclusion we show Lapatinib can reduce DCIS CSC activity, suggesting that the use of Lapatinib in high-risk DCIS patients has the potential to reduce recurrence and the progression of DCIS to invasive disease.
Collapse
Affiliation(s)
- Gillian Farnie
- Cancer Stem Cell Research; University of Manchester; Institute of Cancer Sciences; Manchester Academic Health Science Centre; Paterson Building; The Christie NHS Foundation Trust; Manchester, UK
| | - Rachael L Johnson
- Surgical Oncology; University Hospital of South Manchester NHS Foundation Trust; Wythenshawe Hospital; Manchester, UK
| | - Kathryn E Williams
- Cancer Stem Cell Research; University of Manchester; Institute of Cancer Sciences; Manchester Academic Health Science Centre; Paterson Building; The Christie NHS Foundation Trust; Manchester, UK; Surgical Oncology; University Hospital of South Manchester NHS Foundation Trust; Wythenshawe Hospital; Manchester, UK
| | - Robert B Clarke
- Breast Biology Group; University of Manchester; Institute of Cancer Sciences; Manchester Academic Health Science Centre; Paterson Building; The Christie NHS Foundation Trust; Manchester, UK
| | - Nigel J Bundred
- Surgical Oncology; University Hospital of South Manchester NHS Foundation Trust; Wythenshawe Hospital; Manchester, UK
| |
Collapse
|
39
|
Preliminary results of centralized HER2 testing in ductal carcinoma in situ (DCIS): NSABP B-43. Breast Cancer Res Treat 2013; 142:415-21. [PMID: 24202240 DOI: 10.1007/s10549-013-2755-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 10/24/2013] [Indexed: 12/21/2022]
Abstract
NSABP B-43 is the first prospective, randomized phase III multi-institution clinical trial targeting high-risk, HER2-positive DCIS. It compares whole breast irradiation alone with WBI given concurrently with trastuzumab in women with HER2-positive DCIS treated by lumpectomy. The primary aim is to determine if trastuzumab plus radiation will reduce in-breast tumor recurrence. HER2-positive DCIS was previously estimated at >50 %, occurring primarily in ER-negative, comedo-type DCIS of high nuclear grade. There has been no documented centralized multi-institutional HER2 analysis of DCIS. NSABP B-43 provides a unique opportunity to evaluate this in a large cohort of DCIS patients. Patients undergoing lumpectomy for DCIS without evidence of an invasive component are eligible. A central review of each patient's pure DCIS lesion is carried out by immunohistochemistry analysis. If the lesion is 2+, FISH analysis is performed. Patients whose tumors are HER2 3+ or FISH-positive are randomly assigned to receive two doses of trastuzumab during WBI or WBI alone. NSABP B-43 opened 11/9/08. As of 7/31/2013, 5,861 patients have had specimens received centrally, and 5,645 of those had analyzable blocks; 1,969 (34.9 %) were HER2 positive. A total of 1,428 patients have been accrued, 1,137 (79.6 %) of whom have follow-up information. The average follow-up time for the 1,137 patients is 23.3 months. No grade 4 or 5 toxicity has been observed. In NSABP B-43 the HER2-positive rate for pure DCIS among patients undergoing breast-preserving surgery is 34.9 %, lower than the previously reported rate. No trastuzumab-related safety signals have been observed. Interest in this trial has been robust.
Collapse
|
40
|
den Hollander P, Savage MI, Brown PH. Targeted therapy for breast cancer prevention. Front Oncol 2013; 3:250. [PMID: 24069582 PMCID: PMC3780469 DOI: 10.3389/fonc.2013.00250] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 09/09/2013] [Indexed: 12/13/2022] Open
Abstract
With a better understanding of the etiology of breast cancer, molecularly targeted drugs have been developed and are being testing for the treatment and prevention of breast cancer. Targeted drugs that inhibit the estrogen receptor (ER) or estrogen-activated pathways include the selective ER modulators (tamoxifen, raloxifene, and lasofoxifene) and aromatase inhibitors (AIs) (anastrozole, letrozole, and exemestane) have been tested in preclinical and clinical studies. Tamoxifen and raloxifene have been shown to reduce the risk of breast cancer and promising results of AIs in breast cancer trials, suggest that AIs might be even more effective in the prevention of ER-positive breast cancer. However, these agents only prevent ER-positive breast cancer. Therefore, current research is focused on identifying preventive therapies for other forms of breast cancer such as human epidermal growth factor receptor 2 (HER2)-positive and triple-negative breast cancer (TNBC, breast cancer that does express ER, progesterone receptor, or HER2). HER2-positive breast cancers are currently treated with anti-HER2 therapies including trastuzumab and lapatinib, and preclinical and clinical studies are now being conducted to test these drugs for the prevention of HER2-positive breast cancers. Several promising agents currently being tested in cancer prevention trials for the prevention of TNBC include poly(ADP-ribose) polymerase inhibitors, vitamin D, and rexinoids, both of which activate nuclear hormone receptors (the vitamin D and retinoid X receptors). This review discusses currently used breast cancer preventive drugs, and describes the progress of research striving to identify and develop more effective preventive agents for all forms of breast cancer.
Collapse
Affiliation(s)
- Petra den Hollander
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center , Houston, TX , USA
| | | | | |
Collapse
|
41
|
Siziopikou KP. Ductal carcinoma in situ of the breast: current concepts and future directions. Arch Pathol Lab Med 2013; 137:462-6. [PMID: 23544935 DOI: 10.5858/arpa.2012-0078-ra] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT In situ carcinomas of the breast constitute 15% to 30% of all newly diagnosed breast cancer cases; 80% of these in situ lesions belong to the ductal carcinoma in situ (DCIS) category. Similar to invasive breast carcinomas, DCIS is not a single disease but rather many distinct diseases with different histopathologic and molecular characteristics, a propensity to progress to invasive disease, and differential response to treatment. OBJECTIVE To review the classic pathologic parameters of clinical significance and the differential diagnosis of the DCIS lesions, present our new understanding of the importance of biomarkers, and discuss innovative approaches for targeted therapy in DCIS. DATA SOURCES Extensive review of the relevant peer-reviewed literature. CONCLUSIONS In DCIS, improved understanding of the underlying biologic pathways of tumor progression is expected to lead to more accurate classification and innovative targeted treatment approaches for the management of these lesions.
Collapse
Affiliation(s)
- Kalliopi P Siziopikou
- Breast Pathology Service, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
42
|
Mackey A, Greenup R, Hwang ES. New Treatment Paradigms for Patients with Ductal Carcinoma In Situ. CURRENT BREAST CANCER REPORTS 2013. [DOI: 10.1007/s12609-013-0109-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
43
|
Noh JM, Lee J, Choi DH, Cho EY, Huh SJ, Park W, Nam SJ, Lee JE, Kil WH. HER-2 overexpression is not associated with increased ipsilateral breast tumor recurrence in DCIS treated with breast-conserving surgery followed by radiotherapy. Breast 2013; 22:894-7. [PMID: 23643805 DOI: 10.1016/j.breast.2013.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/23/2013] [Accepted: 04/03/2013] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND We evaluated the clinical implications of human epidermal growth factor receptor (HER)-2 overexpression after adjuvant radiotherapy (RT) for ductal carcinoma in situ (DCIS). METHODS We reviewed 215 patients with DCIS who underwent breast-conserving surgery followed by RT. The association between HER-2 overexpression and ipsilateral breast tumor recurrence (IBTR) was evaluated. RESULTS HER-2 overexpression was associated with comedo-type architecture, high nuclear grade, and negative hormonal receptors. The median follow-up duration was 75 months. Sixteen patients experienced IBTR; seven as DCIS recurrence and nine as invasive recurrence. The IBTR rate was 11.4% at 10 years. There was no significant difference in IBTR according to HER-2 expression (P = 0.1764), neither in invasive nor DCIS recurrence. Time to recurrence was shorter in HER-2 positive tumors (P = 0.0697). CONCLUSION Adjuvant RT seems to counteract the negative effect of HER-2 overexpression in DCIS, while time to recurrence was relatively shorter.
Collapse
Affiliation(s)
- Jae Myoung Noh
- Department of Radiation Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul 135-710, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Masson S, Bahl A. The management of ductal carcinoma in situ: current controversies and future directions. Clin Oncol (R Coll Radiol) 2013; 25:275-82. [PMID: 23453783 DOI: 10.1016/j.clon.2013.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 11/30/2022]
Abstract
The incidence of ductal carcinoma in situ (DCIS) has increased in recent decades, primarily due to the widespread implementation of breast cancer screening. Traditionally, the management of DCIS has mirrored that of invasive breast cancer, with a focus on adequate surgical excision, breast-conserving surgery, adjuvant radiotherapy and endocrine therapy. However, an increasing understanding of the biology of this spectrum of conditions many mean that some cases may be managed more conservatively, reserving aggressive therapies for those patients at high risk of progression to invasive disease, ultimately aiming for a personalised approach based on individual risk factors. This overview highlights the key evidence behind current practice and discusses the rationale for current and future clinical trials in DCIS.
Collapse
Affiliation(s)
- S Masson
- Bristol Haematology and Oncology Centre, Bristol, UK.
| | | |
Collapse
|
45
|
Boyle DP, Mullan P, Salto-Tellez M. Molecular mapping the presence of druggable targets in preinvasive and precursor breast lesions: a comprehensive review of biomarkers related to therapeutic interventions. Biochim Biophys Acta Rev Cancer 2013; 1835:230-42. [PMID: 23403165 DOI: 10.1016/j.bbcan.2013.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 01/28/2013] [Accepted: 01/31/2013] [Indexed: 01/29/2023]
Abstract
The analysis of clinical breast samples using biomarkers is integral to current breast cancer management. Currently, a limited number of targeted therapies are standard of care in breast cancer treatment. However, these targeted therapies are only suitable for a subset of patients and resistance may occur. Strategies to prevent the occurrence of invasive lesions are required to reduce the morbidity and mortality associated with the development of cancer. In theory, application of targeted therapies to pre-invasive lesions will prevent their progression to invasive lesions with full malignant potential. The diagnostic challenge for pathologists is to make interpretative decisions on early detected pre-invasive lesions. Overall, only a small proportion of these pre-invasive lesions will progress to invasive carcinoma and morphological assessment is an imprecise and subjective means to differentiate histologically identical lesions with varying malignant potential. Therefore differential biomarker analysis in pre-invasive lesions may prevent overtreatment with surgery and provide a predictive indicator of response to therapy. There follows a review of established and emerging potential druggable targets in pre-invasive lesions and correlation with lesion morphology.
Collapse
Affiliation(s)
- David P Boyle
- Centre for Cancer Research and Cell Biology, Queen's University, Belfast BT9 7BL, UK.
| | | | | |
Collapse
|
46
|
Serce N, Gnatzy A, Steiner S, Lorenzen H, Kirfel J, Buettner R. Elevated expression of LSD1 (Lysine-specific demethylase 1) during tumour progression from pre-invasive to invasive ductal carcinoma of the breast. BMC Clin Pathol 2012; 12:13. [PMID: 22920283 PMCID: PMC3511290 DOI: 10.1186/1472-6890-12-13] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 08/13/2012] [Indexed: 02/02/2023] Open
Abstract
Background Lysine-specific demethylase1 (LSD1) is a nuclear protein which belongs to the aminooxidase-enzymes playing an important role in controlling gene expression. It has also been found highly expressed in several human malignancies including breast carcinoma. Our aim was to detect LSD1 expression also in pre-invasive neoplasias of the breast. In the current study we therefore analysed LSD1 protein expression in ductal carcinoma in situ (DCIS) in comparison to invasive ductal breast cancer (IDC). Methods Using immunohistochemistry we systematically analysed LSD1 expression in low grade DCIS (n = 27), intermediate grade DCIS (n = 30), high grade DCIS (n = 31) and in invasive ductal breast cancer (n = 32). SPSS version 18.0 was used for statistical analysis. Results LSD1 was differentially expressed in DCIS and invasive ductal breast cancer. Interestingly, LSD1 was significantly overexpressed in high grade DCIS versus low grade DCIS. Differences in LSD1 expression levels were also statistically significant between low/intermediate DCIS and invasive ductal breast carcinoma. Conclusions LSD1 is also expressed in pre-invasive neoplasias of the breast. Additionally, there is a gradual increase of LSD1 expression within tumour progression from pre-invasive DCIS to invasive ductal breast carcinoma. Therefore upregulation of LSD1 may be an early tumour promoting event.
Collapse
Affiliation(s)
- Nuran Serce
- Institute of Pathology, University of Bonn, Sigmund-Freud-Str. 25, Bonn 53127, Germany
| | - Annette Gnatzy
- Institute of Pathology, University of Bonn, Sigmund-Freud-Str. 25, Bonn 53127, Germany
| | - Susanne Steiner
- Institute of Pathology, University of Bonn, Sigmund-Freud-Str. 25, Bonn 53127, Germany
| | - Henning Lorenzen
- Institute of Medical Biometrics, Informatics and Epidemiology, University of Bonn, Sigmund-Freud-Str. 25, Bonn 53127, Germany
| | - Jutta Kirfel
- Institute of Pathology, University of Bonn, Sigmund-Freud-Str. 25, Bonn 53127, Germany
| | - Reinhard Buettner
- Institute of Pathology, University of Cologne, Kerpener Str. 62, Cologne, 50924, Germany
| |
Collapse
|
47
|
Abstract
Preventing breast cancer is possible with selective estrogen receptor (ER) modulators and aromatase inhibitors, which reduce the risk of invasive disease by up to 65% (up to 73% for ER-positive and no effect for ER-negative cancer) and the risk of preinvasive disease [ductal carcinoma in situ (DCIS)] by up to 50%. Clearly, approaches for preventing ER-negative, and increased prevention of ER-positive breast cancers would benefit public health. A growing body of work (including recent preclinical and clinical data) support targeting the HER family [epidermal growth factor receptor (EGFR), or human epidermal growth factor receptor (HER) 1 or ErbB1) and HER2, HER3, and HER4] for preventing ER-negative and possibly ER-positive breast cancer. Preclinical studies of HER family-targeting drugs in mammary neoplasia show suppression of (i) ER-negative tumors in HER2-overexpressing mouse strains, (ii) ER-negative tumors in mutant Brca1/p53(+/-) mice, and (iii) ER-positive tumors in the methylnitrosourea (MNU) rat model; tumors arising in both the MNU and mutant Brca1/p53(+/-) models lack HER2 overexpression. Clinical trials include a recent placebo-controlled phase IIb presurgical trial of the dual EGFR HER2 inhibitor lapatinib that suppressed growth of breast premalignancy [including atypical ductal hyperplasia (ADH) and DCIS] and invasive cancer in patients with early-stage, HER2-overexpressing or -amplified breast cancer. These results suggest that lapatinib can clinically suppress the progression of ADH and DCIS to invasive breast cancer, an effect previously observed in a mouse model of HER2-overexpressing, ER-negative mammary cancer. The preclinical and clinical signals provide a compelling rationale for testing HER-targeting drugs for breast cancer prevention in women at moderate-to-high risk, leading perhaps to combinations that prevent ER-negative and ER-positive breast cancer.
Collapse
Affiliation(s)
- Louise R Howe
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, USA
| | | |
Collapse
|
48
|
Sioshansi S, Huber KE, Wazer DE. The implications of breast cancer molecular phenotype for radiation oncology. Front Oncol 2011; 1:12. [PMID: 22649753 PMCID: PMC3355956 DOI: 10.3389/fonc.2011.00012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 06/12/2011] [Indexed: 12/31/2022] Open
Abstract
The identification of distinct molecular subtypes of breast cancer has advanced the understanding and treatment of breast cancer by providing insight into prognosis, patterns of recurrence, and effectiveness of therapy. The prognostic significance of molecular phenotype with regard to distant recurrences and overall survival are well established in the literature and has been readily incorporated into systemic therapy management decisions. However, despite the accumulating data suggesting similar prognostic significance for locoregional recurrence, integration of molecular phenotype into local management decision making has lagged. Although there are some conflicting reports, collectively the literature supports a low risk of local recurrence (LR) in the hormone receptor (HR) positive luminal subtypes compared to HR negative subtypes [triple negative (TN) and HER2-enriched]. The development of targeted therapies, such as trastuzumab for the treatment of HER2-enriched subtype, has been shown to mitigate the increased risk of LR. Unfortunately, no such remedy exists to address the increased risk of LR for patients with TN tumors, making it a clinical challenge for radiation oncologists. In this review we discuss the correlation between molecular subtype and LR following either breast conservation therapy or mastectomy. We also explore the possible mechanisms for increased LR in TN breast cancer and radiotherapeutic implications for this population, such as the safety of breast conservation, consideration of dose escalation, and the appropriateness of accelerated partial breast irradiation.
Collapse
Affiliation(s)
- Shirin Sioshansi
- Department of Radiation Oncology, Tufts Medical Center, Tufts University School of Medicine Boston, MA, USA
| | | | | |
Collapse
|
49
|
von Minckwitz G, Darb-Esfahani S, Loibl S, Huober J, Tesch H, Solbach C, Holms F, Eidtmann H, Dietrich K, Just M, Clemens MR, Hanusch C, Schrader I, Henschen S, Hoffmann G, Tiemann K, Diebold K, Untch M, Denkert C. Responsiveness of adjacent ductal carcinoma in situ and changes in HER2 status after neoadjuvant chemotherapy/trastuzumab treatment in early breast cancer--results from the GeparQuattro study (GBG 40). Breast Cancer Res Treat 2011; 132:863-70. [PMID: 21667238 DOI: 10.1007/s10549-011-1621-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 05/29/2011] [Indexed: 11/30/2022]
Abstract
Adjacent ductal carcinoma in situ (DCIS) is found in approximately 45% of invasive ductal carcinomas (IDC) of the breast. Pure DCIS overexpresses HER2 in approximately 45%. There is uncertainty whether adjacent DCIS impacts on the response to neoadjuvant chemotherapy and trastuzumab as well as whether HER2 expression in IDC component or adjacent DCIS changes throughout treatment. Core biopsies and surgical tissue from participants of the GeparQuattro study with HER2-positive IDC were centrally examined for the area of invasive ductal component and adjacent DCIS before and after receiving neoadjuvant anthracycline-taxane-trastuzumab containing chemotherapy. HER2 overexpression in IDC and adjacent DCIS was quantified separately by immunohistochemistry using the Ventana automated staining system. Pathological complete response (pCR) was defined as no residual invasive or non-invasive tumor tissue. Fifty-nine (37.3%) of 158 IDCs presented with adjacent DCIS at diagnosis. These tumors showed lower regression grades than pure IDC (P = 0.033). The presence of adjacent DCIS was an independent negative predictor of pCR [odds ratio 0.42 (95% CI 0.2-0.9), P = 0.027]. Adjacent DCIS area decreased from pre-treatment to surgery (r = 0.205) with 30 (50.8%) IDCs with adjacent DCIS showing complete eradication of adjacent DCIS. HER2 status of adjacent DCIS was highly correlated with HER2 status of IDC component before (r = 0.892) and after treatment (r = 0.676). Degree of HER2 overexpression of the IDC component decreased in 16 (33.3%) out of 49 patients without a pCR. These 16 IDCs showed lower RGs compared to the 33 IDCs with unchanged HER2 expression (P = 0.055). HER2-positive IDCs with adjacent DCIS is less responsive to neoadjuvant chemotherapy and trastuzumab compared to pure IDC. However, complete eradication of adjacent DCIS is frequently observed. HER2-overexpression of the invasive ductal component decreases in a subset of tumors, which showed less tumor regression.
Collapse
Affiliation(s)
- Gunter von Minckwitz
- German Breast Group, c/o GBG Forschungs GmbH, Martin-Behaim-Straße 12, 63263 Neu-Isenburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lari SA, Kuerer HM. Biological Markers in DCIS and Risk of Breast Recurrence: A Systematic Review. J Cancer 2011; 2:232-61. [PMID: 21552384 PMCID: PMC3088863 DOI: 10.7150/jca.2.232] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 04/30/2011] [Indexed: 12/12/2022] Open
Abstract
Understanding of the biology and clinical behavior of ductal carcinoma in situ (DCIS) is currently inadequate. The aim of this comprehensive review was to identify important molecular biological markers associated with DCIS and candidate markers associated with increased risk of ipsilateral recurrence after diagnosis of DCIS. A comprehensive systematic review was performed to identify studies published in the past 10 years that investigated biological markers in DCIS. To be included in this review, studies that investigated the rate of biological expression of markers had to report on at least 30 patients; studies that analyzed the recurrence risk associated with biomarker expression had to report on at least 50 patients. There were 6,252 patients altogether in our review. Biological markers evaluated included steroid receptors, proliferation markers, cell cycle regulation and apoptotic markers, angiogenesis-related proteins, epidermal growth factor receptor family receptors, extracellular matrix-related proteins, and COX-2. Although the studies in this review provide valuable preliminary information regarding the expression and prognostic significance of biomarkers in DCIS, common limitations of published studies (case-series, cohort, and case-control studies) were that they were limited to small patient cohorts in which the extent of surgery and use of radiotherapy or endocrine therapy varied from patient to patient, and variable methods of determining biomarker expression. These constraints made it difficult to interpret the absolute effect of expression of various biomarkers on risk of local recurrence. No prospective validation studies were identified. As the study of biomarkers are in their relative infancy in DCIS compared with invasive breast cancer, key significant prognostic and predictive markers associated with invasive breast cancer have not been adequately studied in DCIS. There is a critical need for prospective analyses of novel and other known breast cancer molecular markers in large cohorts of patient with DCIS to differentiate indolent from aggressive DCIS and better tailor the need and extent of current therapies.
Collapse
Affiliation(s)
- Sara A Lari
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|