BACKGROUND
The widespread adoption of screening mammography in the United States has led to an associated increase in the incidence of both ductal carcinoma in situ (DCIS; stage 0 breast cancer) and early-stage invasive breast cancer. One study has estimated that in the United States alone, the incidence of early-stage breast cancer has more than doubled in the last three decades, with this increase almost exclusively observed in women of screening age, ≥ 40 years.1 Outcomes following current treatment guidelines for stage 0 and I breast cancer are excellent; between 2009 and 2015, 62% of new breast cancer was localized to the breast at the time of diagnosis, and the 5-year mortality for these early-stage patients was 98.8%.2
Such improvements in outcome have been accompanied by important treatment-related sequelae; thus, the Breast Cancer Steering Committee of the National Cancer Institute, which provides guidance for prioritization and conduct for cooperative group trials in the United States, has declared “decreasing toxicity/treatment/costs associated with therapy with negligible clinically meaningful benefits” to be one of its highest priorities.3(p1) The notion of de-escalation has been a challenging one for both patients and physicians. However, early breast cancer, with its attendant excellent outcomes, provides the ideal setting in which to de-escalate locoregional management and continues to be a proving ground for incorporation of biomarker-driven care. For both DCIS and early-stage invasive cancer, there are important ongoing opportunities to “right-size” both surgery and radiation to reduce treatment toxicities while better understanding the trade-offs of omitting some treatment modalities. While doing so, the highest priority will be to achieve excellent oncologic outcomes and high quality of life for these patients.
PUBLISHED RANDOMIZED TRIALS FOR DCIS
Five prospective, randomized clinical trials have evaluated the role of radiation treatment after lumpectomy for patients with DCIS.4-11 In these randomized trials, radiation treatment was typically given to the whole breast, usually without a boost to the site of the primary tumor. On balance, these randomized trials have demonstrated that adding radiation treatment to the whole breast after surgical excision reduces the risk of local recurrence in the ipsilateral breast, as well as the subset of invasive local recurrence in the ipsilateral breast. Although these improvements in local recurrence with the addition of radiation treatment are large (typically by about half) and statistically significant, no associated improvement in breast cancer survival or overall survival (OS) has been found with the addition of radiation treatment after lumpectomy. However, the lack of a survival benefit should be interpreted cautiously because any potential survival benefit is an underpowered endpoint based on the number of patients in this setting.
The meta-analysis from the Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) analyzed the first four reported randomized trials of radiation treatment after lumpectomy for 3,729 women.12 The 10-year rate of local recurrence in the ipsilateral breast was reduced from 28.1% to 12.9% (P < .00001) by adding radiation treatment. Similarly, the 10-year rate of the subset of invasive local recurrence in the ipsilateral breast was reduced from 15.4% to 6.8% (P < .001). Despite these large differences, no differences were found for the 10-year rates of breast cancer mortality (3.7% v 4.1%, respectively; P > .1) or all-cause mortality (8.2% v 8.4%, respectively; P > .1).
The EBCTCG meta-analysis defined a potentially favorable subgroup of patients based on the combination of low nuclear grade, negative margins, and tumor size ≤ 2 cm. In this subset of 291 potentially favorable patients, the 10-year rate of local recurrence after lumpectomy was reduced from 30.1% without radiotherapy to 12.1% with radiotherapy (P = .002). After the EBCTCG meta-analysis, McCormick et al8,9 published the results from a randomized clinical trial of radiation treatment after lumpectomy for DCIS prospectively defined as having good risk characteristics (unicentric disease, tumor size ≤ 2.5 cm, low or intermediate nuclear grade, and negative margins ≥ 3 mm). This study was the only randomized trial specifically restricted to DCIS with favorable, low-risk clinical characteristics. The trial was coordinated by NRG (National Surgical Adjuvant Breast and Bowel Project [NSABP], Radiation Therapy Oncology Group [RTOG], and Gynecologic Oncology Group [GOG]). In the most recent results from this study (NRG/RTOG 9804 [ClinicalTrials.gov identifier: NCT00003857]; formerly RTOG 9804), adding radiation treatment reduced the 12-year rates of local recurrence from 11.4% to 2.8% (hazard ratio [HR], 0.26; P = .0001) and the subset of invasive local recurrence from 5.8% to 1.5% (HR, 0.34; P = .016). However, no differences were found for OS or disease-free survival.
Randomized clinical trials have studied the value of adjuvant endocrine treatment after lumpectomy, as well as the role of aromatase inhibitors in the postmenopausal patient.4,7,10,13-17 After lumpectomy plus radiation treatment in the NSABP B-24 study, patients were randomly assigned to receive adjuvant tamoxifen versus not receiving it.7,10,13 Adding tamoxifen reduced the 10-year risk of any breast cancer event (defined as ipsilateral plus contralateral invasive carcinoma or DCIS) only for estrogen receptor (ER)-positive DCIS (HR, 0.49; P < .001), but not for ER-negative DCIS (HR, 0.84; P = .59). In the UK/ANZ (United Kingdom, Australia, and New Zealand) study, adding tamoxifen reduced all breast cancer events for the overall group of patients (HR, 0.71; P = .002) and for the subset of patients not receiving radiotherapy (P = .002).4 However, no similar benefit for tamoxifen was seen for the subset of patients receiving radiotherapy (P = .8).
OMISSION OF RADIATION TREATMENT AFTER BREAST-CONSERVING SURGERY FOR DCIS
Using Clinical and Pathologic Characteristics to Define Low-Risk DCIS and to De-Escalate Treatment After Lumpectomy
A number of arguments support de-escalation of treatment and omitting radiotherapy after lumpectomy for DCIS. First, no randomized trial has shown a benefit for survival or distant metastatic disease by adding either radiation treatment or adjuvant endocrine therapy after lumpectomy, although recognizing that randomized studies are underpowered for these endpoints. Second, the long-term breast cancer–specific mortality rates are similar and low whether patients are treated with mastectomy, lumpectomy, or lumpectomy plus radiation treatment.18,19 Third, after an initial lumpectomy, only about half of the local recurrences in the ipsilateral breast are invasive carcinoma. Fourth, salvage treatment of local recurrence after an initial lumpectomy is often successful.
Many retrospective studies have attempted to individualize patient management and to omit radiation treatment after lumpectomy in low-risk DCIS as defined by clinical and pathologic characteristics.20-30 However, the specific clinical and pathologic characteristics used to select for low-risk DCIS vary widely across studies, and no consensus has emerged. The most common characteristics used to define low-risk DCIS include (1) tumor size, (2) negative margins of resection, (3) grade, and (4) patient age. The lack of consensus is highlighted by the National Comprehensive Cancer Network, which does not include any specific definition of “low risk” for DCIS.20
More recently, three clinical studies, including RTOG 9804, have attempted to define prospectively low-risk DCIS using clinical and pathologic characteristics (Table 1).8,9,31-33 When restricted to the patients with grade 1-2 DCIS treated without radiation treatment after lumpectomy, the 10-year rates of local recurrence were 9.8%-15.6%, which appear to be less than historical controls. In addition, an increasing use of endocrine therapy across these three studies appears to be associated with a decreasing risk of local recurrence for patients treated with lumpectomy only, without radiation treatment.
TABLE 1.
Summary of Open and Enrolling Studies Prospectively Identifying Patients With Low-Risk DCIS for Treatment Using Breast-Conservation Surgery With or Without RT
Using Molecular Profiling to Define Low-Risk DCIS and to De-Escalate Treatment After Lumpectomy
Given the successful integration of multigene molecular profiling in the management of invasive breast carcinoma, similar efforts have been made to develop and implement multigene molecular profiling in the management of DCIS, especially with the goal of de-escalation by omitting radiation treatment after lumpectomy34-40 De-escalation of therapy is further supported by the lack of improvement in survival by adding radiation treatment as well as by the small, but real, potential adverse effects of adding radiation treatment. Thus, molecular profiling has the potential to help individualize management, for example, by allowing for the individual patient with low-risk disease to weigh the relative risk-benefit of decreasing upfront treatment in exchange for a small increase in subsequent local recurrence.
The 12-gene Oncotype DX DCIS Score (also referred to as the DCIS Score) is a prognostic test that was developed to determine the 10-year risks of local recurrence and the subset of invasive local recurrence after initial lumpectomy without radiation treatment.37,39 The DCIS Score consists of seven tumor genes and five reference genes. This molecular test was developed and validated for clinical use in accordance with the rigorous criteria for tumor biomarkers as specified by Simon et al.41 Importantly, two independent validation studies for the DCIS Score were performed, highlighting the reproducibility of this molecular test.37,39
The first validation of the DCIS Score was performed using DCIS tumor specimens and clinical information from 327 patients enrolled in ECOG-ACRIN (Eastern Cooperative Oncology Group American College of Radiology Imaging Network) E5194.39 In this validation study, the 10-year risks of local recurrence were 10.6% for the low-risk group, 26.7% for the intermediate-risk group, and 25.9% for the high-risk group (P = .006). The 10-year risks for the subset of invasive local recurrence were 3.7%, 12.3%, and 19.2%, respectively (P = .003). On multivariable analysis, the DCIS Score, tumor size, and menopausal status were statistically significant for local recurrence (all P ≤ .02). Similar findings were reported by Rakovitch et al37 in a second, independent validation study using a population-based cohort of patients from Ontario, Canada.
The DCIS Score identifies a significant fraction of patients with underlying higher risk biology that is not seen using standard clinical and pathologic characteristics to select for lower risk biology. In the E5194 study, 30% (97/327) of the patients were found to have intermediate- or high-risk molecular profiling, and in the Ontario cohort, 38% (216/571).
The prognostic information from the DCIS Score has been further refined by combining the DCIS Score with the clinical and pathologic features of patient age and tumor size.36 Using this combination of factors increases the number of lower-risk patients, thus increasing the number of patients eligible for omitting radiation treatment. For example, if one judges the threshold for eligibility for omitting radiation treatment to be ≤ 10% risk of local recurrence at 10 years, then 47% of patients meet this threshold, and if ≤ 8%, then 26% meet this threshold. Given that the risk of developing a contralateral breast cancer is approximately 7%-8% at 10 years, many patients have molecular profiling that indicates similar, or even lower, risk in the ipsilateral breast.
In an analysis of the interaction of the DCIS Score with radiation treatment, Rakovitch et al38 reported that adding radiation treatment after lumpectomy reduces the 10-year rate of local recurrence by about half, regardless of the DCIS Score. For a lower risk DCIS Score, the 10-year rate of local recurrence was reduced from 10.6% without radiotherapy to 5.0% with radiotherapy, and for a higher risk DCIS Score, the respective 10-year rates were 25.4% without radiotherapy and 12.6% with radiotherapy.
The Decision Score is a second test developed for patients with DCIS.34,40 The Decision Score is derived from seven cancer-related genes combined with four clinical and pathologic factors (patient age, tumor size, margin status, and palpability). The Decision Score is then dichotomized into two groups (low risk and elevated risk).
On multivariate analysis for the overall group of patients, the Decision Score, the use of radiation treatment, and year of diagnosis were statistically significant for local recurrence. The Decision Score and the use of radiation treatment were statistically significant for the subset of invasive local recurrence. When the Decision Score was separated into low-risk and high-risk groups, the use of radiation treatment remained statistically significant for the elevated-risk group for local recurrence and for the subset of invasive local recurrence, but not for the low-risk group for local recurrence or the subset of invasive local recurrence. Based on the published findings from these various molecularly based studies, a number of prospective studies have been designed and are currently open to enrollment (Table 2). The cost-effectiveness of molecular testing for DCIS in the context of omission of radiation has been evaluated, and has not been shown to offer a value advantage from a population perspective.42 Thus, the decision of when to use molecular diagnostics remains an individual choice, specifically benefiting those patients whose treatment could be altered by this information.
TABLE 2.
Summary of Open and Enrolling Prospective Studies Using Molecularly Based Approaches for DCIS of the Breast
OMISSION OF SURGERY FOR LOW-RISK DCIS
The treatment of DCIS has been predicated on the assumption that without treatment, all DCIS would progress to invasive cancer. The excellent prognosis of patients treated for DCIS with the current treatment approach has been well documented, with 10-year breast cancer–specific survival approaching 98%.19 However, there has been increasing recognition of the variable rate and likelihood of invasive progression among different DCIS lesions. A few case reports from single institutions as well as from the Nurses’ Health study indicated that up to 50% of DCIS may progress to invasive cancer over a 30-year period.43,44 However, these reports were small, with limited data. A recent evaluation of data from the SEER registry of women diagnosed with DCIS from 1992-2014 who did not undergo surgery at the time of diagnosis indicates that the rate of invasive progression is highly variable between individuals and that the 10-year net risk of ipsilateral invasive breast cancer ranged from 15%-28%, depending on age at diagnosis and histologic features.45
Moreover, there are some data to support that some DCIS may resolve without surgery, with endocrine therapy alone. CALGB 40903 (ClinicalTrials.gov identifier: NCT01439711) was a prospective study in which postmenopausal women diagnosed with ER-positive DCIS received treatment with 6 months of preoperative letrozole. Overall, over 80% of patients exhibited an imaging response to treatment (Fig 1). More notably, however, there was a 15% rate of pathologic complete response (pCR), including a 75% rate of pCR among patients with low-grade disease.46 Such data support a more individualized approach for DCIS, which could be based on projected risk of invasive progression rather than merely the diagnosis of DCIS.
FIG 1.
Magnetic resonance imaging (MRI)–based response pattern of hormone receptor–positive ductal carcinoma in situ to preoperative letrozole therapy in CALGB 40903. Patients exhibited variable patterns of response, with almost one third of study patients demonstrating complete or near-complete imaging response to therapy by 6 months. Example of patient with near-complete response to treatment at (A) baseline, (B) 3-month, and (C) 6-month MRI timepoints.
Based on these observations, there has been growing interest in testing whether there exists a group of patients with DCIS with sufficiently low risk of invasive progression such that close surveillance, rather than surgery and/or radiation, could be reasonably considered. Since 2015, there has been activation of 3 prospective randomized trials in the United States, United Kingdom, and Europe, and one registry study in Japan for low-risk DCIS, all of which seek to determine whether an active surveillance strategy, with or without endocrine therapy, may be noninferior to usual care (Table 3).47-50 For patients on active surveillance, surgery is only undertaken if there is progression of disease. The definition of “low-risk” disease differs slightly among studies, but all studies exclude high-grade DCIS, and both the COMET (Clinical Trials.gov identifier: NCT02926911) and LORETTA trials (UMIN000028298), which include the use of endocrine therapy, require that DCIS be ER and/or progesterone receptor (PR) positive.
TABLE 3.
Summary of Open and Enrolling International Prospective Trials for Omission of Surgery in Low-Risk DCIS
The primary endpoint of these studies is either time to progression to invasive cancer or 2-, 5-, and 10-year rate of invasive progression, and the overall study designs are sufficiently similar to allow for informative meta-analyses. However, additional meaningful endpoints, such as quality of life, mastectomy-free survival, and OS, will also be evaluated to assess the quality-of-life trade-offs between these two approaches. Biospecimen collection is integrated into these trials, and it is hoped that these efforts will permit discovery of novel markers of invasive progression for DCIS.
BREAST-CONSERVING SURGERY WITHOUT RADIATION TREATMENT OF EARLY-STAGE INVASIVE BREAST CANCER
In the setting of breast-conservation treatment of early-stage invasive breast carcinoma, omitting radiotherapy for the lowest-risk patient subgroups has been an active area of clinical investigation. The search for these patient subgroups is supported by the decreasing rates of local recurrence in more contemporary groups of patients who received breast-conservation surgery. Early studies focused on using older patient age to select for low-risk subgroups for omitting radiation treatment. More recent and ongoing studies have focused on using biologic approaches to select for low-risk patient subgroups (Table 4).
TABLE 4.
Summary of Published and Enrolling Prospective De-Escalation Studies in Low-Risk Patients With Early-Stage Invasive Breast Carcinoma Treated With Breast-Conservation Surgery and Endocrine Therapy, With or Without RT
The approach to omitting radiation treatment in low-risk subgroups is based on the assumption that even though adding radiation treatment results in a small improvement in local control, there is no demonstrated improvement in other endpoints, such as OS, breast cancer–specific survival, and freedom from distant metastatic disease. However, because prospective studies, including randomized trials, used local failure as the primary study endpoint to design the respective study sample sizes, these studies were substantially underpowered for other endpoints. In the EBCTCG meta-analyses of randomized trials of radiation treatment after breast-conservation surgery for early-stage invasive breast carcinoma, four local recurrences in the breast were directly associated with one avoidable breast cancer death.51,52 Thus, even in low-risk subgroups, omitting radiation treatment is potentially associated with a small increase (eg, 1%-2%) in breast cancer deaths, which is too small to be evaluated with the numbers of patients reported to date.
The rate of local recurrence has been shown to be lower in older patients. Based on this observation, a number of randomized clinical trials as well as SEER data have evaluated the role of omitting radiation treatment after lumpectomy using patient age as a key eligibility factor, typically for patients ≥ 65 or ≥ 70 years of age.53-58 These randomized trials demonstrated a small, but statistically significant, improvement in the 5-year and 10-year rates of local recurrence associated with adding radiation treatment, but without any demonstrable impact on breast cancer–specific mortality or OS. The study with the longest reported follow-up, CALGB 9343, found no significant differences in time to mastectomy, time to distant metastasis, breast cancer–specific survival, or OS between patients randomly assigned to tamoxifen alone or tamoxifen and radiation therapy at a median of follow-up of 12.6 years.54 Importantly, when individualizing treatment options for the older patient, comorbidity and life expectancy are also key considerations.
Retrospective studies have demonstrated low rates of 10-year local recurrence (eg, ≤ 5%) after breast-conservation treatment with low-risk molecular profiling.59-65 Based on these observations, as well as the generally decreasing risk of local recurrence in contemporary studies, a number of prospective studies based on molecular profiling have been designed with the goal of potentially eliminating radiation treatment of the lowest-risk patient subgroups (Table 4). One limitation of these studies is that only one was a randomized study to evaluate the potential improvement in local recurrence associated with adding radiation treatment, even in these low-risk subgroups. Another limitation is that these studies are underpowered for events other than local control (eg, breast cancer mortality or distant metastatic disease). Regardless, these studies will begin to identify potential patient subgroups for de-escalation of radiation in the setting of breast-conservation treatment.
SELECTIVE DE-ESCALATION OF AXILLARY SURGERY FOR EARLY-STAGE INVASIVE BREAST CANCER
De-escalation of locoregional treatment of early-stage invasive cancer has also included consideration of omission of sentinel node biopsy in select individuals. Despite the low morbidity of sentinel lymph node biopsy (SLNB) compared with axillary node dissection (ALND), prospective trials have nevertheless reported up to a 10% risk of neuropathy, lymphedema, and limited shoulder mobility after SLNB, all of which are more pronounced among older women.66,67 Thus, efforts have focused on reduction of axillary surgery in those patients who are either at low risk for axillary metastases or in those for whom this information would not likely alter treatment recommendations. Although some have argued that SLNB lacks benefit in women who present with a clinically negative axilla and for whom chemotherapy would clearly be recommended (eg, T1 N0 human epidermal growth factor receptor 2–positive breast cancer), omission of SLNB has primarily been studied in women ≥ 70 years, including those with small ER-negative tumors for which chemotherapy might not be advised, even in the setting of low-volume nodal disease.
Evidence for safe omission of surgical staging of the axilla was provided by the CALGB 9343 study, a prospective trial of women ≥ 70 years of age who underwent breast-conserving surgery for stage I ER-positive disease and who were then randomly assigned to either tamoxifen and radiation or tamoxifen alone. Of interest, over 60% of women in the study did not undergo any axillary surgery. Of the 244 women who had ALND, there were no axillary recurrences, but importantly, there were also no axillary recurrences among women who had tamoxifen and radiation and only 6/200 axillary recurrences among those who received tamoxifen alone without radiation. At a median follow-up of 12.6 years, there was no difference in breast cancer survival or OS between groups.54
Two additional prospective randomized trials have also addressed outcomes after omission of axillary surgery among older women with early-stage breast cancer.68,69 Similar to CALGB 9343, both studies found a small increase in axillary recurrence in patients who did not have axillary surgery compared with those who underwent axillary surgery. In an analysis that combined the data from the two studies, there were no differences in in-breast recurrence, breast cancer–specific survival, or OS.
Among women ≥ 70 years of age in the National Cancer Database diagnosed with stage I ER-positive breast cancer between 2004 and 2010, 89% of patients had some type of axillary surgery, including 95% of patients who had mastectomy. However, even among node-positive patients, only 23% of patients 76-79 years of age and 11% of women 80-85 years of age received adjuvant chemotherapy.70
In 2016, the Society of Surgical Oncology published its “Choosing Wisely” guidelines, which included a recommendation to consider omission of SLNB in clinically node-negative women > 70 years of age with hormone receptor–positive invasive breast cancer.71 The European Society of Breast Cancer Specialists had issued similar guidance in 2012, stating that although SLNB should be considered routine in healthy older women, omission of SLNB and completion ALND might be reasonable in some older patients whose treatment recommendations and outcomes were not likely to be affected by axillary staging.72
Currently, there is only 1 active single-institution trial at Cedars Sinai Hospital (ClinicalTrials.gov identifier: NCT02564848), a single-arm registry trial studying the safety of SLNB omission in women ≥ 65 years of age with clinically node-negative T1-2 ER-positive breast cancer. There are several other trials in development using biomarkers to identify women at lowest risk. These efforts to de-escalate axillary surgery in selected patients will require multidisciplinary teamwork, including geriatric oncologists who can help provide a careful definition of physiologic age and life expectancy, to identify those patients who benefit least from axillary surgery.
In conclusion, the ultimate goal of personalized medicine is to right size therapies by using clinical criteria, emerging biomarkers, and medical evidence. Achievement of this objective will require both more and less: greater use of biologically targeted therapies as well as identifying opportunities to scale back those treatments that do not offer meaningful clinical benefit. Conceptually, the latter is more challenging than the former, because omission of certain treatments may be mistakenly confounded with either negligence or increased risk. Thus, future efforts focused on selective de-escalation of treatments will pose unique challenges and will require new tools, including innovative approaches to noninferiority trial design, thoughtful definition of more meaningful clinical endpoints (including addressing the fundamental question of how to define “clinically meaningful”), rational biomarker development, and more effective approaches to both eliciting and determining patient preferences for care. These efforts will require more creative engagement and communication with patients to realize the most patient-centered and equitable outcomes for patients with early-stage breast cancer.
ACKNOWLEDGMENT
The author and editors for this Special Series wish to acknowledge Dr. Larry Solin, a treasured teacher, mentor, and colleague. We are grateful for his invaluable contributions to advance care for patients with breast cancer and will miss his keen insight and friendship.
SUPPORT
E.S.H. was supported in part by National Institutes of Health Grants No. U2C CA17035, R01 CA170595, and R01 CA185138, as well as Congressionally Directed Medical Research Programs Breast Cancer Research Program Award BC132057, the Breast Cancer Research Foundation, and the Cancer Research UK Grand Challenge Award.
AUTHOR CONTRIBUTIONS
Conception and design: E. Shelley Hwang
Collection and assembly of data: E. Shelley Hwang
Data analysis and interpretation: E. Shelley Hwang
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
De-Escalation of Locoregional Therapy in Low-Risk Disease for DCIS and Early-Stage Invasive Cancer
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Lawrence Solin
Consulting or Advisory Role: Genomic Health
Speakers' Bureau: Genomic Health
No other potential conflicts of interest were reported.
REFERENCES
- 1.Bleyer A, Welch HG. Effect of three decades of screening mammography on breast-cancer incidence. N Engl J Med. 2012;367:1998–2005. doi: 10.1056/NEJMoa1206809. [DOI] [PubMed] [Google Scholar]
- 2. Howlader N, Noone AM, Krapcho M, et al: SEER Cancer Statistics (CSR) 1975-2016. https://seer.cancer.gov/csr/1975_2016.
- 3.National Cancer Institute 2015 Strategic Priorities: Breast Cancer Steering Committee. https://www.cancer.gov/about-nci/organization/ccct/steering-committees/nctn/breast-cancer/bcsc-2015-strategic-priorities.pdf
- 4.Cuzick J, Sestak I, Pinder SE, et al. Effect of tamoxifen and radiotherapy in women with locally excised ductal carcinoma in situ: Long-term results from the UK/ANZ DCIS trial. Lancet Oncol. 2011;12:21–29. doi: 10.1016/S1470-2045(10)70266-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Donker M, Litière S, Werutsky G, et al. Breast-conserving treatment with or without radiotherapy in ductal carcinoma in situ: 15-year recurrence rates and outcome after a recurrence, from the EORTC 10853 randomized phase III trial. J Clin Oncol. 2013;31:4054–4059. doi: 10.1200/JCO.2013.49.5077. [DOI] [PubMed] [Google Scholar]
- 6.Fisher B, Dignam J, Wolmark N, et al. Lumpectomy and radiation therapy for the treatment of intraductal breast cancer: Findings from National Surgical Adjuvant Breast and Bowel Project B-17. J Clin Oncol. 1998;16:441–452. doi: 10.1200/JCO.1998.16.2.441. [DOI] [PubMed] [Google Scholar]
- 7.Fisher B, Land S, Mamounas E, et al. Prevention of invasive breast cancer in women with ductal carcinoma in situ: An update of the National Surgical Adjuvant Breast and Bowel Project experience. Semin Oncol. 2001;28:400–418. doi: 10.1016/s0093-7754(01)90133-2. [DOI] [PubMed] [Google Scholar]
- 8. McCormick B: Randomized trial evaluating radiation following surgical excision for “good risk” DCIS: 12-year report from NRG/RTOG 9804. American Society for Radiation Oncology Annual Meeting, San Antonio, TX, October 21-24, 2018. [Google Scholar]
- 9.McCormick B, Winter K, Hudis C, et al. RTOG 9804: A prospective randomized trial for good-risk ductal carcinoma in situ comparing radiotherapy with observation. J Clin Oncol. 2015;33:709–715. doi: 10.1200/JCO.2014.57.9029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wapnir IL, Dignam JJ, Fisher B, et al. Long-term outcomes of invasive ipsilateral breast tumor recurrences after lumpectomy in NSABP B-17 and B-24 randomized clinical trials for DCIS. J Natl Cancer Inst. 2011;103:478–488. doi: 10.1093/jnci/djr027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wärnberg F, Garmo H, Emdin S, et al. Effect of radiotherapy after breast-conserving surgery for ductal carcinoma in situ: 20 years follow-up in the randomized SweDCIS Trial. J Clin Oncol. 2014;32:3613–3618. doi: 10.1200/JCO.2014.56.2595. [DOI] [PubMed] [Google Scholar]
- 12.Correa C, McGale P, Taylor C, et al. Overview of the randomized trials of radiotherapy in ductal carcinoma in situ of the breast. J Natl Cancer Inst Monogr. 2010;2010:162–177. doi: 10.1093/jncimonographs/lgq039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Allred DC, Anderson SJ, Paik S, et al. Adjuvant tamoxifen reduces subsequent breast cancer in women with estrogen receptor-positive ductal carcinoma in situ: A study based on NSABP protocol B-24. J Clin Oncol. 2012;30:1268–1273. doi: 10.1200/JCO.2010.34.0141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Forbes JF, Sestak I, Howell A, et al. Anastrozole versus tamoxifen for the prevention of locoregional and contralateral breast cancer in postmenopausal women with locally excised ductal carcinoma in situ (IBIS-II DCIS): A double-blind, randomised controlled trial. Lancet. 2016;387:866–873. doi: 10.1016/S0140-6736(15)01129-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ganz PA, Cecchini RS, Julian TB, et al. Patient-reported outcomes with anastrozole versus tamoxifen for postmenopausal patients with ductal carcinoma in situ treated with lumpectomy plus radiotherapy (NSABP B-35): A randomised, double-blind, phase 3 clinical trial. Lancet. 2016;387:857–865. doi: 10.1016/S0140-6736(15)01169-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Margolese RG, Cecchini RS, Julian TB, et al. Anastrozole versus tamoxifen in postmenopausal women with ductal carcinoma in situ undergoing lumpectomy plus radiotherapy (NSABP B-35): A randomised, double-blind, phase 3 clinical trial. Lancet. 2016;387:849–856. doi: 10.1016/S0140-6736(15)01168-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Sestak I, Smith SG, Howell A, et al. Early participant-reported symptoms as predictors of adherence to anastrozole in the International Breast Cancer Intervention Studies II. Ann Oncol. 2018;29:504–509. doi: 10.1093/annonc/mdx713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Narod SA, Iqbal J, Giannakeas V, et al. Breast cancer mortality after a diagnosis of ductal carcinoma in situ. JAMA Oncol. 2015;1:888–896. doi: 10.1001/jamaoncol.2015.2510. [DOI] [PubMed] [Google Scholar]
- 19.Worni M, Akushevich I, Greenup R, et al. Trends in treatment patterns and outcomes for ductal carcinoma in situ. J Natl Cancer Inst. 2015;107:djv263. doi: 10.1093/jnci/djv263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gradishar WJ, Anderson BO, Balassanian R, et al. Breast cancer, version 4.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2018;16:310–320. doi: 10.6004/jnccn.2018.0012. [DOI] [PubMed] [Google Scholar]
- 21.Kuerer HM, Smith BD, Chavez-MacGregor M, et al. DCIS margins and breast conservation: MD Anderson Cancer Center multidisciplinary practice guidelines and outcomes. J Cancer. 2017;8:2653–2662. doi: 10.7150/jca.20871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Moran MS, Bai HX, Harris EE, et al. ACR appropriateness criteria(®) ductal carcinoma in situ. Breast J. 2012;18:8–15. doi: 10.1111/j.1524-4741.2011.01197.x. [DOI] [PubMed] [Google Scholar]
- 23.Punglia RS, Jiang W, Lipsitz SR, et al. Clinical risk score to predict likelihood of recurrence after ductal carcinoma in situ treated with breast-conserving surgery. Breast Cancer Res Treat. 2018;167:751–759. doi: 10.1007/s10549-017-4553-5. [DOI] [PubMed] [Google Scholar]
- 24.Rudloff U, Jacks LM, Goldberg JI, et al. Nomogram for predicting the risk of local recurrence after breast-conserving surgery for ductal carcinoma in situ. J Clin Oncol. 2010;28:3762–3769. doi: 10.1200/JCO.2009.26.8847. [DOI] [PubMed] [Google Scholar]
- 25.Silverstein MJ, Lagios MD. Treatment selection for patients with ductal carcinoma in situ (DCIS) of the breast using the University of Southern California/Van Nuys (USC/VNPI) prognostic index. Breast J. 2015;21:127–132. doi: 10.1111/tbj.12368. [DOI] [PubMed] [Google Scholar]
- 26.Silverstein MJ, Lagios MD, Groshen S, et al. The influence of margin width on local control of ductal carcinoma in situ of the breast. N Engl J Med. 1999;340:1455–1461. doi: 10.1056/NEJM199905133401902. [DOI] [PubMed] [Google Scholar]
- 27.Solin LJ, Fourquet A, Vicini FA, et al. Long-term outcome after breast-conservation treatment with radiation for mammographically detected ductal carcinoma in situ of the breast. Cancer. 2005;103:1137–1146. doi: 10.1002/cncr.20886. [DOI] [PubMed] [Google Scholar]
- 28.Solin LJ, Kurtz J, Fourquet A, et al. Fifteen-year results of breast-conserving surgery and definitive breast irradiation for the treatment of ductal carcinoma in situ of the breast. J Clin Oncol. 1996;14:754–763. doi: 10.1200/JCO.1996.14.3.754. [DOI] [PubMed] [Google Scholar]
- 29.Van Zee KJ, Subhedar P, Olcese C, et al. Relationship between margin width and recurrence of ductal carcinoma in situ: Analysis of 2996 women treated with breast-conserving surgery for 30 years. Ann Surg. 2015;262:623–631. doi: 10.1097/SLA.0000000000001454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Yi M, Meric-Bernstam F, Kuerer HM, et al. Evaluation of a breast cancer nomogram for predicting risk of ipsilateral breast tumor recurrences in patients with ductal carcinoma in situ after local excision. J Clin Oncol. 2012;30:600–607. doi: 10.1200/JCO.2011.36.4976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hughes LL, Wang M, Page DL, et al. Local excision alone without irradiation for ductal carcinoma in situ of the breast: A trial of the Eastern Cooperative Oncology Group. J Clin Oncol. 2009;27:5319–5324. doi: 10.1200/JCO.2009.21.8560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Solin LJ, Gray R, Hughes LL, et al. Surgical excision without radiation for ductal carcinoma in situ of the breast: 12-year results from the ECOG-ACRIN E5194 study. J Clin Oncol. 2015;33:3938–3944. doi: 10.1200/JCO.2015.60.8588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wong JS, Chen YH, Gadd MA, et al. Eight-year update of a prospective study of wide excision alone for small low- or intermediate-grade ductal carcinoma in situ (DCIS) Breast Cancer Res Treat. 2014;143:343–350. doi: 10.1007/s10549-013-2813-6. [DOI] [PubMed] [Google Scholar]
- 34.Bremer T, Whitworth PW, Patel R, et al. A biological signature for breast ductal carcinoma in situ to predict radiotherapy benefit and assess recurrence risk. Clin Cancer Res. 2018;24:5895–5901. doi: 10.1158/1078-0432.CCR-18-0842. [DOI] [PubMed] [Google Scholar]
- 35.Lehman CD, Gatsonis C, Romanoff J, et al. Association of magnetic resonance imaging and a 12-gene expression assay with breast ductal carcinoma in situ treatment. JAMA Oncol. 2019;5:1036–1042. doi: 10.1001/jamaoncol.2018.6269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rakovitch E, Gray R, Baehner FL, et al. Refined estimates of local recurrence risks by DCIS score adjusting for clinicopathological features: A combined analysis of ECOG-ACRIN E5194 and Ontario DCIS cohort studies. Breast Cancer Res Treat. 2018;169:359–369. doi: 10.1007/s10549-018-4693-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Rakovitch E, Nofech-Mozes S, Hanna W, et al. A population-based validation study of the DCIS Score predicting recurrence risk in individuals treated by breast-conserving surgery alone. Breast Cancer Res Treat. 2015;152:389–398. doi: 10.1007/s10549-015-3464-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rakovitch E, Nofech-Mozes S, Hanna W, et al. Multigene expression assay and benefit of radiotherapy after breast conservation in ductal carcinoma in situ. J Natl Cancer Inst. 2017;109:djw256. doi: 10.1093/jnci/djw256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Solin LJ, Gray R, Baehner FL, et al. A multigene expression assay to predict local recurrence risk for ductal carcinoma in situ of the breast. J Natl Cancer Inst. 2013;105:701–710. doi: 10.1093/jnci/djt067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Warnberg F, Garmo H, Folkvaljon Y, et al: A validation of DCIS biological risk profile in a randomised study for radiation therapy with 20 year follow-up (SweDCIS). San Antonio Breast Cancer Symposium, San Antonio, Texas, December 5-9, 2017. [Google Scholar]
- 41.Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101:1446–1452. doi: 10.1093/jnci/djp335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Raldow AC, Sher D, Chen AB, et al. Cost effectiveness of the Oncotype DX DCIS Score for guiding treatment of patients with ductal carcinoma in situ. J Clin Oncol. 2016;34:3963–3968. doi: 10.1200/JCO.2016.67.8532. [DOI] [PubMed] [Google Scholar]
- 43.Collins LC, Tamimi RM, Baer HJ, et al. Outcome of patients with ductal carcinoma in situ untreated after diagnostic biopsy: Results from the Nurses’ Health Study. Cancer. 2005;103:1778–1784. doi: 10.1002/cncr.20979. [DOI] [PubMed] [Google Scholar]
- 44.Sanders ME, Schuyler PA, Dupont WD, et al. The natural history of low-grade ductal carcinoma in situ of the breast in women treated by biopsy only revealed over 30 years of long-term follow-up. Cancer. 2005;103:2481–2484. doi: 10.1002/cncr.21069. [DOI] [PubMed] [Google Scholar]
- 45.Ryser MD, Weaver DL, Zhao F, et al. Cancer outcomes in DCIS patients without locoregional treatment. J Natl Cancer Inst. 2019;111:952–960. doi: 10.1093/jnci/djy220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Hwang E, Duong S, Bedrosian I, et al: Abstract GS5-05: Primary endocrine therapy for ER-positive ductal carcinoma in situ (DCIS) CALGB 40903 (Alliance). San Antonio Breast Cancer Symposium, San Antonio, TX, December 5-9, 2017. [Google Scholar]
- 47.Elshof LE, Tryfonidis K, Slaets L, et al. Feasibility of a prospective, randomised, open-label, international multicentre, phase III, non-inferiority trial to assess the safety of active surveillance for low risk ductal carcinoma in situ - The LORD study. Eur J Cancer. 2015;51:1497–1510. doi: 10.1016/j.ejca.2015.05.008. [DOI] [PubMed] [Google Scholar]
- 48.Francis A, Thomas J, Fallowfield L, et al. Addressing overtreatment of screen detected DCIS; the LORIS trial. Eur J Cancer. 2015;51:2296–2303. doi: 10.1016/j.ejca.2015.07.017. [DOI] [PubMed] [Google Scholar]
- 49.Hwang ES, Hyslop T, Lynch T, et al. The COMET (Comparison of Operative versus Monitoring and Endocrine Therapy) trial: A phase III randomised controlled clinical trial for low-risk ductal carcinoma in situ (DCIS) BMJ Open. 2019;9:e026797. doi: 10.1136/bmjopen-2018-026797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kanbayashi C, Iwata H. Current approach and future perspective for ductal carcinoma in situ of the breast. Jpn J Clin Oncol. 2017;47:671–677. doi: 10.1093/jjco/hyx059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Clarke M, Collins R, Darby S, et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: An overview of the randomised trials. Lancet. 2005;366:2087–2106. doi: 10.1016/S0140-6736(05)67887-7. [DOI] [PubMed] [Google Scholar]
- 52.Darby S, McGale P, Correa C, et al. Effect of radiotherapy after breast-conserving surgery on 10-year recurrence and 15-year breast cancer death: Meta-analysis of individual patient data for 10,801 women in 17 randomised trials. Lancet. 2011;378:1707–1716. doi: 10.1016/S0140-6736(11)61629-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fyles AW, McCready DR, Manchul LA, et al. Tamoxifen with or without breast irradiation in women 50 years of age or older with early breast cancer. N Engl J Med. 2004;351:963–970. doi: 10.1056/NEJMoa040595. [DOI] [PubMed] [Google Scholar]
- 54.Hughes KS, Schnaper LA, Bellon JR, et al. Lumpectomy plus tamoxifen with or without irradiation in women age 70 years or older with early breast cancer: Long-term follow-up of CALGB 9343. J Clin Oncol. 2013;31:2382–2387. doi: 10.1200/JCO.2012.45.2615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kunkler IH, Williams LJ, Jack WJ, et al. Breast-conserving surgery with or without irradiation in women aged 65 years or older with early breast cancer (PRIME II): A randomised controlled trial. Lancet Oncol. 2015;16:266–273. doi: 10.1016/S1470-2045(14)71221-5. [DOI] [PubMed] [Google Scholar]
- 56.Smith BD, Gross CP, Smith GL, et al. Effectiveness of radiation therapy for older women with early breast cancer. J Natl Cancer Inst. 2006;98:681–690. doi: 10.1093/jnci/djj186. [DOI] [PubMed] [Google Scholar]
- 57.Blamey RW, Bates T, Chetty U, et al. Radiotherapy or tamoxifen after conserving surgery for breast cancers of excellent prognosis: British Association of Surgical Oncology (BASO) II trial. Eur J Cancer. 2013;49:2294–2302. doi: 10.1016/j.ejca.2013.02.031. [DOI] [PubMed] [Google Scholar]
- 58.Pötter R, Gnant M, Kwasny W, et al. Lumpectomy plus tamoxifen or anastrozole with or without whole breast irradiation in women with favorable early breast cancer. Int J Radiat Oncol Biol Phys. 2007;68:334–340. doi: 10.1016/j.ijrobp.2006.12.045. [DOI] [PubMed] [Google Scholar]
- 59. doi: 10.1007/s10549-014-3188-z. Drukker CA, Elias SG, Nijenhuis MV, et al: Gene expression profiling to predict the risk of locoregional recurrence in breast cancer: A pooled analysis. Breast Cancer Res Treat 148:599-613, 2014 [Erratum: Breast Cancer Res Treat 149:567, 2015] [DOI] [PubMed] [Google Scholar]
- 60.Horton JK, Jagsi R, Woodward WA, et al. Breast cancer biology: Clinical implications for breast radiation therapy. Int J Radiat Oncol Biol Phys. 2018;100:23–37. doi: 10.1016/j.ijrobp.2017.08.025. [DOI] [PubMed] [Google Scholar]
- 61.Kirwan CC, Coles CE, Bliss J, et al. It’s PRIMETIME. Postoperative avoidance of radiotherapy: Biomarker selection of women at very low risk of local recurrence. Clin Oncol (R Coll Radiol) 2016;28:594–596. doi: 10.1016/j.clon.2016.06.007. [DOI] [PubMed] [Google Scholar]
- 62.Liu FF, Shi W, Done SJ, et al. Identification of a low-risk luminal A breast cancer cohort that may not benefit from breast radiotherapy. J Clin Oncol. 2015;33:2035–2040. doi: 10.1200/JCO.2014.57.7999. [DOI] [PubMed] [Google Scholar]
- 63.Mamounas EP, Liu Q, Paik S, et al. 21-gene recurrence score and locoregional recurrence in node-positive/ER-positive breast cancer treated with chemo-endocrine therapy. J Natl Cancer Inst. 2017;109:djw25. doi: 10.1093/jnci/djw259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mamounas EP, Tang G, Fisher B, et al. Association between the 21-gene recurrence score assay and risk of locoregional recurrence in node-negative, estrogen receptor-positive breast cancer: Results from NSABP B-14 and NSABP B-20. J Clin Oncol. 2010;28:1677–1683. doi: 10.1200/JCO.2009.23.7610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Solin LJ, Gray R, Goldstein LJ, et al. Prognostic value of biologic subtype and the 21-gene recurrence score relative to local recurrence after breast conservation treatment with radiation for early stage breast carcinoma: Results from the Eastern Cooperative Oncology Group E2197 study. Breast Cancer Res Treat. 2012;134:683–692. doi: 10.1007/s10549-012-2072-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Krag DN, Anderson SJ, Julian TB, et al. Sentinel-lymph-node resection compared with conventional axillary-lymph-node dissection in clinically node-negative patients with breast cancer: Overall survival findings from the NSABP B-32 randomised phase 3 trial. Lancet Oncol. 2010;11:927–933. doi: 10.1016/S1470-2045(10)70207-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Mansel RE, Fallowfield L, Kissin M, et al. Randomized multicenter trial of sentinel node biopsy versus standard axillary treatment in operable breast cancer: The ALMANAC Trial. J Natl Cancer Inst. 2006;98:599–609. doi: 10.1093/jnci/djj158. [DOI] [PubMed] [Google Scholar]
- 68.Martelli G, Boracchi P, Ardoino I, et al. Axillary dissection versus no axillary dissection in older patients with T1N0 breast cancer: 15-year results of a randomized controlled trial. Ann Surg. 2012;256:920–924. doi: 10.1097/SLA.0b013e31827660a8. [DOI] [PubMed] [Google Scholar]
- 69.Rudenstam CM, Zahrieh D, Forbes JF, et al. Randomized trial comparing axillary clearance versus no axillary clearance in older patients with breast cancer: First results of International Breast Cancer Study Group Trial 10-93. J Clin Oncol. 2006;24:337–344. doi: 10.1200/JCO.2005.01.5784. [DOI] [PubMed] [Google Scholar]
- 70.Pesce C, Czechura T, Winchester DJ, et al. Axillary surgery among estrogen receptor positive women 70 years of age or older with clinical stage I breast cancer, 2004-2010: A report from the National Cancer Data Base. Ann Surg Oncol. 2013;20:3259–3265. doi: 10.1245/s10434-013-3153-9. [DOI] [PubMed] [Google Scholar]
- 71.Society of Surgical Oncology Choosing Wisely. https://www.choosingwisely.org/
- 72.Biganzoli L, Wildiers H, Oakman C, et al. Management of elderly patients with breast cancer: Updated recommendations of the International Society of Geriatric Oncology (SIOG) and European Society of Breast Cancer Specialists (EUSOMA) Lancet Oncol. 2012;13:e148–e160. doi: 10.1016/S1470-2045(11)70383-7. [DOI] [PubMed] [Google Scholar]
- 73. doi: 10.1007/s11912-019-0777-3. Solin LJ: Management of ductal carcinoma in situ (DCIS) of the breast: Present approaches and future directions. Curr Oncol Rep 21:33, 2019. [DOI] [PubMed] [Google Scholar]





