Vol 27, No 3 (2022)
Review paper
Published online: 2022-03-09

open access

Page views 6358
Article views/downloads 998
Get Citation

Connect on Social Media

Connect on Social Media

Triple-negative breast cancer and radiation therapy

Jordana de Faria Bessa1, Gustavo Nader Marta23
Rep Pract Oncol Radiother 2022;27(3):545-551.

Abstract

Background: This study aimed to review specific indications of radiation therapy for triple-negative breast cancer (TNBC), and to introduce the hypothesis of TNBC as an independent predictor for postmastectomy radiation therapy (PMRT).

Materials and methods: Two reviewers independently searched two electronic databases (Pubmed and Embase), with the inclusion dates of January 2000 to December 2021, for the following terms: “mastectomy” or “breast conserving surgery” or “lumpectomy”, and “radiation” or “radiotherapy”, and “triple negative” and “recurrence”. All evidence was explored by two reviewers, then organized into a narrative review considering grades of recommendation.

Results: Patients with TNBC are candidates for breast conserving surgery (grade of recommendation B). Postoperative whole-breast irradiation must be offered following breast conserving surgery (grade of recommendation A). Do not omit postoperative radiation therapy in older patients with TNBC (grade of recommendation B). Do not use partial-breast irradiation in patients with TNBC (grade of recommendation B). Postmastectomy radiation therapy should be offered for women with T3–T4 or node- positive TNBC, for any number of positive nodes (grade of recommendation A). Radiation therapy following mastectomy might also benefit patients with T1–T2 node-negative TNBC (grade of recommendation B). For patients treated with neoadjuvant systemic therapy, radiation therapy indication is based on pretreatment features. Retrospective studies suggest that residual TNBC is sensitive to radiation therapy to optimize locoregional control (grade of recommendation C).

Conclusions: Postoperative radiation therapy should be offered for most patients with TNBC. Upcoming studies, preferably prospective randomized trials, should evaluate the indications of radiation therapy, especially in the context of novel systemic treatments.

review article

Reports of Practical Oncology and Radiotherapy

2022, Volume 27, Number 3, pages: 545–551

DOI: 10.5603/RPOR.a2022.0025

Submitted: 18.01.2021

Accepted: 16.02.2022

© 2022 Greater Poland Cancer Centre.
Published by Via Medica.
All rights reserved.

e-ISSN 2083–4640

ISSN 1507–1367

Triple-negative breast cancer and radiation therapy

Jordana de Faria Bessa1Gustavo Nader Marta23
1Breast Surgery, Hospital São Luiz, São Paulo, Brazil
2Department of Radiation Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
3Latin America Cooperative Oncology Group (LACOG), Brazil

Address for correspondence: Jordana de Faria Bessa, Rua Antonio Camardo, 1001 São Paulo – SP, Brazil 03309-060; e-mail: jordana.bessa@gmail.com

This article is available in open access under Creative Common Attribution-Non-Commercial-No Derivatives 4.0 International (CC BY-NC-ND 4.0) license, allowing to download articles and share them with others as long as they credit the authors and the publisher, but without permission to change them in any way or use them commercially

Abstract
Background: This study aimed to review specific indications of radiation therapy for triple-negative breast cancer (TNBC), and to introduce the hypothesis of TNBC as an independent predictor for postmastectomy radiation therapy (PMRT).
Materials and methods: Two reviewers independently searched two electronic databases (Pubmed and Embase), with the inclusion dates of January 2000 to December 2021, for the following terms: “mastectomy” or “breast conserving surgery” or “lumpectomy”, and “radiation” or “radiotherapy”, and “triple negative” and “recurrence”. All evidence was explored by two reviewers, then organized into a narrative review considering grades of recommendation.
Results: Patients with TNBC are candidates for breast conserving surgery (grade of recommendation B). Postoperative whole-breast irradiation must be offered following breast conserving surgery (grade of recommendation A). Do not omit postoperative radiation therapy in older patients with TNBC (grade of recommendation B). Do not use partial-breast irradiation in patients with TNBC (grade of recommendation B). Postmastectomy radiation therapy should be offered for women with T3–T4 or node- positive TNBC, for any number of positive nodes (grade of recommendation A). Radiation therapy following mastectomy might also benefit patients with T1–T2 node-negative TNBC (grade of recommendation B). For patients treated with neoadjuvant systemic therapy, radiation therapy indication is based on pretreatment features. Retrospective studies suggest that residual TNBC is sensitive to radiation therapy to optimize locoregional control (grade of recommendation C).
Conclusions: Postoperative radiation therapy should be offered for most patients with TNBC. Upcoming studies, preferably prospective randomized trials, should evaluate the indications of radiation therapy, especially in the context of novel systemic treatments.
Key words: triple negative breast neoplasms; radiotherapy; mastectomy; segmental mastectomy
Rep Pract Oncol Radiother 2022;27(3):545–551

Introduction

Triple-negative breast cancer (TNBC) is a subset of breast cancer that does not express estrogen, progesterone, or HER-2 receptors, as observed on immunohistochemistry. TNBC has worse outcomes than non-TNBC after either breast-conserving surgery (BCS) or mastectomy [1]. Unlike hormone-receptor-positive and HER-2-positive cancers, early TNBC only had modest advances in treatment over the years [2–5].

TNBC is further subdivided into basal and non-basal subtypes. Basal subtypes express cytokeratins 5/6 or epidermal growth factor receptor on immunohistochemistry [6]. Their gene expression patterns matched with those cells that originated in the basal/myoepithelial layer of the ductal epithelium [6]. Basal TNBC has less lymphocyte infiltration, which is associated with better prognosis [7–9].

In contrast, the non-basal subtype contains apocrine tumors that express androgen receptors and claudin-low tumors, with features suggestive of epithelial-to-mesenchymal transition [6]. Compared with patients with non-apocrine TNBC, those with apocrine TNBC are older and have smaller, lower-grade tumors and better survival rates [10]. Historical randomized controlled trials on breast oncology did not consider different molecular subtypes. Yet, uniform treatments for such heterogeneous diseases are no longer suitable in the era of precision, evidence-based medicine.

This study aimed to review specific indications of radiation therapy for TNBC, and to introduce the hypothesis of TNBC as an independent predictor for postmastectomy radiation therapy (PMRT).

Materials and methods

Two reviewers independently searched two electronic databases (Pubmed and Embase), with the inclusion dates of January 2000 to November 2021, for the following terms: “mastectomy” or “breast conserving surgery” or “lumpectomy”, and “radiation” or “radiotherapy”, and “triple negative” and “recurrence”. Filters were applied to retrieve only randomized or observational clinical trials and meta-analyses.

Non-comparative studies, reviews, case reports, case series, comments, editorials and studies published in other languages than English were excluded. References from included studies were also considered to construct the narrative review.

All evidence was then organized into a narrative review considering the grades of recommendation as proposed by Hadorn and modified by Harbour and Miller [11, 12].

Results

Our search retrieved 26 studies from Pubmed and 34 studies from Embase (Fig. 1). After excluding duplicated and non-comparative studies (by the prescription of radiotherapy in TNBC), our analysis was limited to 11 studies [13–24]. These are further detailed in the following narrative review.

125640.png
Figure 1. Search results of comparative studies by prescription of radiation therapy among triple-negative breast cancer (TNBC)
Breast-conserving surgery

All molecular subtypes, including TNBC, benefit from whole-breast radiation therapy following BCS [14, 15]. In TNBC, age is not determinant of radiation therapy prescription. In older patients (aged more than 65–70 years), randomized trials show that tamoxifen alone, without radiation therapy, provides low rates of recurrence without significant differences in mortality, in luminal breast cancer [25, 26]. For triple-negative, though, large population-based retrospective studies showed that radiation therapy improved survival [27–29].

A meta-analysis suggested that triple-negative tumors benefit from BCS with radiation therapy over mastectomy. However, low-quality trials, retrospective designs, wide confidence intervals (CIs), and high heterogeneity (including patients with T3/T4 and N2/N3 disease) make it unwise to state any conclusions [16].

The MA.20 trial, which addressed the role of nodal (internal mammary, supraclavicular, and axillary) irradiation following BCS for node-negative or high-risk N1 tumors, failed to show significant differences for the overall population, but a subgroup analysis suggests that estrogen-negative and/or progesterone-negative tumors benefit from nodal irradiation [30]. The impact of the molecular subtype was even more important than the number of positive nodes or location of tumor [30].

As for ultrafractionation (5 fractions) whole-breast radiation, the triple negative subtype was included in the FAST-Forward trial but represented less than 10% of the sample [31]. Yet, there is no data indicating that those patients had worse outcomes. The European Society for Radiotherapy and Oncology (ESTRO) Advisory Committee in Radiation Oncology Practice consensus states that ultrafractionation can be offered for non-nodal breast or chest wall (without reconstruction) radiation therapy regardless of the molecular subtype [32].

Patients with TNBC are not candidates for partial-breast radiation therapy. Triple negative subtype was the only independent predictor of recurrence in a prospective trial of accelerated partial-breast irradiation (APBI) using 32 Gy in 8 twice-daily sessions [18].

In ELIOT, a randomized trial, patients with TNBC who received intraoperative radiation therapy had much higher rates of ipsilateral breast tumor recurrence [33]. The American Society for Radiation Oncology (ASTRO) and ESTRO consensus states that only patients with T1 or small (non-high grade, screen-detected) DCIS are suitable for APBI, with caution for estrogen-receptor negative tumors [32, 34].

In conclusion, patients with TNBC are candidates for BCS (grade B recommendation). Therefore, postoperative whole-breast irradiation must be offered following BCS (grade A recommendation). Postoperative radiation therapy must not be omitted in older patients with TNBC (grade B recommendation), and partial-breast irradiation must not be administered to patients with TNBC (grade B recommendation).

Postmastectomy radiation therapy

Nearly 20 years ago, PMRT promoted an increase in non-breast cancer deaths, possibly related to cardiac adverse effects [35]. Most expert panels retained recommendations for PMRT in patients with a high risk of recurrence, such as those with T3/T4 disease or 4 positive nodes [36, 37]. In 2014, one large meta-analysis conducted by the EBCTCG group, including individual patient data of 8,135 women, concluded that PMRT significantly affected mortality of patients with node-positive breast cancer, including those with any extent of positive nodes [38]. The authors highlighted the safety of current radiation therapy, with tridimensional planning and better coverage of target areas. Conversely for patients with node-negative breast cancer who underwent axillary dissection, outcomes following mastectomy without radiation therapy were excellent and PMRT provided no additional benefit. As an important limitation, molecular subtype classification, axillary surgery and systemic therapy were outdated in a substantial proportion of studies, thus affecting the external validity.

One of the trialists included in the EBCTCG meta-analysis later presented results stratified by molecular subtype. At first, TNBC seemed markedly radioresistant [22]. The same was observed in a large national retrospective study [17]. However, subsequent analyses showed that resistance was specific for basal TNBC [23]. Non-basal TNBC had a great benefit from PMRT, reducing recurrence from 64% to 22% (p = 0.03). Conversely, for basal TNBC, the adoption of PMRT had no impact at all (20% and 19%, p = 0.87).

Tumor-infiltrating lymphocytes (TILs), another feature commonly seen in TNBC, is also predictive of response to PMRT [24].

Recently, a subset analysis of the BEATRICE trial, which randomized patients with operable pT1a–pT3 TNBC to adjuvant bevacizumab, suggested that patients with N1 disease who went through radiation therapy had fewer locoregional recurrences than those with mastectomy alone (4% and 9%, respectively; HR = 0.46) [13].

Only one randomized controlled trial specifically included patients with TNBC as the target population [19]. A total of 681 women with stage I–II TNBC treated with mastectomy and partial axillary dissection were randomized to receive systemic chemotherapy plus radiation therapy or chemotherapy alone. After a median follow-up time of 86.5 months, PMRT improved the overall recurrence (HR = 0.77, 95% CI: 0.72–0.98, p = 0.02) and overall survival (HR = 0.79, 95% CI: 0.74–0.97, p = 0.03). Although this study has important limitations (imbalance in the number of subjects to each arm, lack of sample size calculation, and dropout rates), this result is in accordance with the EBCTCG data for mastectomy with axillary sampling [38].

Skin-sparing mastectomy (SSM) and nipple-sparing mastectomy (NSM) are techniques that allow immediate reconstruction with implants. A meta-analysis of 11 observational studies that included 6,502 patients showed increased rates of local recurrence for NSM and no difference in overall survival among the groups; however, the authors considered that the quality of evidence was very low to make a definite conclusion [39]. Whether SSM and NSM retain the same indications for radiotherapy as simple mastectomy remains unclear. In an international survey that included 298 radiation oncologists and 252 breast surgeons, most responders affirmed that PMRT should be offered to patients with adverse prognostic features, such as young age, lymph node involvement, lymphovascular invasion, histological grade III, and triple-negative subtype [40].

The EORTC 22922 trial, although with outdated chemotherapy regimens, showed that nodal (internal mammary and medial supraclavicular) irradiation improved breast-cancer specific survival and recurrence rates for stage I–III central or medially located tumors [41]. Subgroup analyses were not provided, but recalling the MA.20 trial (following BCS) the addition of nodal irradiation might benefit estrogen-negative and progesteron-receptor negative tumors [30].

In conclusion, PMRT should be offered to women with node-positive TNBC for any number of positive nodes (grade A recommendation). In addition, one randomized controlled trial and consistent observational trials suggested that radiation therapy following mastectomy could benefit patients with T1–T2 node-negative TNBC (grade B recommendation). Following SSM and NSM, radiation therapy should be offered to patients with node-positive (grade B recommendation) and T1–T2 node-negative TNBC (grade D recommendation).

PMRT after neoadjuvant systemic therapy

Long-term results of 10 randomized trials confirm the equivalence of preoperative and postoperative chemotherapy in terms of breast cancer-specific survival and overall survival [42]. Current guidelines state that radiation therapy is based on the maximum disease stage at diagnosis (before chemotherapy) [43].

It is unknown if stage III complete responders could be spared of adjuvant radiation therapy, as well as if stage I/II non-responders, instead, should not [44]. Retrospective studies have suggested that patients with residual HER-2-positive and TNBC are sensitive to radiation therapy to optimize locoregional control [45]. Stage II–III patients, especially with the triple negative subtype, also benefit from internal mammary nodal irradiation, according to a propensity-score matching retrospective study [21].

In conclusion, patients with TNBC are candidates for preoperative chemotherapy (grade A recommendation). Following BCS, all patients should be offered adjuvant radiation therapy (grade A recommendation). Prescription of radiation therapy is based on the maximum disease stage at diagnosis (grade of recommendation D). It is unknown if patients with stage I/II TNBC and complete pathological response may forego adjuvant radiation therapy.

Discussion

Although TNBC foreshadows, in a substantial number of women, poorer prognosis than that observed for other breast cancer subtypes, the current status of available prospective and retrospective data suggests that postoperative radiation therapy has an important role and should be performed in most patients. Figure 2 represents a suggested clinical recommendation for postoperative radiation therapy based on the literature review.

125717.png
Figure 2. Clinical recommendation. RT radiation therapy; N+ positive node

Future studies, preferably prospective randomized trials, should be conducted to evaluate the indications of radiation therapy, especially in the context of novel systemic treatments for patients with TNBC.

Conflict of interest

The authors declare that they do not have any conflict of interest.

Funding

The authors declare that no funding was granted in the development of this article.

Author contribution

The authors have developed the design of this article, both engaged in article search and data extraction. Bessa made the first draft, Marta revised it critically, and both approved the final version. Both agreed to be accountable for all aspects of the work.

References

  1. Lowery AJ, Kell MR, Glynn RW, et al. Locoregional recurrence after breast cancer surgery: a systematic review by receptor phenotype. Breast Cancer Res Treat. 2012; 133(3): 831–841, doi: 10.1007/s10549-011-1891-6, indexed in Pubmed: 22147079.
  2. Thomssen C, Balic M, Harbeck N, et al. St. Gallen/Vienna 2021: A Brief Summary of the Consensus Discussion on Customizing Therapies for Women with Early Breast Cancer. Breast Care (Basel). 2021; 16(2): 135–143, doi: 10.1159/000516114, indexed in Pubmed: 34002112.
  3. Masuda N, Lee SJ, Ohtani S, et al. Adjuvant Capecitabine for Breast Cancer after Preoperative Chemotherapy. N Engl J Med. 2017; 376(22): 2147–2159, doi: 10.1056/NEJMoa1612645, indexed in Pubmed: 28564564.
  4. Schmid P, Cortes J, Pusztai L, et al. KEYNOTE-522 Investigators. Pembrolizumab for Early Triple-Negative Breast Cancer. N Engl J Med. 2020; 382(9): 810–821, doi: 10.1056/NEJMoa1910549, indexed in Pubmed: 32101663.
  5. Tutt ANJ, Garber JE, Kaufman B, et al. OlympiA Clinical Trial Steering Committee and Investigators. Adjuvant Olaparib for Patients with - or -Mutated Breast Cancer. N Engl J Med. 2021; 384(25): 2394–2405, doi: 10.1056/NEJMoa2105215, indexed in Pubmed: 34081848.
  6. Badve S, Dabbs DJ, Schnitt SJ, et al. Basal-like and triple-negative breast cancers: a critical review with an emphasis on the implications for pathologists and oncologists. Mod Pathol. 2011; 24(2): 157–167, doi: 10.1038/modpathol.2010.200, indexed in Pubmed: 21076464.
  7. Tavares MC, Sampaio CD, Lima GE, et al. A high CD8 to FOXP3 ratio in the tumor stroma and expression of PTEN in tumor cells are associated with improved survival in non-metastatic triple-negative breast carcinoma. BMC Cancer. 2021; 21(1): 901, doi: 10.1186/s12885-021-08636-4, indexed in Pubmed: 34362334.
  8. Kovács A, Stenmark Tullberg A, Werner Rönnerman E, et al. Effect of Radiotherapy After Breast-Conserving Surgery Depending on the Presence of Tumor-Infiltrating Lymphocytes: A Long-Term Follow-Up of the SweBCG91RT Randomized Trial. J Clin Oncol. 2019; 37(14): 1179–1187, doi: 10.1200/JCO.18.02157, indexed in Pubmed: 30939091.
  9. Masili-Oku SM, Almeida BG, Bacchi CE, et al. Lymphocyte-predominant triple-negative breast carcinomas in premenopausal patients: Lower expression of basal immunohistochemical markers. Breast. 2017; 31: 34–39, doi: 10.1016/j.breast.2016.10.012, indexed in Pubmed: 27810697.
  10. Saridakis A, Berger ER, Harigopal M, et al. Apocrine Breast Cancer: Unique Features of a Predominantly Triple-Negative Breast Cancer. Ann Surg Oncol. 2021; 28(10): 5610–5616, doi: 10.1245/s10434-021-10518-9, indexed in Pubmed: 34426884.
  11. Harbour R, Miller J. A new system for grading recommendations in evidence based guidelines. BMJ. 2001; 323(7308): 334–336, doi: 10.1136/bmj.323.7308.334, indexed in Pubmed: 11498496.
  12. Hadorn DC, Baker D, Hodges JS, et al. Rating the quality of evidence for clinical practice guidelines. J Clin Epidemiol. 1996; 49(7): 749–754, doi: 10.1016/0895-4356(96)00019-4, indexed in Pubmed: 8691224.
  13. Kayali M, Abi Jaoude J, Mohammed M, et al. Post-mastectomy Radiation Therapy in Triple-Negative Breast Cancer Patients: Analysis of the BEATRICE Trial. Ann Surg Oncol. 2022; 29(1): 460–466, doi: 10.1245/s10434-021-10511-2, indexed in Pubmed: 34324113.
  14. Wickberg Å, Magnuson A, Holmberg L, et al. Influence of the subtype on local recurrence risk of breast cancer with or without radiation therapy. Breast. 2018; 42: 54–60, doi: 10.1016/j.breast.2018.08.097, indexed in Pubmed: 30176553.
  15. Sjöström M, Lundstedt D, Hartman L, et al. Response to Radiotherapy After Breast-Conserving Surgery in Different Breast Cancer Subtypes in the Swedish Breast Cancer Group 91 Radiotherapy Randomized Clinical Trial. J Clin Oncol. 2017; 35(28): 3222–3229, doi: 10.1200/JCO.2017.72.7263, indexed in Pubmed: 28759347.
  16. O’Rorke MA, Murray LJ, Brand JS, et al. The value of adjuvant radiotherapy on survival and recurrence in triple-negative breast cancer: A systematic review and meta-analysis of 5507 patients. Cancer Treat Rev. 2016; 47: 12–21, doi: 10.1016/j.ctrv.2016.05.001, indexed in Pubmed: 27214603.
  17. Tseng YD, Uno H, Hughes ME, et al. Biological Subtype Predicts Risk of Locoregional Recurrence After Mastectomy and Impact of Postmastectomy Radiation in a Large National Database. Int J Radiat Oncol Biol Phys. 2015; 93(3): 622–630, doi: 10.1016/j.ijrobp.2015.07.006, indexed in Pubmed: 26461004.
  18. Pashtan IM, Recht A, Ancukiewicz M, et al. External beam accelerated partial-breast irradiation using 32 gy in 8 twice-daily fractions: 5-year results of a prospective study. Int J Radiat Oncol Biol Phys. 2012; 84(3): e271–e277, doi: 10.1016/j.ijrobp.2012.04.019, indexed in Pubmed: 22652104.
  19. Wang J, Shi M, Ling R, et al. Adjuvant chemotherapy and radiotherapy in triple-negative breast carcinoma: a prospective randomized controlled multi-center trial. Radiother Oncol. 2011; 100(2): 200–204, doi: 10.1016/j.radonc.2011.07.007, indexed in Pubmed: 21852010.
  20. Thorpe CS, DeWees TA, Corbin KS, et al. MC1635: Randomized Phase III Trial of Hypofractionated Radiotherapy to the Whole Breast After Breast Conserving Surgery. Int J Radiat Oncol Biol Phys. 2021; 111(3): S6–S7, doi: 10.1016/j.ijrobp.2021.07.048.
  21. Kim KH, Noh JM, Kim YB, et al. Does internal mammary node irradiation affect treatment outcome in clinical stage II-III breast cancer patients receiving neoadjuv ant chemotherapy? Breast Cancer Res Treat. 2015; 152(3): 589–599, doi: 10.1007/s10549-015-3505-1, indexed in Pubmed: 26202053.
  22. Kyndi M, Sørensen FB, Knudsen H, et al. Danish Breast Cancer Cooperative Group. Estrogen receptor, progesterone receptor, HER-2, and response to postmastectomy radiotherapy in high-risk breast cancer: the Danish Breast Cancer Cooperative Group. J Clin Oncol. 2008; 26(9): 1419–1426, doi: 10.1200/JCO.2007.14.5565, indexed in Pubmed: 18285604.
  23. Tramm T, Kyndi M, Myhre S, et al. Relationship between the prognostic and predictive value of the intrinsic subtypes and a validated gene profile predictive of loco-regional control and benefit from post-mastectomy radiotherapy in patients with high-risk breast cancer. Acta Oncol. 2014; 53(10): 1337–1346, doi: 10.3109/0284186X.2014.925580, indexed in Pubmed: 24957550.
  24. Tramm T, Vinter H, Vahl P, et al. Tumor-infiltrating lymphocytes predict improved overall survival after post-mastectomy radiotherapy: a study of the randomized DBCG82bc cohort. Acta Oncol. 2022; 61(2): 153–162, doi: 10.1080/0284186X.2021.1989629, indexed in Pubmed: 34705573.
  25. Kunkler IH, Williams LJ, Jack WJL, et al. PRIME II investigators. 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(3): 266–273, doi: 10.1016/S1470-2045(14)71221-5, indexed in Pubmed: 25637340.
  26. Hughes KS, Schnaper LA, Bellon JR, et al. Cancer and Leukemia Group B, Radiation Therapy Oncology Group, Eastern Cooperative Oncology Group. Lumpectomy plus tamoxifen with or without irradiation in women 70 years of age or older with early breast cancer. N Engl J Med. 2004; 351(10): 971–977, doi: 10.1056/NEJMoa040587, indexed in Pubmed: 15342805.
  27. Zhai Z, Zheng Yi, Yao J, et al. Evaluation of Adjuvant Treatments for T1 N0 M0 Triple-Negative Breast Cancer. JAMA Netw Open. 2020; 3(11): e2021881, doi: 10.1001/jamanetworkopen.2020.21881, indexed in Pubmed: 33211105.
  28. Haque W, Verma V, Hsiao KY, et al. Omission of radiation therapy following breast conservation in older (≥70 years) women with T1-2N0 triple-negative breast cancer. Breast J. 2019; 25(6): 1126–1133, doi: 10.1111/tbj.13443, indexed in Pubmed: 31273872.
  29. Algan O, Zhao YD, Herman T. Radiotherapy in Patients 70 Years and Older With Triple-Negative Breast Cancer. Clin Breast Cancer. 2016; 16(4): e99–e9e106, doi: 10.1016/j.clbc.2016.05.011, indexed in Pubmed: 27292180.
  30. Whelan TJ, Olivotto IA, Parulekar WR, et al. MA.20 Study Investigators. Regional Nodal Irradiation in Early-Stage Breast Cancer. N Engl J Med. 2015; 373(4): 307–316, doi: 10.1056/NEJMoa1415340, indexed in Pubmed: 26200977.
  31. Brunt AM, Haviland J, Wheatley D, et al. Hypofractionated breast radiotherapy for 1 week versus 3 weeks (FAST-Forward): 5-year efficacy and late normal tissue effects results from a multicentre, non-inferiority, randomised, phase 3 trial. Lancet. 2020; 395(10237): 1613–1626, doi: 10.1016/s0140-6736(20)30932-6, indexed in Pubmed: 32580883.
  32. Meattini I, Becherini C, Boersma L, et al. European Society for Radiotherapy and Oncology Advisory Committee in Radiation Oncology Practice consensus recommendations on patient selection and dose and fractionation for external beam radiotherapy in early breast cancer. Lancet Oncol. 2022; 23(1): e21–e31, doi: 10.1016/S1470-2045(21)00539-8, indexed in Pubmed: 34973228.
  33. Orecchia R, Veronesi U, Maisonneuve P, et al. Intraoperative irradiation for early breast cancer (ELIOT): long-term recurrence and survival outcomes from a single-centre, randomised, phase 3 equivalence trial. Lancet Oncol. 2021; 22(5): 597–608, doi: 10.1016/S1470-2045(21)00080-2, indexed in Pubmed: 33845035.
  34. Correa C, Harris EE, Leonardi MC, et al. Accelerated Partial Breast Irradiation: Executive summary for the update of an ASTRO Evidence-Based Consensus Statement. Pract Radiat Oncol. 2017; 7(2): 73–79, doi: 10.1016/j.prro.2016.09.007, indexed in Pubmed: 27866865.
  35. Favourable and unfavourable effects on long-term survival of radiotherapy for early breast cancer: an overview of the randomised trials. Lancet. 2000; 355(9217): 1757–1770, doi: 10.1016/s0140-6736(00)02263-7, indexed in Pubmed: 10832826.
  36. Recht A, Edge SB, Solin LJ, et al. American Society of Clinical Oncology. Postmastectomy radiotherapy: clinical practice guidelines of the American Society of Clinical Oncology. J Clin Oncol. 2001; 19(5): 1539–1569, doi: 10.1200/JCO.2001.19.5.1539, indexed in Pubmed: 11230499.
  37. Eifel P, Axelson JA, Costa J, et al. National Institutes of Health Consensus Development Conference Statement: adjuvant therapy for breast cancer, November 1-3, 2000. J Natl Cancer Inst. 2001; 93(13): 979–989, doi: 10.1093/jnci/93.13.979, indexed in Pubmed: 11438563.
  38. EBCTCG (Early Breast Cancer Trialists’ Collaborative Group). Effect of radiotherapy after mastectomy and axillary surgery on 10-year recurrence and 20-year breast cancer mortality: meta-analysis of individual patient data for 8135 women in 22 randomised trials. Lancet. 2014; 383(9935): 2127–2135, doi: 10.1016/s0140-6736(14)60488-8, indexed in Pubmed: 24656685.
  39. Janssen S, Holz-Sapra E, Rades D, et al. Nipple-sparing mastectomy in breast cancer patients: The role of adjuvant radiotherapy (Review). Oncol Lett. 2015; 9(6): 2435–2441, doi: 10.3892/ol.2015.3084, indexed in Pubmed: 26137086.
  40. Marta GN, Poortmans P, de Barros AC, et al. Multidisciplinary international survey of post-operative radiation therapy practices after nipple-sparing or skin-sparing mastectomy. Eur J Surg Oncol. 2017; 43(11): 2036–2043, doi: 10.1016/j.ejso.2017.09.014, indexed in Pubmed: 28967564.
  41. Poortmans PM, Weltens C, Fortpied C, et al. European Organisation for Research and Treatment of Cancer Radiation Oncology and Breast Cancer Groups. Internal mammary and medial supraclavicular lymph node chain irradiation in stage I-III breast cancer (EORTC 22922/10925): 15-year results of a randomised, phase 3 trial. Lancet Oncol. 2020; 21(12): 1602–1610, doi: 10.1016/S1470-2045(20)30472-1, indexed in Pubmed: 33152277.
  42. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials. Lancet Oncol. 2018; 19(1): 27–39, doi: 10.1016/S1470-2045(17)30777-5, indexed in Pubmed: 29242041.
  43. National Comprehensive Cancer Network. NCCN Guidelines Version 8.2021 Breast Cancer 2021. https://www.nccn.org/professionals/physician_gls/pdf/breast.pdf (2021 Sep 23).
  44. Bellon JR, Wong JS, Burstein HJ. Should response to preoperative chemotherapy affect radiotherapy recommendations after mastectomy for stage II breast cancer? J Clin Oncol. 2012; 30(32): 3916–3920, doi: 10.1200/JCO.2012.44.3358, indexed in Pubmed: 23032626.
  45. Caudle AS, Yu TK, Tucker SL, et al. Local-regional control according to surrogate markers of breast cancer subtypes and response to neoadjuvant chemotherapy in breast cancer patients undergoing breast conserving therapy. Breast Cancer Res. 2012; 14(3): R83, doi: 10.1186/bcr3198, indexed in Pubmed: 22621334.



Reports of Practical Oncology and Radiotherapy