Next Article in Journal
Association between Immunosuppressive Therapy Utilized in the Treatment of Autoimmune Disease or Transplant and Cancer Progression
Next Article in Special Issue
Comparing the Biology of Young versus Old Age Estrogen-Receptor-Positive Breast Cancer through Gene and Protein Expression Analyses
Previous Article in Journal
A Novel TLR4-SYK Interaction Axis Plays an Essential Role in the Innate Immunity Response in Bovine Mammary Epithelial Cells
Previous Article in Special Issue
The Role of Matrix Metalloproteinase Single-Nucleotide Polymorphisms in the Clinicopathological Properties of Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Prognostic and Predictive Value of Genomic Assays in Guiding Adjuvant Breast Radiation Therapy

1
Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
2
Department of Radiation Oncology, The University of Kansas Medical Center, Kansas City, KS 66160, USA
3
Department of Radiation Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2023, 11(1), 98; https://doi.org/10.3390/biomedicines11010098
Submission received: 16 November 2022 / Revised: 16 December 2022 / Accepted: 21 December 2022 / Published: 30 December 2022

Abstract

:
Many patients with non-metastatic breast cancer benefit from adjuvant radiation therapy after lumpectomy or mastectomy on the basis of many randomized trials. However, there are many patients that have such low risks of recurrence after surgery that de-intensification of therapy by either reducing the treatment volume or omitting radiation altogether may be appropriate options. On the other hand, dose intensification may be necessary for more aggressive breast cancers. Until recently, these treatment decisions were based solely on clinicopathologic factors. Here, we review the current literature on the role of genomic assays as prognostic and/or predictive biomarkers to help guide adjuvant radiation therapy decision-making.

1. Breast Cancer and Breast Conservation Therapy

Breast conservation therapy (BCT) consisting of a lumpectomy followed by radiation therapy was first shown in the 1980’s to have equivalent outcomes to mastectomy for early stage invasive breast cancers [1,2,3,4,5,6]. Breast radiation after breast conservation surgery improves local recurrence rates and overall survival at 15 years [1,2]. BCT has therefore become a standard treatment paradigm for treating invasive breast cancers [1,7,8]. Current treatment guidelines recommend radiation therapy for most patients having breast conservation surgery.
While adjuvant radiation has therapeutic advantages, daily radiation treatments can be inconvenient, costly and may involve toxicity to the heart, lungs, skin and adverse cosmesis [7,8,9]. For these reasons, there has been a trend toward decreasing the burden and inconvenience of daily radiation treatments among low-risk breast cancer patients (defined as post-menopausal women with lymph node negative, estrogen receptor (ER) positive cancers of small size < 2 cm and negative margins). Multiple large randomized studies have shown that reducing radiation treatment volumes with accelerated partial breast irradiation (APBI) [10,11,12,13,14] or reducing overall treatment times with an ultrahypofractionated approach [15,16] does not compromise cancer control compared to conventional whole breast radiation in select low risk breast cancer subpopulations.
The identification of low risk breast cancers in which radiation may be safely omitted and the side effects of radiation can be spared is an area of investigation. Historical clinical trials omitting radiation therapy for low risk breast cancers based on clinical and pathologic factors alone have shown conflicting results. More recently, prognostic genomic assays have shown promise in improving our ability to identify patients with a low risk of local recurrence in which adjuvant radiation may be safely omitted to prevent radiation-associated toxicities. Prospective clinical trials to investigate the ability of prognostic genomic assays to identify low risk breast cancers in which radiation can be safely omitted are currently ongoing. While prognostic genomic assays may help stratify breast cancers according to their risk of recurrence, the development and validation of predictive genomic assays may help us to better understand the likelihood of breast cancers to respond to radiation therapy, thus allowing for a more tailored locoregional approach to improve radiation outcomes. In this review, we summarize the current evidence for prognostic genomic assays and discuss the potential for predictive genomic assays to facilitate adjuvant radiation therapy decisions in the clinic.

2. De-Escalation of Radiation Therapy Based on Clinical Factors in Breast Cancer

The historical NSABP-06 trial randomized a non-selective cohort of n = 1851 early stage breast cancer patients to lumpectomy, lumpectomy and radiation, or mastectomy. The 20-year outcomes showed the incidence of recurrent tumor in the ipsilateral breast was 14.3% in women undergoing lumpectomy followed by breast irradiation compared to 39.2% in the women undergoing lumpectomy alone (p < 0.001) [4]. This study, which included all subtypes of early stage invasive breast cancers, indicated that radiation after lumpectomy substantially improved the rates of local recurrence, thus confirming lumpectomy followed by radiation as standard therapy for early stage breast cancers. Further retrospective studies have also suggested that omission of whole breast radiation post-lumpectomy in higher risk early stage breast cancers may decrease survival [17,18].
Several clinical studies have made attempts to identify “low risk” ER+ early stage breast cancers in which radiation can be omitted with similar rates of local control. NSABP-21 evaluated whether radiation could safely be omitted in select early stage breast cancer patients with small ER+ or ER− tumors < 1 cm and negative surgical margins [19]. In this study, n = 1009 patients with T1a or T1b breast cancers were randomized to tamoxifen alone, radiation + placebo, or radiation + tamoxifen. At 8 years, the cumulative incidence of in breast tumor recurrence was 16.5% with tamoxifen alone, 9.3% with radiation and placebo, and 2.8% with radiation and tamoxifen. Thus, radiation reduced local recurrence below the level achieved with tamoxifen alone [19]. Similar results were shown in the Austrian Breast and Colorectal Cancer Study Group (ABCSG) trial 8A, a prospective, multicenter clinical trial that randomized favorable early stage breast cancer patients (<3 cm, Grade 1 or 2, ER/PR+) receiving lumpectomy with radiation versus lumpectomy without radiation [20]. The 5-year recurrence rates were 2.1% in the radiation group versus 6.1% in the no-radiation group (p = 0.002) [20]. The results of the Swedish Breast Cancer Group 91 RT (SweBCG 91 RT) also confirmed that small early stage invasive breast cancers identified on screening mammogram had an improved in-breast tumor recurrence benefit from radiation therapy (23.9% vs. 11.5%, p < 00.1) [21,22].
While these randomized trials demonstrated a local control benefit of radiation in ER+ early stage breast cancers, elderly women with small, low grade ER+ breast cancers appeared to derive the least benefit [23]. In the Cancer and Leukemia Group B (CALGB) 9343 trial, 636 women aged ≥ 70 years old with T1 early stage ER+ breast cancers were randomized to receive tamoxifen alone versus tamoxifen plus radiation therapy post-lumpectomy [24,25]. At a long term follow up of 12.6 years, the local recurrence rates were only mildly improved with radiation (10% in the tamoxifen alone group vs. 2% in the tamoxifen and radiation group). However, the secondary endpoints of overall survival, distant metastases and mastectomy-free survival were the same between the two groups [24]. The PRIME-II trial, which included ER+ early stage breast cancer patients ≥ 65 years old with T1/T2 tumors demonstrated similar results with only mildly improved rates of local recurrence with radiation post-lumpectomy (4.1% vs. 1.3%) [26]. Given only a modest local control benefit of radiation, NCCN guidelines were revised to allow for the omission of radiation in patients ≥ 70 yo in 2005 [27].
In 2004, Fyles et al. [28] randomized n = 769 women ≥ 50 yo with T1/T2 early stage ER+, node-negative breast cancers to tamoxifen alone compared to tamoxifen plus radiation. Despite this favorable early stage ER+ breast cancer patient population, radiation significantly reduced the rate of local relapse. After a median follow up of 5.6 years, the risk of local relapse was 7.7% in the tamoxifen alone group compared to only 0.6% in the radiation plus tamoxifen group [28]. However, a further retrospective analysis by Liu et al. [29], showed a minimal rate of recurrence regardless of radiation in a subtype of breast cancer defined as ER+/PR+/Her-2 negative with a low Ki-67 value. These results prompted the question of whether better characterizing the biology of breast cancer may help identify breast cancers in which radiation has minimal benefit and can be safely omitted.
Most recently, the LUMINA single arm prospective clinical trial [30] showed that women ≥ 55 yo with Grade 1–2 invasive breast cancer of the Luminal A subtype (defined as: ER ≥ 1%, PR > 20%, HER2 negative and Ki67 ≤ 13.25%) had very low rates of local recurrence of 2.3% at 5 years when patients were treated with lumpectomy followed by endocrine therapy alone [31]. While these preliminary results show promise that Luminal A breast cancers may be candidates for radiation omission, the results of multiple prospective clinical trials to further characterize the role of adjuvant radiation among breast cancer molecular subtypes using genomic assays are ongoing.

3. Genomic Profiling of Breast Cancers

It is well-known that breast cancers represent a heterogenous group of diseases composed of several subtypes. Seminal genomic profiling studies have shown that breast cancer is composed of intrinsic molecular subtypes [32,33,34,35]. The Luminal A, Luminal B, Her-2 enriched and Basal-like molecular subtypes demonstrate different disease-specific outcomes and treatment responses. Multiple studies have shown that these molecular subtypes of breast cancer strongly predict locoregional recurrence in addition to distant recurrence [36,37,38,39,40]. While molecular subtypes facilitate adjuvant systemic and radiation treatment decisions, the use of genome-wide gene expression profiling microarrays can be costly, time consuming and challenging to implement in the clinic.
Consequently, molecular subtypes are often approximated in the clinic by immunohistochemical staining for ER, PR and Her-2 status, grade and Ki-67 proliferation rate. Multiple studies have shown that Luminal A breast cancers had the lowest rate of regional local recurrence and best prognosis. In contrast, Her-2 enriched and Basal-like subtypes demonstrated an increased risk of local recurrence [37,38,39,40]. Arvold et al. [37] approximated these subtypes based on immunohistochemistry and grade and determined the 5-year cumulative incidence of local recurrence was 0.8% for Luminal A, 2.3% for Luminal B, 1.1% for Luminal HER-2, 10.8% for HER2-enriched and 6.7% for Basal-like early stage breast cancers. Moreover, Voduc et al. [39] defined these molecular subtypes based on a six gene panel, including ER, PR, Her-2, Ki-67, epidermal growth factor receptor (EGFR), and cytokeratin (CK) 5/6. Bane et al. [38] classified tumors by molecular subtype, including Luminal A, Luminal B, Her-2 enriched, Basal-like or unclassified based on immunohistochemical staining for ER, PR, Her-2, Ki-67, CK 5/6, EGFR status in addition to HIF-1alpha, CAIX and GLUT1. In this study, the 10 yr cumulative local recurrence incidence was 4.5% for Luminal A and Basal-like, 7.9% for Luminal B and 16.9% for Her-2 enriched tumors [38].
Commercially available genomic assays have now been developed and are commonly implemented in the clinic as prognostic and predictive tools for adjuvant systemic therapy. In the next section, we discuss OncotypeDx, Mammoprint, and other commercially available gene assays for guiding adjuvant systemic therapy decisions in the clinic.

4. Genome Expression Assays as Prognostic and Predictive Biomarkers for Adjuvant Systemic Therapy

Genomic assays are now routinely used for making adjuvant systemic treatment decisions in the clinic [41,42,43,44,45,46]. OncotypeDx (Exact Sciences, Madison, WI, USA) is a multigene assay using RNA expression profiling on a panel of 21 genes, including 16 cancer-related genes associated with proliferation, invasion, and estrogen signaling in addition to 5 reference genes [47]. Gene expression levels are included in a mathematical algorithm to generate a Recurrence Score (RS) from 0–100 that stratifies patients into three different risk categories of low (0–18), intermediate (18–31), and high (31–100) risk of distant recurrence.
Multiple retrospective studies showed that Recurrence Scores using this 21 gene assay could not only estimate the risk of distant recurrence in ER+, node-negative breast cancers on NSABP B-14 and NSABP B-20 [48,49,50,51,52], however it could also predict breast cancers most likely to benefit from adjuvant chemotherapy [49]. Indeed, Paik et al. [49] showed that patients with high Recurrence Scores (≥31) experienced a large benefit from cyclophosphamide, methotrexate, and fluorouracil chemotherapy (27.6% absolute mean decrease in distant recurrence rate at 10 years), while patients that had low Recurrence Scores (<18) experienced minimal benefit from chemotherapy (1.1% absolute mean decrease in distant recurrence at 10 years). While the role for chemotherapy was clear in high and low risk cases, it was less certain in patients with intermediate Recurrence Scores [49].
The Trial Assigning Individualized Options for Treatment (TAILORx) was a prospective randomized clinical trial designed to address this uncertainty in chemotherapy benefit among intermediate Recurrence Scores [53]. ER+, node-negative breast cancer patients with intermediate (11–25) Recurrence Scores were randomized to receive either chemotherapy and endocrine therapy or endocrine therapy alone [53]. Patients with Recurrence Scores between 11 and 25 had similar outcomes whether treated with chemoendocrine therapy or endocrine therapy alone. Both groups had a disease-free survival rate of ~84%, disease-free metastasis of ~95% and overall survival of ~93% at a follow up of 9 years. However, some survival benefit was observed in women < 50 yo with Recurrence Scores of 16 to 25, with benefit increasing as the Recurrence Score increased [53,54].
In regard to node positive patients, Albain et al. [55] performed a retrospective analysis that showed the 21-gene Recurrence Score to also be prognostic and predictive of chemotherapy benefit in a subset of post-menopausal, ER+, node positive women on the Southwest Oncology Group (SWOG)-8814 INT-0100 trial [55,56]. In this study, patients with Recurrence Scores < 18 showed no benefit to chemotherapy, however patients with Recurrence Scores ≥ 31 showed increased disease-free survival, overall survival and breast cancer specific survival in patients treated with chemotherapy [55]. The prognostic and predictive role of Recurrence Scores in node-positive breast cancers was evaluated by RxPonder (A Clinical Trial RX for Positive Node, Endocrine Responsive Breast Cancer) [57], a prospective clinical trial randomizing women with ER+, node positive (1–3 positive lymph nodes) and Recurrence Scores of 0–25 to chemotherapy plus endocrine therapy vs. endocrine therapy alone. The RxPonder trials showed that ER+ post-menopausal women with 1–3 positive lymph nodes and a recurrence score of 0–25 showed no clinically relevant benefit to adjuvant chemotherapy. However, for the 33% of pre-menopausal women enrolled on the trial, an increase in disease-free survival of 40% and relapse-free survival of 42% was seen with the addition of chemotherapy regardless of recurrence score [57]. As a result, genomic assays are used in making adjuvant systemic therapy decisions for post-menopausal women, but not pre-menopausal women, with 1–3 positive nodes.
MammaPrint (Agendia Precision Oncology, Irvine, CA, USA) is a prognostic 70-gene signature developed by the Netherlands Cancer Institute that out-performed all clinical variables in predicting distant metastases over five years. The 70 gene signature involving genes associated with cell cycle, invasion, metastases and signal transduction was able to classify breast cancer patients with tumor size < 5 cm, lymph node negative and age < 55 yo into “poor prognosis” and “good prognosis” subtypes [58,59]. The initial validation study showed that node-negative breast cancer patients stratified as low risk by the 70-gene signature demonstrated a 10-year overall survival of 92%, whereas patients classified as high risk had a 59.5% 10-year overall survival [60]. The prospective Phase III MINDACT (Microarray In Node-Negative and 1 to 3 Positive Lymph Node Disease May Avoid Chemotherapy Trial) later showed that the 70 gene signature identified patients with high clinical risk and low genomic risk of distant metastases that did not benefit from the addition of chemotherapy to endocrine therapy at a median follow up of 8.7 years [61,62]. More recent studies suggest that this 70-gene Mammaprint signature may identify ultra-low risk breast cancers that could be candidates for further de-escalation of endocrine therapy [63,64].
Based on these previously discussed multiple prospective clinical trials, ASCO guidelines provide evidence-based recommendations for the use of genomic assays for guiding adjuvant systemic therapies among early-stage breast cancers [43,45,46].

5. Prognostic Genomic Assays and Adjuvant Radiation Therapy

While the past decade has seen a significant increase in the utilization of genomic assays in making adjuvant systemic therapy recommendations, the relevance of genomic assays in guiding radiation therapy recommendations remains uncertain. Clinicopathologic factors such as grade, tumor size, lymph node status and age independently correlate with local recurrence. However, prospective clinical trials aimed to identify low-risk breast cancers based on clinicopathologic factors alone in which radiation therapy can be omitted with minimal effect on disease control have been unsuccessful. Radiation provided at least a modest benefit in local control in all the studies [19,20,24,25,26,65].
The previously described Oncotype and Mammaprint assays are not only prognostic for distant recurrence, but have also been shown to be prognostic for local recurrence [50,66,67,68,69,70]. In 895 tamoxifen-treated ER+, node negative patients from NSABP B-14 and B-20, locoregional recurrence (LRR) was significantly associated with Recurrence Score. The 10-year LRR was 4.3% for patients with a low RS (<18), 7.2% for those with intermediate RS (18–30), and 15.8% for those with a high RS (≥30) [50]. Recurrence Score remained an independent predictor of LRR in ER+, node-positive patients treated with adjuvant chemotherapy plus tamoxifen in NSABP B-28 [71].
The risk-of-recurrence (ROR) score derived from the Predictor Analysis of Microarray (PAM) 50 (Prosigna) is a 50 gene assay that describes individual breast cancers based on intrinsic subtype and has also been shown to be an independent prognostic factor for risk of local recurrence in post-menopausal ER+ early stage breast cancer patients [72,73,74]. Women with breast cancer who had a low-risk PAM-50 ROR score (defined as <57) after breast conservation therapy on the Austrian Breast and Colorectal Cancer Study Group (ABCSG) 8 randomized trial had only a 0.9% risk of local recurrence [72]. These prognostic genomic assays have been summarized in Table 1.
Current prospective clinical trials are evaluating whether OncotypeDx and PAM-50 prognostic gene assays can be used to identify low risk ER+ early breast cancer patients in which radiation therapy can be safely omitted (Table 2). The IDEA (Individualized Decisions for Endocrine Therapy Alone) is a single arm Phase II prospective trial evaluating locoregional recurrence rates in low risk women (defined as post-menopausal, ER+ with a RS ≤ 18 who plan to receive endocrine therapy) in which radiation therapy will be omitted after breast conservation surgery [75]. The PRECISION (Profiling Early Breast Cancer for Radiation Omission) is a prospective Phase II single arm trial evaluating 5 year locoregional recurrence after the omission of radiation ER+ early stage breast cancers < 2 cm with a low risk PAM-50-based ROR in post-menopausal woman > 50 yo [76]. Both the IDEA and PRECISION trials have completed accrual. NRG-BR007 DEBRA (De-Escalation of Breast Radiation) is a currently accruing Phase III prospective, randomized trial evaluating omission of breast radiation in ER+/Her-2 negative early stage breast cancers with Oncotype Recurrence Scores ≤ 18 in women age 50–69 yo [77]. The TAILOR-RT Phase III is randomizing patients to regional nodal radiation that have ER+/Her2-negative biomarker low risk (defined as Oncotype ≤ 25) breast cancers with low lymph node burden (1–3+ lymph nodes) [78]. The EXPERT (Examining Personalized Radiation Therapy for Low Risk Early Breast Cancer) trial is a Phase III randomized trial evaluating the omission of breast radiation in ER+/Her-2 negative early stage breast cancers with PAM-50 ROR < 60 in women ≥ 50 yo [79]. Until the results of these prospective clinical trials become available, adjuvant breast radiation remains the standard of care after breast conservation surgery for most patients.

6. The Potential for Predictive Genomic Assays to Guide Adjuvant Radiation Therapy

While multiple studies have successfully shown that prognostic genomic assays are able to predict local recurrence in breast cancer [37,38,39], predictive genomic assays could potentially identify subpopulations of breast cancer that are most or least likely to benefit from radiation therapy (Figure 1). Genomic classifiers capable of predicting response to radiation are still under development. However, data has shown that the radiosensitivity of breast cancers appears to be independent of intrinsic molecular subtype and the signaling mechanisms associated with radiosensitivity or resistance may differ among breast cancer subtypes [29,38,80,81].
Speers et al. studied the intrinsic radiosensitivity of 16 human breast cancer cell lines using clonogenic survival assays to develop a 51 gene molecular signature that would identify women at an increased risk of local recurrence most likely to benefit from radiation [82]. This gene classifier was enriched with genes associated with cell cycle arrest and DNA damage response. Moreover, Nimeus-Malmstrom et al. found a reliable gene expression profile capable of predicting local recurrence despite radiation after breast conservation surgery in ER+ breast cancers [83]. Another study identified a 7 gene profile for node positive patients from the Danish Breast Cancer Cooperative Group (DBCG82bc) treated with systemic treatment and randomized to receive or not to receive post-mastectomy radiation therapy (PMRT). This 6 gene expression panel identified patients with “high LRR risk” in which PMRT significantly reduced the risk of LR and “low LRR risk” group in which PMRT showed no additional reduction in LR rate and the response to radiation was independent of intrinsic molecular subtype [81,84].
The Adjuvant Radiotherapy Intensification Classifier (ARTIC), comprised of 27 genes related to cell proliferation, cell cycle and kinase activity as well as patient age, is a gene classifier for radiation sensitivity in patients with high-risk node negative early stage breast cancers was developed using three publicly available cohorts. The ARTIC classifier was the first predictive assay to be later validated with a Phase III clinical trial in which patients were randomized to radiation [85].
The RSI radiosensitivity index is a linear model based on 10 genes (related to DNA damage response, histone deacetylation, cell cycle, apoptosis and proliferation) associated with the fraction of cells surviving after 2 Gy (SF2) of radiation. The RSI has been validated to predict clinical outcomes and benefit from radiation therapy among several cancer types, including breast cancer [86,87,88,89,90]. Torres-Roca et al. [88] later found that RSI may not predict radiation sensitivity among all breast cancer subtypes and may be influenced by ER status. Scott et al. later combined RSI with the linear quadratic model to create the Genome Adjusted Radiation Dose (GARD), a tool believed to predict a tumor’s response to a particular dose of radiation based on its inherent radiosensitivity [86,91].
Other groups have found genomic classifiers for radiation sensitivity that depend on the immune microenvironment. Shen et al. developed an 11 gene signature that stratify tumors based on response to radiation therapy and the tumor immune microenvironment. In this study, CD4+ T cells and B cells infiltrated the radiosensitive tumors and macrophages infiltrated the radioresistant tumors [92]. Cui et al. [93] also developed gene expression signatures based on intrinsic radiosensitivity and antitumor immunity. Validation of these predictive genomic classifiers in prospective clinical trials are warranted before implementation in the clinic.
In summary, while confirmatory studies are needed, many of these pre-clinical studies have identified genes involved in cell proliferation, DNA damage response, immune microenvironment and wound response signaling to be associated with radiation resistance in breast cancer. The results of these studies have been summarized in Table 3.

7. Summary and Conclusions

Clinical trials have shown that even the most low-risk breast cancers defined by clinical and pathologic factors alone demonstrate even a modest local control benefit from radiation. Therefore, current treatment guidelines recommend radiation therapy for nearly all breast cancer patients after breast conservation surgery. However, genomic assays are believed to better risk stratify breast cancers based on their inherent biology. While genomic assays for guiding adjuvant systemic therapy decisions in the clinic have been implemented over the past decade, the use of genomic assays in guiding radiation therapy decisions has yet to be adopted.
Retrospective studies have shown the ability of prognostic genomic assays, such as Oncotype and PAM-50, to predict risk of local recurrence among breast cancer subtypes. These data show promise for identifying low risk breast cancers in which radiation may be safely omitted. Several prospective clinical trials that use Oncotype and PAM-50 genomic assays to guide adjuvant radiation therapy decisions are currently underway and their results are eagerly awaited. Until these clinical trial results are available, post-lumpectomy radiation is recommended for all breast cancer patients < 70 years old regardless of genomic assay results.
While prognostic genomic assays may help stratify breast cancers according to their risk of local recurrence, the development of predictive genomic assays may help stratify breast cancers according to their likelihood to respond to radiation therapy. The use of predictive genomic assays may one day allow for a more tailored locoregional approach to improve radiation outcomes, however these assays are still under development and there is no consensus on the genes predominantly involved in radiation resistance. For example, lower doses of radiation could be considered in more radiosensitive breast cancer subtypes. In contrast, aggressive radioresistant breast cancers may still experience local recurrence despite radiation therapy. These patients may require more intensive locoregional therapies, such as mastectomy or an increase in radiation dose with tumor bed boost. Currently, the decision to boost the tumor bed is based on clinical factors such as patient age and tumor grade [94], however if a genomic assay were to suggest a more radioresistant breast cancer, radiation dose intensification with tumor bed boost may be considered. In cases of more radioresistant breast cancers, the use of chemotherapies and radiosensitizers may also be considered.

Author Contributions

Conceptualization, S.J.B. and J.G.B.; writing—original draft preparation, S.J.B. and M.T.; writing—review and editing, S.J.B., M.T., S.R.J., J.R.W. and J.G.B.; supervision, J.G.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Early Breast Cancer Trialists’ Collaborative, Group. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Clarke, M.; Collins, R.; Darby, S.; Davies, C.; Elphinstone, P.; Evans, V.; Godwin, J.; Gray, R.; Hicks, C.; James, 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. [Google Scholar] [PubMed]
  3. Blichert-Toft, M.; Nielsen, M.; Düring, M.; Møller, S.; Rank, F.; Overgaard, M.; Mouridsen, H.T. Long-term results of breast conserving surgery vs. mastectomy for early stage invasive breast cancer: 20-year follow-up of the Danish randomized DBCG-82TM protocol. Acta Oncol. 2008, 47, 672–681. [Google Scholar] [CrossRef] [PubMed]
  4. Fisher, B.; Anderson, S.; Bryant, J.; Margolese, R.G.; Deutsch, M.; Fisher, E.R.; Jeong, J.-H.; Wolmark, N. Twenty-year follow-up of a randomized trial comparing total mastectomy, lumpectomy, and lumpectomy plus irradiation for the treatment of invasive breast cancer. N. Engl. J. Med. 2002, 347, 1233–1241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Litière, S.; Werutsky, G.; Fentiman, I.S.; Rutgers, E.; Christiaens, M.-R.; Van Limbergen, E.; Baaijens, M.H.; Bogaerts, J.; Bartelink, H. Breast conserving therapy versus mastectomy for stage I–II breast cancer: 20 year follow-up of the EORTC 10801 phase 3 randomised trial. Lancet Oncol. 2012, 13, 412–419. [Google Scholar] [CrossRef]
  6. Veronesi, U.; Cascinelli, N.; Mariani, L.; Greco, M.; Saccozzi, R.; Luini, A.; Aguilar, M.; Marubini, E. Twenty-year follow-up of a randomized study comparing breast-conserving surgery with radical mastectomy for early breast cancer. N. Engl. J. Med. 2002, 347, 1227–1232. [Google Scholar] [CrossRef] [PubMed]
  7. Darby, S.C.; Ewertz, M.; McGale, P.; Bennet, A.M.; Blom-Goldman, U.; Brønnum, D.; Correa, C.; Cutter, D.; Gagliardi, G.; Gigante, B.; et al. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N. Engl. J. Med. 2013, 368, 987–998. [Google Scholar] [CrossRef] [Green Version]
  8. Lingos, T.I.; Recht, A.; Vicini, F.; Abner, A.; Silver, B.; Harris, J.R. Radiation pneumonitis in breast cancer patients treated with conservative surgery and radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 1991, 21, 355–360. [Google Scholar] [CrossRef]
  9. Back, M. Impact of radiation therapy on acute toxicity in breast conservation therapy for early breast cancer. Clin. Oncol. 2004, 16, 12–16. [Google Scholar] [CrossRef]
  10. Vicini, F.A.; Cecchini, R.S.; White, J.R.; Arthur, D.W.; Julian, T.B.; A Rabinovitch, R.; Kuske, R.R.; A Ganz, P.; Parda, D.S.; Scheier, M.F.; et al. Long-term primary results of accelerated partial breast irradiation after breast-conserving surgery for early-stage breast cancer: A randomised, phase 3, equivalence trial. Lancet 2019, 394, 2155–2164. [Google Scholar] [CrossRef]
  11. Meattini, I.; Marrazzo, L.; Saieva, C.; Desideri, I.; Scotti, V.; Simontacchi, G.; Bonomo, P.; Greto, D.; Mangoni, M.; Scoccianti, S.; et al. Accelerated Partial-Breast Irradiation Compared With Whole-Breast Irradiation for Early Breast Cancer: Long-Term Results of the Randomized Phase III APBI-IMRT-Florence. Trial. J. Clin. Oncol. 2020, 38, 4175–4183. [Google Scholar] [CrossRef] [PubMed]
  12. NSABP B-39, RTOG 0413: A Randomized Phase III Study of conventional whole breast irradiation versus partial breast irradiation for women with stage 0, I, or II breast cancer. Clin. Adv. Hematol. Oncol. 2006, 4, 719–721.
  13. Coles, C.E.; Griffin, C.L.; Kirby, A.M.; Titley, J.; Agrawal, R.K.; Alhasso, A.; Bhattacharya, I.S.; Brunt, A.M.; Ciurlionis, L.; Chan, C.; et al. Partial-breast radiotherapy after breast conservation surgery for patients with early breast cancer (UK IMPORT LOW trial): 5-year results from a multicentre, randomised, controlled, phase 3, non-inferiority trial. Lancet 2017, 390, 1048–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Correa, C.; Harris, E.E.; Leonardi, M.C.; Smith, B.D.; Taghian, A.G.; Thompson, A.M.; White, J.; Harris, J.R. Accelerated Partial Breast Irradiation: Executive summary for the update of an ASTRO Evidence-Based Consensus Statement. Pract. Radiat. Oncol. 2017, 7, 73–79. [Google Scholar] [CrossRef]
  15. Murray Brunt, A.; Haviland, J.S.; Wheatley, D.A.; Sydenham, M.A.; Alhasso, A.; Bloomfield, D.J.; Chan, C.; Churn, M.; Cleator, S.; Coles, C.E.; 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, 1613–1626. [Google Scholar] [CrossRef]
  16. Brunt, A.M.; Haviland, J.; Sydenham, M.; Agrawal, R.K.; Algurafi, H.; Alhasso, A.; Barrett-Lee, P.; Bliss, P.; Bloomfield, D.; Bowen, J.; et al. Ten-Year Results of FAST: A Randomized Controlled Trial of 5-Fraction Whole-Breast Radiotherapy for Early Breast Cancer. J. Clin. Oncol. 2020, 38, 3261–3272. [Google Scholar] [CrossRef]
  17. Haque, W.; Verma, V.; Butler, E.B.; Teh, B.S. Omission of radiotherapy in elderly women with early stage metaplastic breast cancer. Breast 2018, 38, 154–159. [Google Scholar] [CrossRef] [PubMed]
  18. Bazan, J.G.; Jhawar, S.R.; Stover, D.; Park, K.U.; Beyer, S.; Healy, E.; White, J.R. De-escalation of radiation therapy in patients with stage I, node-negative, HER2-positive breast cancer. NPJ Breast Cancer 2021, 7, 33. [Google Scholar] [CrossRef]
  19. Fisher, B.; Bryant, J.; Dignam, J.J.; Wickerham, D.L.; Mamounas, E.P.; Fisher, E.R.; Margolese, R.G.; Nesbitt, L.; Paik, S.; Pisansky, T.M.; et al. Tamoxifen, radiation therapy, or both for prevention of ipsilateral breast tumor recurrence after lumpectomy in women with invasive breast cancers of one centimeter or less. J. Clin. Oncol. 2002, 20, 4141–4149. [Google Scholar] [CrossRef]
  20. Pötter, R.; Gnant, M.; Kwasny, W.; Tausch, C.; Handl-Zeller, L.; Pakisch, B.; Taucher, S.; Hammer, J.; Luschin-Ebengreuth, G.; Schmid, M.; 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. [Google Scholar] [CrossRef]
  21. Malmström, P.; Holmberg, L.; Anderson, H.; Mattsson, J.; Jönsson, P.-E.; Tennvall-Nittby, L.; Balldin, G.; Lovén, L.; Svensson, J.-H.; Ingvar, C.; et al. Breast conservation surgery, with and without radiotherapy, in women with lymph node-negative breast cancer: A randomised clinical trial in a population with access to public mammography screening. Eur. J. Cancer 2003, 39, 1690–1697. [Google Scholar] [CrossRef] [PubMed]
  22. Killander, F.; Karlsson, P.; Anderson, H.; Mattsson, J.; Holmberg, E.; Lundstedt, D.; Malmström, P. No breast cancer subgroup can be spared postoperative radiotherapy after breast-conserving surgery. Fifteen-year results from the Swedish Breast Cancer Group randomised trial, SweBCG 91 RT. Eur. J. Cancer 2016, 67, 57–65. [Google Scholar] [CrossRef] [PubMed]
  23. Fisher, E.R.; Anderson, S.; Tan-Chiu, E.; Fisher, B.; Eaton, L.; Wolmark, N. Fifteen-year prognostic discriminants for invasive breast carcinoma: National Surgical Adjuvant Breast and Bowel Project Protocol-06. Cancer 2001, 91, 1679–1687. [Google Scholar] [CrossRef] [PubMed]
  24. Hughes, K.S.; Schnaper, L.A.; Bellon, J.R.; Cirrincione, C.T.; Berry, D.A.; Mc Cormick, B.; Muss, H.B.; Smith, B.L.; Hudis, C.A.; Winer, E.P.; 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. [Google Scholar] [CrossRef] [Green Version]
  25. Hughes, K.S.; Schnaper, L.A.; Berry, D.; Cirrincione, C.; McCormick, B.; Shank, B.; Wheeler, J.; Champion, L.A.; Smith, T.J.; Smith, B.L.; et al. 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, 971–977. [Google Scholar] [CrossRef] [Green Version]
  26. Kunkler, I.H.; Williams, L.J.; Jack, W.J.L.; A Cameron, D.; Dixon, J.M. 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. [Google Scholar] [CrossRef]
  27. N.C.C.N Clinical Practice Guidelines in Oncology Breast Cancer. 2022. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1419 (accessed on 7 November 2022).
  28. Fyles, A.; McCready, D.; Manchul, L.; Trudeau, M.; Merante, P.; Pintilie, M.; Weir, L.; Olivotto, I. A randomized trial of tamoxifen with or without breast radiation in women with early breast cancer 50 years of age and older. Radiother. Oncol. 2004, 73, S14. [Google Scholar]
  29. Liu, F.-F.; Shi, W.; Done, S.J.; Miller, N.; Pintilie, M.; Voduc, D.; Nielsen, T.O.; Nofech-Mozes, S.; Chang, M.C.; Whelan, T.J.; 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. [Google Scholar] [CrossRef]
  30. ClinicalTrials.gov. A Prospective Cohort Study Evaluating Risk of Local Recurrence Following Breast Conserving Surgery and Endocrine Therapy in Low Risk Luminal A Breast Cancer (LUMINA). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT01791829 (accessed on 10 November 2022).
  31. Whelan, T.J.; Smith, S.; Nielsen, T.O.; Parpia, S.; Fyles, A.W.; Bane, A.; Liu, F.-F.; Grimard, L.; Stevens, C.; Bowen, J.; et al. LUMINA: A prospective trial omitting radiotherapy (RT) following breast conserving surgery (BCS) in T1N0 luminal A breast cancer (BC). J. Clin. Oncol. 2022, 40, LBA501. [Google Scholar] [CrossRef]
  32. Perou, C.M.; Sørlie, T.; Eisen, M.B.; Van De Rijn, M.; Jeffrey, S.S.; Rees, C.A.; Pollack, J.R.; Ross, D.T.; Johnsen, H.; Akslen, L.A.; et al. Molecular portraits of human breast tumours. Nature 2000, 406, 747–752. [Google Scholar] [CrossRef] [Green Version]
  33. Sørlie, T.; Perou, C.M.; Tibshirani, R.; Aas, T.; Geisler, S.; Johnsen, H.; Hastie, T.; Eisen, M.B.; van de Rijn, M.; Jeffrey, S.S.; et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc. Natl. Acad. Sci. USA 2001, 98, 10869–10874. [Google Scholar] [CrossRef] [Green Version]
  34. Sørlie, T.; Tibshirani, R.; Parker, J.; Hastie, T.; Marron, J.S.; Nobel, A.; Deng, S.; Johnsen, H.; Pesich, R.; Geisler, S.; et al. Repeated observation of breast tumor subtypes in independent gene expression data sets. Proc. Natl. Acad. Sci. USA 2003, 100, 8418–8423. [Google Scholar] [CrossRef] [PubMed]
  35. The Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Nguyen, P.L.; Taghian, A.G.; Katz, M.S.; Niemierko, A.; Raad, R.F.A.; Boon, W.L.; Bellon, J.R.; Wong, J.S.; Smith, B.L.; Harris, J.R. Breast cancer subtype approximated by estrogen receptor, progesterone receptor, and HER-2 is associated with local and distant recurrence after breast-conserving therapy. J. Clin. Oncol. 2008, 26, 2373–2378. [Google Scholar] [CrossRef] [PubMed]
  37. Arvold, N.D.; Taghian, A.G.; Niemierko, A.; Raad, R.F.A.; Sreedhara, M.; Nguyen, P.L.; Bellon, J.R.; Wong, J.S.; Smith, B.L.; Harris, J.R.; et al. Age, breast cancer subtype approximation, and local recurrence after breast-conserving therapy. J. Clin. Oncol. 2011, 29, 3885–3891. [Google Scholar] [CrossRef]
  38. Bane, A.L.; Whelan, T.; Pond, G.; Parpia, S.; Gohla, G.; Fyles, A.; Pignol, J.-P.; Pritchard, K.; Chambers, S.; Levine, M. Tumor factors predictive of response to hypofractionated radiotherapy in a randomized trial following breast conserving therapy. Ann. Oncol. 2014, 25, 992–998. [Google Scholar] [CrossRef]
  39. Voduc, K.D.; Cheang, M.C.U.; Tyldesley, S.; Gelmon, K.; Nielsen, T.O.; Kennecke, H. Breast cancer subtypes and the risk of local and regional relapse. J. Clin. Oncol. 2010, 28, 1684–1691. [Google Scholar] [CrossRef] [Green Version]
  40. Lowery, A.J.; Kell, M.R.; Glynn, R.W.; Kerin, M.J.; Sweeney, K.J. Locoregional recurrence after breast cancer surgery: A systematic review by receptor phenotype. Breast Cancer Res. Treat. 2012, 133, 831–841. [Google Scholar] [CrossRef]
  41. Carlson, J.J.; Roth, J.A. The impact of the Oncotype Dx breast cancer assay in clinical practice: A systematic review and meta-analysis. Breast Cancer Res. Treat. 2013, 141, 13–22. [Google Scholar] [CrossRef]
  42. Harris, L.; Fritsche, H.; Mennel, R.; Norton, L.; Ravdin, P.; Taube, S.; Somerfield, M.R.; Hayes, D.F.; Bast, R.C. American society of clinical oncology 2007 update of recommendations for the use of tumor markers in breast cancer. J. Clin. Oncol. 2007, 25, 5287–5312. [Google Scholar] [CrossRef] [Green Version]
  43. Harris, L.N.; Ismaila, N.; McShane, L.M.; Hayes, D.F. Use of Biomarkers to Guide Decisions on Adjuvant Systemic Therapy for Women With Early-Stage Invasive Breast Cancer: American Society of Clinical Oncology Clinical Practice Guideline Summary. J. Oncol. Pract. 2016, 12, 384–389. [Google Scholar] [CrossRef] [PubMed]
  44. Goldhirsch, A.; Winer, E.P.; Coates, A.S.; Gelber, R.D.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223. [Google Scholar] [CrossRef] [PubMed]
  45. Raab, R.; Ismaila, N.; Andre, F.; Stearns, V.; Kalinsky, K. Biomarkers for Adjuvant Endocrine and Chemotherapy in Early-Stage Breast Cancer: ASCO Guideline Update Q and A. Jco Oncol. Pract. 2022, 18, 646–648. [Google Scholar] [CrossRef] [PubMed]
  46. Andre, F.; Ismaila, N.; Allison, K.H.; Barlow, W.E.; Collyar, D.E.; Damodaran, S.; Henry, N.L.; Jhaveri, K.; Kalinsky, K.; Kuderer, N.M.; et al. Biomarkers for Adjuvant Endocrine and Chemotherapy in Early-Stage Breast Cancer: ASCO Guideline Update. J. Clin. Oncol. 2022, 40, 1816–1837. [Google Scholar] [CrossRef] [PubMed]
  47. Sparano, J.A.; Paik, S. Development of the 21-gene assay and its application in clinical practice and clinical trials. J. Clin. Oncol. 2008, 26, 721–728. [Google Scholar] [CrossRef]
  48. Paik, S.; Shak, S.; Tang, G.; Kim, C.; Baker, J.; Cronin, M.; Baehner, F.L.; Walker, M.G.; Watson, D.; Park, T.; et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N. Engl. J. Med. 2004, 351, 2817–2826. [Google Scholar] [CrossRef] [Green Version]
  49. Paik, S.; Tang, G.; Shak, S.; Kim, C.; Baker, J.; Kim, W.; Cronin, M.; Baehner, F.L.; Watson, D.; Bryant, J.; et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor—positive breast cancer. J. Clin. Oncol. 2006, 24, 3726–3734. [Google Scholar] [CrossRef]
  50. Mamounas, E.P.; Tang, G.; Fisher, B.; Paik, S.; Shak, S.; Costantino, J.P.; Watson, D.; Geyer, C.E.; Wickerham, D.L.; Wolmark, N. 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. [Google Scholar] [CrossRef]
  51. Tang, G.; Shak, S.; Paik, S.; Anderson, S.; Costantino, J.P.; Geyer, C.E., Jr.; Mamounas, E.P.; Wickerham, D.L.; Wolmark, N. Comparison of the prognostic and predictive utilities of the 21-gene Recurrence Score assay and Adjuvant! for women with node-negative, ER-positive breast cancer: Results from NSABP B-14 and NSABP B-20. Breast Cancer Res. Treat. 2011, 127, 133–142. [Google Scholar] [CrossRef] [Green Version]
  52. Habel, L.A.; Shak, S.; Jacobs, M.K.; Capra, A.; Alexander, C.; Pho, M.; Baker, J.; Walker, M.; Watson, D.; Hackett, J.; et al. A population-based study of tumor gene expression and risk of breast cancer death among lymph node-negative patients. Breast Cancer Res. 2006, 8, R25. [Google Scholar] [CrossRef] [Green Version]
  53. Sparano, J.A.; Gray, R.J.; Makower, D.F.; Pritchard, K.I.; Albain, K.S.; Hayes, D.F.; Geyer, C.E., Jr.; Dees, E.C.; Goetz, M.P.; Olson, J.A.; et al. Adjuvant Chemotherapy Guided by a 21-Gene Expression Assay in Breast Cancer. N. Engl. J. Med. 2018, 379, 111–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Sparano, J.A.; Gray, R.J.; Ravdin, P.M.; Makower, D.F.; Pritchard, K.I.; Albain, K.S.; Hayes, D.F.; Geyer, C.E.; Dees, E.C.; Goetz, M.P.; et al. Clinical and Genomic Risk to Guide the Use of Adjuvant Therapy for Breast Cancer. N. Engl. J. Med. 2019, 380, 2395–2405. [Google Scholar] [CrossRef] [PubMed]
  55. Albain, K.S.; Barlow, W.E.; Shak, S.; Hortobagyi, G.N.; Livingston, R.B.; Yeh, I.-T.; Ravdin, P.; Bugarini, R.; Baehner, F.L.; Davidson, N.; et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: A retrospective analysis of a randomised trial. Lancet Oncol. 2010, 11, 55–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Albain, K.S.; Barlow, W.E.; Ravdin, P.M.; Farrar, W.B.; Burton, G.V.; Ketchel, S.J.; Cobau, C.D.; Levine, E.G.; Ingle, J.N.; Pritchard, K.I.; et al. Adjuvant chemotherapy and timing of tamoxifen in postmenopausal patients with endocrine-responsive, node-positive breast cancer: A phase 3, open-label, randomised controlled trial. Lancet 2009, 374, 2055–2063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kalinsky, K.; Barlow, W.E.; Gralow, J.R.; Meric-Bernstam, F.; Albain, K.S.; Hayes, D.F.; Lin, N.U.; Perez, E.A.; Goldstein, L.J.; Chia, S.K.; et al. 21-Gene Assay to Inform Chemotherapy Benefit in Node-Positive Breast Cancer. N. Engl. J. Med. 2021, 385, 2336–2347. [Google Scholar] [CrossRef]
  58. Vijver, M.J.v.d.; He, Y.D.; Veer, L.J.v.; Dai, H.; Hart, A.A.M.; Voskuil, D.W.; Schreiber, G.J.; Peterse, J.L.; Roberts, C.; Marton, M.J.; et al. A gene-expression signature as a predictor of survival in breast cancer. N. Engl. J. Med. 2002, 347, 1999–2009. [Google Scholar] [CrossRef] [Green Version]
  59. Van’t Veer, L.J.; Dai, H.; Van De Vijver, M.J.; He, Y.D.; Hart, A.A.M.; Mao, M.; Peterse, H.L.; Van Der Kooy, K.; Marton, M.J.; Witteveen, A.T.; et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 2002, 415, 530–536. [Google Scholar] [CrossRef] [Green Version]
  60. Buyse, M.; Loi, S.; Veer, L.V.; Viale, G.; Delorenzi, M.; Glas, A.; D’Assignies, M.S.; Bergh, J.; Lidereau, R.; Ellis, P.; et al. Validation and clinical utility of a 70-gene prognostic signature for women with node-negative breast cancer. J. Natl. Cancer Inst. 2006, 98, 1183–1192. [Google Scholar] [CrossRef] [Green Version]
  61. Piccart, M.; van’t Veer, L.J.; Poncet, C.; Cardozo, J.M.N.L.; Delaloge, S.; Pierga, J.-Y.; Vuylsteke, P.; Brain, E.; Vrijaldenhoven, S.; Neijenhuis, P.A.; et al. 70-gene signature as an aid for treatment decisions in early breast cancer: Updated results of the phase 3 randomised MINDACT trial with an exploratory analysis by age. Lancet Oncol. 2021, 22, 476–488. [Google Scholar]
  62. Cardoso, F.; van’t Veer, L.J.; Bogaerts, J.; Slaets, L.; Viale, G.; Delaloge, S.; Pierga, J.Y.; Brain, E.; Causeret, S.; Delorenzi, M.; et al. 70-Gene Signature as an Aid to Treatment Decisions in Early-Stage Breast Cancer. N. Engl. J. Med. 2016, 375, 717–729. [Google Scholar] [CrossRef] [Green Version]
  63. Cardozo, J.M.N.L.; Drukker, C.A.; Rutgers, E.J.T.; Schmidt, M.K.; Glas, A.M.; Witteveen, A.; Cardoso, F.; Piccart, M.; Esserman, L.J.; Poncet, C.; et al. Outcome of Patients With an Ultralow-Risk 70-Gene Signature in the MINDACT. Trial. J. Clin. Oncol. 2022, 40, 1335–1345. [Google Scholar] [CrossRef] [PubMed]
  64. Delahaye, L.J.M.J.; Drukker, C.A.; Dreezen, C.; Witteveen, A.; Chan, B.; Snel, M.; Beumer, I.J.; Bernards, R.; Audeh, M.W.; Veer, L.J.V.; et al. A breast cancer gene signature for indolent disease. Breast Cancer Res. Treat. 2017, 164, 461–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Jayasekera, J.; Schechter, C.B.; Sparano, J.A.; Jagsi, R.; White, J.; Chapman, J.-A.W.; Whelan, T.; Anderson, S.J.; Fyles, A.W.; Sauerbrei, W.; et al. Effects of Radiotherapy in Early-Stage, Low-Recurrence Risk, Hormone-Sensitive Breast Cancer. J. Natl. Cancer Inst. 2018, 110, 1370–1379. [Google Scholar] [CrossRef] [PubMed]
  66. Woodward, W.A.; Barlow, W.E.; Jagsi, R.; Buchholz, T.; Shak, S.; Baehner, F.; Whelan, T.J.; Davidson, N.E.; Ingle, J.N.; King, T.A.; et al. Association Between 21-Gene Assay Recurrence Score and Locoregional Recurrence Rates in Patients With Node-Positive Breast Cancer. JAMA Oncol. 2020, 6, 505–511. [Google Scholar] [CrossRef] [PubMed]
  67. Solin, L.J.; Gray, R.; Goldstein, L.J.; Recht, A.; Baehner, F.L.; Shak, S.; Badve, S.; Perez, E.A.; Shulman, L.N.; Martino, S.; 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. [Google Scholar] [CrossRef] [Green Version]
  68. Cheng, S.H.; Horng, C.-F.; West, M.; Huang, E.; Pittman, J.; Tsou, M.-H.; Dressman, H.; Chen, C.-M.; Tsai, S.Y.; Jian, J.J.; et al. Genomic prediction of locoregional recurrence after mastectomy in breast cancer. J. Clin. Oncol. 2006, 24, 4594–4602. [Google Scholar] [CrossRef] [Green Version]
  69. Nuyten, D.S.A.; Kreike, B.; Hart, A.A.M.; Chi, J.-T.A.; Sneddon, J.B.; Wessels, L.F.A.; Peterse, H.J.; Bartelink, H.; Brown, P.O.; Chang, H.Y.; et al. Predicting a local recurrence after breast-conserving therapy by gene expression profiling. Breast Cancer Res. 2006, 8, R62. [Google Scholar] [CrossRef] [Green Version]
  70. Drukker, C.A.; Elias, S.G.L.; Nijenhuis, M.V.L.; Wesseling, J.; Bartelink, H.; Elkhuizen, P.; Fowble, B.; Whitworth, P.W.; Patel, R.R.; Snoo, F.A.d.; et al. Gene expression profiling to predict the risk of locoregional recurrence in breast cancer: A pooled analysis. Breast Cancer Res. Treat. 2015, 149, 599–613. [Google Scholar] [CrossRef] [Green Version]
  71. Mamounas, E.P.; Liu, Q.; Paik, S.; Baehner, F.L.; Tang, G.; Jeong, J.-H.; Kim, S.R.; Butler, S.M.; Jamshidian, F.; Cherbavaz, D.B.; 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, djw259. [Google Scholar] [CrossRef]
  72. Fitzal, F.; Filipits, M.; Fesl, C.; Rudas, M.; Greil, R.; Balic, M.; Moinfar, F.; Herz, W.; Dubsky, P.; Bartsch, R.; et al. PAM-50 predicts local recurrence after breast cancer surgery in postmenopausal patients with ER+/HER2-disease: Results from 1204 patients in the randomized ABCSG-8 trial. Br. J. Surg. 2021, 108, 308–314. [Google Scholar] [CrossRef]
  73. Wallden, B.; Storhoff, J.; Nielsen, T.; Dowidar, N.; Schaper, C.; Ferree, S.; Liu, S.; Leung, S.; Geiss, G.; Snider, J.; et al. Development and verification of the PAM50-based Prosigna breast cancer gene signature assay. BMC Med. Genom. 2015, 8, 54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Gnant, M.; Filipits, M.; Greil, R.; Stoeger, H.; Rudas, M.; Bago-Horvath, Z.; Mlineritsch, B.; Kwasny, W.; Knauer, M.; Singer, C.; et al. Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: Using the PAM50 Risk of Recurrence score in 1478 postmenopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann. Oncol. 2014, 25, 339–345. [Google Scholar] [CrossRef] [PubMed]
  75. ClinicalTrials.gov. The IDEA Study (Individualized Decisions for Endocrine Therapy Alone). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT02400190 (accessed on 10 November 2022).
  76. ClinicalTrials.gov. The PRECISION Trial (Profiling Early Breast Cancer for Radiotherapy Omission): A Phase II Study of Breast-Conserving Surgery Without Adjuvant Radiotherapy for Favorable-Risk Breast Cancer. 2022. Available online: https://www.clinicaltrials.gov/ct2/show/NCT02653755 (accessed on 10 November 2022).
  77. ClinicalTrials.gov. De-Escalation of Breast Radiation Trial for Hormone Sensitive, HER-2 Negative, Oncotype Recurrence Score Less Than or Equal to 18 Breast Cancer (DEBRA). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT04852887 (accessed on 10 November 2022).
  78. ClinicalTrials.gov. Regional Radiotherapy in Biomarker Low-Risk Node Positive and T3N0 Breast Cancer (TAILOR RT). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT03488693 (accessed on 10 November 2022).
  79. ClinicalTrials.gov. EXamining PErsonalised Radiation Therapy for Low-Risk Early Breast Cancer. 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT02889874 (accessed on 10 November 2022).
  80. He, L.; Lv, Y.; Song, Y.; Zhang, B. The prognosis comparison of different molecular subtypes of breast tumors after radiotherapy and the intrinsic reasons for their distinct radiosensitivity. Cancer Manag. Res. 2019, 11, 5765–5775. [Google Scholar] [CrossRef] [PubMed]
  81. Tramm, T.; Kyndi, M.; Myhre, S.; Nord, S.; Alsner, J.; Sørensen, F.B.; Sørlie, T.; Overgaard, J. 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, 1337–1346. [Google Scholar] [CrossRef]
  82. Speers, C.; Zhao, S.; Liu, M.; Bartelink, H.; Pierce, L.J.; Feng, F.Y. Development and Validation of a Novel Radiosensitivity Signature in Human Breast Cancer. Clin. Cancer Res. 2015, 21, 3667–3677. [Google Scholar] [CrossRef] [Green Version]
  83. Niméus-Malmström, E.; Krogh, M.; Malmström, P.; Strand, C.; Fredriksson, I.; Karlsson, P.; Nordenskjöld, B.; Stål, O.; Östberg, G.; Peterson, C.; et al. Gene expression profiling in primary breast cancer distinguishes patients developing local recurrence after breast-conservation surgery, with or without postoperative radiotherapy. Breast Cancer Res. 2008, 10, R34. [Google Scholar] [CrossRef] [Green Version]
  84. Tramm, T.; Mohammed, H.; Myhre, S.; Kyndi, M.; Alsner, J.; Børresen-Dale, A.-L.; Sørlie, T.; Frigessi, A.; Overgaard, J. Development and Validation of a Gene Profile Predicting Benefit of Postmastectomy Radiotherapy in Patients with High-Risk Breast Cancer: A Study of Gene Expression in the DBCG82bc Cohort. Clin. Cancer Res. 2014, 20, 5272–5280. [Google Scholar] [CrossRef] [Green Version]
  85. Sjöström, M.; Chang, S.L.; Fishbane, N.; Davicioni, E.; Zhao, S.G.; Hartman, L.; Holmberg, E.; Feng, F.Y.; Speers, C.W.; Pierce, L.J.; et al. Clinicogenomic Radiotherapy Classifier Predicting the Need for Intensified Locoregional Treatment After Breast-Conserving Surgery for Early-Stage Breast Cancer. J. Clin. Oncol. 2019, 37, 3340–3349. [Google Scholar] [CrossRef]
  86. Eschrich, S.A.; Fulp, W.J.; Pawitan, Y.; Foekens, J.A.; Smid, M.; Martens, J.W.; Echevarria, M.; Kamath, V.; Lee, J.-H.; Harris, E.E.; et al. Validation of a radiosensitivity molecular signature in breast cancer. Clin. Cancer Res. 2012, 18, 5134–5143. [Google Scholar] [CrossRef] [Green Version]
  87. Torres-Roca, J.F.; Eschrich, S.; Zhao, H.; Bloom, G.; Sung, J.; McCarthy, S.; Cantor, A.B.; Scuto, A.; Li, C.; Zhang, S.; et al. Prediction of radiation sensitivity using a gene expression classifier. Cancer Res. 2005, 65, 7169–7176. [Google Scholar] [CrossRef] [Green Version]
  88. Torres-Roca, J.F.; Fulp, W.J.; Caudell, J.J.; Servant, N.; Bollet, M.A.; van de Vijver, M.; Naghavi, A.O.; Harris, E.E.; Eschrich, S.A. Integration of a Radiosensitivity Molecular Signature Into the Assessment of Local Recurrence Risk in Breast Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 631–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Eschrich, S.; Zhang, H.; Zhao, H.; Boulware, D.; Lee, J.-H.; Bloom, G.; Torres-Roca, J.F. Systems Biology Modeling of the Radiation Sensitivity Network: A Biomarker Discovery Platform. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 497–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Eschrich, S.A.; Pramana, J.; Zhang, H.; Zhao, H.; Boulware, D.; Lee, J.-H.; Bloom, G.; Rocha-Lima, C.; Kelley, S.; Calvin, D.P.; et al. A Gene Expression Model of Intrinsic Tumor Radiosensitivity: Prediction of Response and Prognosis after Chemoradiation. Int. J. Radiat. Oncol. Biol. Phys. 2009, 75, 489–496. [Google Scholar] [CrossRef] [PubMed]
  91. Scott, J.G.; Berglund, A.; Schell, M.J.; Mihaylov, I.; Fulp, W.J.; Yue, B.; Welsh, E.; Caudell, J.J.; Ahmed, K.; Strom, T.S.; et al. A genome-based model for adjusting radiotherapy dose (GARD): A retrospective, cohort-based study. Lancet Oncol. 2017, 18, 202–211. [Google Scholar] [CrossRef]
  92. Shen, J.; Yan, D.; Bai, L.; Geng, R.; Zhao, X.; Li, H.; Dong, Y.; Cao, J.; Tang, Z.; Liu, S.-B. An 11-Gene Signature Based on Treatment Responsiveness Predicts Radiation Therapy Survival Benefit Among Breast Cancer Patients. Front. Oncol. 2022, 11, 5565. [Google Scholar] [CrossRef]
  93. Cui, Y.; Li, B.; Pollom, E.L.; Horst, K.C.; Li, R. Integrating Radiosensitivity and Immune Gene Signatures for Predicting Benefit of Radiotherapy in Breast Cancer. Clin. Cancer Res. 2018, 24, 4754–4762. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Vrieling, C.; Werkhoven, E.v.; Maingon, P.; Poortmans, P.; Weltens, C.; Fourquet, A.; Schinagl, D.; Oei, B.; Rodenhuis, C.C.; Horior, J.-C.; et al. Prognostic Factors for Local Control in Breast Cancer After Long-term Follow-up in the EORTC Boost vs No Boost Trial A Randomized Clinical Trial. JAMA Oncol. 2017, 3, 42–48. [Google Scholar] [CrossRef]
Figure 1. Schematic illustrating the use of a generic genomic assay to help with the process of staging and subtyping breast cancer but also with the ability to provide predictive information with regard to radiation therapy and chemotherapy.
Figure 1. Schematic illustrating the use of a generic genomic assay to help with the process of staging and subtyping breast cancer but also with the ability to provide predictive information with regard to radiation therapy and chemotherapy.
Biomedicines 11 00098 g001
Table 1. Prognostic genomic assays in breast cancer.
Table 1. Prognostic genomic assays in breast cancer.
MicroarrayNumber of GenesGenesClassificationReference
OncotypeDx21 genes16 cancer-related genes associated with proliferation, invasion, and estrogen signaling as well as 5 reference genesGenerates “Recurrence Score” (RS), from 0–100 which stratifies patients into low (0–18), intermediate (18–31), and high (31–100) risk of distant reccurence.[43]
Mammoprint70 genesGenes associated with cell cycle, invasion, metastases and signal transductionClassifies breast cancer patients into “poor prognosis” or “good prognosis”. Calculates MammaPrint Index (MPI) to stratify Low or High risk of reccurence.[54,55]
PAM 5050 genes50 discriminator genes + 8 controls. These genes identify the intrinsic breast cancer subtypes by indentifying the underlying biology associated with ER and HER2 pathways, and proliferation genes and markers of the basal phenotype.Calculates a “risk-of-recurrence” (ROR) score, 0–100 which classifies low (0–40), intermediate (41–60), or high (61–100) risk for node-negative cancers.[68,69,70]
Table 2. Current prospective clinical trials evaluating whether prognostic gene assays can identify low risk ER+ early breast cancer patients in which radiation therapy can be safely omitted.
Table 2. Current prospective clinical trials evaluating whether prognostic gene assays can identify low risk ER+ early breast cancer patients in which radiation therapy can be safely omitted.
Prospective Clinical TrialsEnrollment CriteriaAimsReference
LUMINAGrade 1–2 invasive breast cancers of luminal A subtype (defined as: ER ≥ 1%, PR > 20%, HER2 negative and Ki67 ≤ 13.25%) in women > 55 yoTo evaluate patients who have low risk of local reccurence following breast conservation surgery and endocrine therapy alone who may be candidates for radiation omission.[26,27]
IDEAER+ invasive breast cancers with RS ≤ 18 who plan to receive endocrine therapy in post-menopausal womenTo evaluate locoregional recurrence rates in low risk women via considering tumor biology including OncotypeDx, in which radiation therapy can be omitted after breast conservation surgery.[71]
PRECISIONER+ early stage breast cancers < 2 cm with a low risk PAM-50-based ROR in women > 50 yoTo evaluate locoregional recurrence rates in patients omitting radiation treatment after lumpectomy.[72]
DEBRAER+/Her-2 negative early stage breast cancers with RS ≤ 18 in women age 50–69 yoTo evaluate omission of breast radiation after breast conservation surgery and endocrine therapy.[73]
EXPERTER+/Her-2 negative early stage breast cancers with PAM-50 ROR < 60 in women ≥ 50 yoTo evaluate the omission of breast radiation therapy compared to obseravation after breast conserving surgery and endocrine therapy.[75]
TAILOR-RTER+/Her2-negative biomarker low risk breast cancers (defined as RS ≤ 25) with low lymph node burden (1–3+ lymph nodes)To evaluate the effects on low risk breast cancer patients after ommission or treatment of regional nodal radiation.[74]
Table 3. Summary of gene expression profiles found to be associated with radiation resistance in breast cancer.
Table 3. Summary of gene expression profiles found to be associated with radiation resistance in breast cancer.
MicroarrayNumber of GenesGenesClassificationReference
Speers et al.51 genesEnriched with genes associated with cell cycle arrest and DNA damage responseIdentify women at an increased risk of local recurrence most likely to benefit from radiation[78]
Danish Breast Cancer Cooperative Group (DBCG82bc)7 genesHLA-DQA, RGS1, DNALI1, hCG2023290, IGKC, OR8G2, and ADH1B. Genes involving immune system, protein signalling, and metabolism enzymesIdentified patients with “high LRR risk” in which PMRT significantly reduced the risk of LR and “low LRR risk” group in which PMRT showed no additional reduction in LR rate and the response to radiation was independent of intrinsic molecular subtype[77,80]
Adjuvant Radiotherapy Intensification Classifier (ARTIC)27 genesGenes related to cell proliferation, cell cycle and kinase activity as well as patient ageA gene classifier for radiation sensitivity in patients with high-risk node negative early stage breast cancers was developed using three publicly available cohorts[81]
RSI radiosensitivity index10 genesRelated to DNA damage response, histone deacetylation, cell cycle, apoptosis and proliferationPredict clinical outcomes and benefit from radiation therapy among several cancer types, including breast cancer[82,83,84,85,86]
Shen et al.11 genesGenes that look at the tumor immune micro-environmentStratify tumors based on response to radiation therapy and the tumor immune microenvironment[88]
Cui et al33 radiation-related genesTumor microenvironment genes such as tumor-associated antigens on the major histocompatibility complex (MHC) molecules.Stratification of patients by predicting benefits from radiotherapy.[89]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Beyer, S.J.; Tallman, M.; Jhawar, S.R.; White, J.R.; Bazan, J.G. The Prognostic and Predictive Value of Genomic Assays in Guiding Adjuvant Breast Radiation Therapy. Biomedicines 2023, 11, 98. https://doi.org/10.3390/biomedicines11010098

AMA Style

Beyer SJ, Tallman M, Jhawar SR, White JR, Bazan JG. The Prognostic and Predictive Value of Genomic Assays in Guiding Adjuvant Breast Radiation Therapy. Biomedicines. 2023; 11(1):98. https://doi.org/10.3390/biomedicines11010098

Chicago/Turabian Style

Beyer, Sasha J., Miranda Tallman, Sachin R. Jhawar, Julia R. White, and Jose G. Bazan. 2023. "The Prognostic and Predictive Value of Genomic Assays in Guiding Adjuvant Breast Radiation Therapy" Biomedicines 11, no. 1: 98. https://doi.org/10.3390/biomedicines11010098

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop