Abstract
Objective
Multigene predictors are being used increasingly in early stage breast cancer patients for prediction and prognosis. However, one consequence of the increased use of multigene predictors, and the heightened efforts towards their incorporation into routine clinical practice, is the potential for future malpractice litigation. It is therefore important to ascertain the strength of the evidence for using the different commercially available multigene predictor assays clinically. We evaluated the literature for evidence of clinical validity of four currently available gene signatures, and to assess the influence of the 21-gene-expression assay Oncotype DX® (Genomic Health Inc., Redwood City, CA, USA) on changes in treatment recommendations.
Methods
A systematic search of the peer-reviewed literature from January 2002 to March 2014 for multigene predictor assays was carried out, and a meta-analysis was conducted.
Results
The adjusted Cox hazard ratio average for studies that met eligibility criteria was 3.538 (95% CI: 1.513 – 8.469). The 21-gene signature showed the highest stability in the estimation of likelihood of distant risk of recurrence. Using recurrence scores resulted in changes in treatment recommendations in 31.8% of all patients in the studies.
Conclusion
This study may provide insight about the use of multigene predictors in clinical practice for prediction and prognosis in breast cancer.
Keywords: Multi-gene predictors, gene-expression profiling, breast cancer, recurrence score, Cox hazard ratios, meta-analysis
INTRODUCTION
Although advances in molecular diagnostics and therapeutic interventions have led to declines in breast cancer mortality, particularly over the last 15 years, breast cancer remains a frequently diagnosed cancer among women [1]. It has been estimated that a total of 232,670 new cases will be diagnosed in the United States in 2014 [1].
A key challenge for physicians and their patients in the management of breast cancer in making decisions about treatment options relates to the use of adjuvant chemotherapy [2,3] following surgical resection of the tumor. Adjuvant chemotherapy has been shown to increase recurrence-free and overall survival in early-stage patients as a whole [4], but it is difficult to identify the patients who would not have experienced recurrence in the first place and do not need chemotherapy. The effectiveness of these interventions are based on risk stratification of patients and has involved the use of validated computer-based models using algorithms designed to calculate the rates for recurrence of the disease such as the Adjuvant! Online (www.adjuvantonline.com) developed using Surveillance, Epidemiology, and End Results (SEER) registry data; the Nottingham Prognostic Index - NPI [5]; and/or the utilization of panel recommendations or clinical guidelines issued by scientific or professional organizations such as the St. Gallen Consensus [6] and the National Comprehensive Cancer Network - NCCN [7].
In more recent years, a number of molecular multigene predictor gene expression profiling (GEP) assays have been developed and increasingly used to identify early-stage breast cancer patients who are most likely to benefit from adjuvant chemotherapy [8].
As a consequence of the increased use of multigene predictors and other molecular companion diagnostics, clinicians, policy-makers and other decision-makers are working towards developing and managing healthcare systems that incorporate personalized genomic medicine into routine clinical practice. This increased use of multigene predictors and other molecular diagnostics thus has the potential to affect future malpractice litigation risks [9,10].
It is therefore important to ascertain: (1) the strength of the evidence for using the different commercially available GEP assays based upon whether these tests provide clinically valid and useful information; and, (2) the influence of the 21-gene-expression assay, commercially marketed as Oncotype DX® (Genomic Health Inc., Redwood City, CA, USA) on physician treatment recommendations, since this is the most frequently used multi-gene predictor GEP assay in the US.
Several studies have been published evaluating the use of the Oncotype DX® recurrence score (RS) on physician recommendations for treatment options (adjuvant chemotherapy and/or hormonal therapy) [11], or whether the gene signatures of interest were comparable in terms of their clinically validity of the estimations for risk of recurrence. However, these studies primarily evaluated how knowledge of the RS affects management of treatment for eligible early-stage breast cancer patients, and did not analyze gene signature adjusted multivariate Cox hazard ratios to allow more direct comparisons of the prognostic value of the gene signature for the likelihood of 10-year relapse-free survival and overall survival of breast cancer between published reports. We systematically reviewed the peer-reviewed published literature and conducted a meta-analysis in order to determine whether (a) sufficient evidence exists in the literature to substantiate the clinical validity of gene-expression profiling to predict recurrence; and, (b) assess the influence of the 21-gene-expression assay on changes in physician treatment recommendations. Given the increasing need for clinicians and patients, and potentially representatives of the legal community, to interpret findings from the literature to make informed decisions about the use of gene expression profiling assays in breast cancer care, we conducted a systematic review of the literature and meta-analysis aimed at summarizing and synthesizing the current evidence.
METHODS
Study Identification and Selection
We systematically searched the peer-reviewed literature from January 2002 through March 2014 for multigene predictor assays. Searches were performed using several databases including PubMed Medline, Ovid (including Medline and Journals@Ovid Full Text) and EMBASE for studies of multigene predictors of clinical outcome in breast cancer. We independently reviewed a list of titles and abstracts for relevance to identify any articles that describe either a validation of any gene-expression profiling assay for early-stage breast cancer and/or the use of the RS to make or change treatment recommendations in breast cancer. Articles reviewed were limited to those published in the English language, and were excluded if they lacked relevance to gene-expression profiling for breast cancer or did not deal with human populations. The full text of any article that passed this initial screening was retrieved for further review. Full articles were also retrieved for any articles for which it was not possible to determine relevance from the abstract in order to ensure a comprehensive search. The references of relevant published articles were also manually searched to identify other potentially relevant articles.
Studies were included for further review if they met the following criteria: original research studies published in peer-reviewed journals; studies focusing on validation of any studies on any of the specific gene-signatures; studies of clinical utilization of the 21-gene expression assay (Oncotype DX®; Genomic Health, Redwood City, CA) recurrence score to make treatment recommendations for patients. Reviews of the literature or meta-analyses that did not include original research were excluded.
Data Extraction and Analysis
Data from each of the articles that were selected for further review was abstracted on to a structured abstraction form. Abstracted data included: a) authors; b) year of publication; c) total number of study participants; d) gene signature adjusted multivariate Cox model hazard ratio and respective 95% confidence intervals; e) total number of patients with changes using RS for treatment recommendations; g) total number of patients changing from a combination of chemotherapy (CHT) plus hormone therapy (HT), CHT-HT, to HT alone or h) from HT alone to a combination of CHT-HT. Data was also extracted about i) the type of study (prospective or retrospective); j) which clinical practice guidelines (if this information was available) and/or algorithm-based estimators of risk (such as Adjuvant! Online and/or the Nottingham Prognostic Index), were used for initial treatment recommendations.
The information available for adjusted multivariate Cox hazard ratios and their 95% confidence intervals (CI) for different gene-signatures was pooled, averaged by gene signature and weighted (using a random effects model). Statistical heterogeneity was calculated using the χ2 test for heterogeneity and the I2 percentage. A probability level for the χ2 statistic less than or equal to 10% (p≤0.10) and/or an I2 greater than 50% would be considered indicative of statistical heterogeneity.
Data about differences between treatment recommendations before and after the availability of recurrence scores was taken directly from the reviewed articles, pooled, and averaged to ascertain how RS were used to change initial treatment recommendations. Effect sizes for differences in treatment recommendations were calculated using Cohen's d and the two-tailed correlation coefficient, rY, using means and standard deviations of two independent groups of equal size. Data were analyzed using IBM® SPSS® Statistics Release 21, (Armonk, New York).
RESULTS
Our initial broad search of the literature yielded 590 articles (figure 1). Of the remaining 52 studies that met eligibility criteria, twenty-five studies were eligible for a meta-analysis of the gene signature associated standardized multivariate Cox hazard ratios. Table 1 summarizes the characteristics of the included studies. These 25 studies used four gene-signatures: Mammaprint® (44%; n=11); Mammostrat® (8.0%; n=2); Oncotype DX® (24.0%; n=6); and the Rotterdam signature (24.0%; n=6). The sample size of these studies ranged between 86 to 4,598 subjects [12,13] with a mean of 702 patients enrolled. We contrasted the prognostic value of the likelihood of 10-year relapse-free survival and overall survival of breast cancer by gene signatures included in this meta-analysis by comparing the reports of adjusted multivariate Cox hazards ratios. The adjusted Cox hazard ratio average for the series of 25 studies was 3.538 (95% CI: 1.513 – 8.469; table 1). Point estimates and their 95% confidence intervals showed some degree of overlap. In our analysis, the 21-gene signature Oncotype DX® showed the highest stability in the estimation of likelihood (Mean: 2.70; 95% CI: 1.56-4.72). Around this range of variation in the adjusted multivariate Cox hazards ratios, the closest one to Oncotype DX® was for Mammaprint® (Mean: 3.43; 95% CI: 1.43-8.64), followed by Mammostrat® (1.61; 95% CI: 1.30-2.02). The Rotterdam 76-gene signature showed the farthest variation overall (Mean: 5.21; 95% CI: 18.50) (Figure 2).
Figure 1.
Search strategy flowchart.
Table 1.
Weighted Adjusted Multivariate Cox Hazard Ratio (HR) and 95% Confidence Intervals (CI)
| Study | Patient Enrollment | Cox HR | SE | Weighted CI lower | Weighted CI upper |
|---|---|---|---|---|---|
| Mook et al, 2010 [41] | 964 | 3.450 | 0.050 | 3.333 | 3.567 |
| Knauer et al, 2010 [42] | 541 | 0.210 | 0.020 | 0.171 | 0.249 |
| Kok et al, 2009 [31] | 364 | 2.210 | 0.078 | 2.057 | 2.363 |
| Buyse et al, 2006 [43] | 307 | 1.500 | 0.067 | 1.363 | 1.637 |
| van de Vijver et al, 2002 [44] | 295 | 4.600 | 0.125 | 4.355 | 4.845 |
| Bueno-de-Mesquita et al, 2009 [45] | 274 | 5.400 | 0.142 | 5.125 | 5.675 |
| Mook et al, 2009 [46] | 241 | 2.990 | 0.111 | 2.772 | 3.208 |
| Saghatchian et al, 2013 [47] | 173 | 2.698 | 0.124 | 2.453 | 2.943 |
| Knauer et al, 2010 [48] | 168 | 5.780 | 0.185 | 5.416 | 6.144 |
| Glas et al, 2006 [49] | 162 | 5.600 | 0.186 | 5.236 | 5.964 |
| Mook et al, 2009 [50] | 148 | 4.400 | 0.172 | 4.062 | 4.738 |
| Avg. Mammaprint | 330.6 | 3.531 | 0.115 | 3.304 | 3.757 |
| Bartlett et al, 2012 [13] | 4,598 | 1.910 | 0.020 | 1.870 | 1.950 |
| Ross et al, 2008 [55] | 1,294 | 1.400 | 0.032 | 1.336 | 1.464 |
| Avg. Mammostrat ® | 2,946.0 | 1.655 | 0.026 | 1.603 | 1.707 |
| Partin et al, 2011 [18] | 1,674 | 2.160 | 0.036 | 2.090 | 2.230 |
| Gray et al, 2011 [52] | 1,436 | 1.430 | 0.062 | 1.368 | 1.492 |
| Dowsett et al, 2010 [53] | 1,372 | 4.360 | 0.042 | 4.250 | 4.470 |
| Tang et al, 2011 [21] | 1,319 | 2.370 | 0.052 | 2.287 | 2.453 |
| Paik et al, 2006 [54] | 651 | 1.780 | 0.086 | 1.678 | 1.882 |
| Albain et al, 2010 [38] | 367 | 2.640 | 0.151 | 2.474 | 2.806 |
| Avg. Oncotype Dx | 1,136.5 | 2.457 | 0.071 | 2.358 | 2.556 |
| Zhang et al, 2009 [55] | 300 | 6.130 | 0.143 | 5.850 | 6.410 |
| Wang et al, 2005 [56] | 286 | 5.550 | 0.139 | 5.277 | 5.823 |
| Desmedt et al, 2008 [20] | 198 | 5.110 | 0.161 | 4.795 | 5.425 |
| Desmedt et al, 2007 [57] | 198 | 2.800 | 0.119 | 2.567 | 3.033 |
| Foekens et al, 2006 [58] | 180 | 11.360 | 0.251 | 2.670 | 48.400 |
| Prat et al, 2012 [59] | 1,380 | 1.490 | 0.015 | 1.426 | 1.554 |
| Avg. Rotterdam 76 | 423.7 | 5.407 | 0.138 | 3.764 | 11.774 |
| Effect Summary | 1,209.2 | 3.262 | 0.088 | 2.757 | 4.949 |
Figure 2. Gene Signature Reported Adjusted Multivariate Cox Hazard Ratios and 95% confidence intervals.

Forest plots showing disease-free survival. Squares: Reported hazard ratios from each study; Diamonds: Each gene signature's average and the overall average of the hazard ratio.
Our search of the literature for evidence of clinical utility of GEP in breast cancer care ultimately yielded 27 studies, all focused on the 21-gene expression-profiling assay, Oncotype DX®. We limited our analysis to studies that reported changes in treatment recommendations from initial recommendations based on using the Oncotype DX® RS. Twenty-seven studies reporting changes in initial treatment recommendations following the use of RS, met eligibility criteria (Table 2), and included 6,655 patients with a mean of 246.5 participants per study (SD: 377.2; 95% CI: 104.2 – 388.8). We found wide variation in the number of participants, from the largest sample of 1,822 patients [14] to the smallest sample of 22 patients[15]. More than two thirds of the studies (70.4%; n=19) were published between 2011 and 2014. Three-fourths of the studies were retrospective (74.1%; n=20). Studies reported that initial treatment recommendations for adjuvant chemotherapy were formulated following clinical practice guidelines endorsed by professional associations, mainly NCCN (35.0%; n=14), St. Gallen Consensus (15.0%; n=6) or ASCO (12.5%; n=5). Several studies reported using algorithms for risk estimation such as Adjuvant! Online (17.5%; n=7) and the Nottingham Prognostic Index (5.0%; n=2).
Table 2.
Proportional Changes to Treatment Recommendations following Utilization of the 21-gene signature Recurrence Score (RS®)
| Study | Type of Study | Country of Pub. | Number of cases/Study Participants | Guidelines or Decision Algorithm Used Pre-RS | Receipt of Adjuvant Therapy | ||
|---|---|---|---|---|---|---|---|
| % Change CT-HT to HT | % Change HT to CT-HT | % Change Total | |||||
| Oratz et al 2007 [60] | Retrospective | USA | 68 | AO | 25.0 | 25.0 | 25.0 |
| Erb et al 2007 [61] | Retrospective | USA | 124 | NCCN | 30.0 | n.a. | 30.0 |
| Lo et al 2010 [25] | Prospective | USA | 89 | n.a. | 22.5 | 3.4 | 25.9 |
| Asad et al 2008 [35] | Retrospective | USA | 85 | NCCN | 38.8 | 4.7 | 43.5 |
| Henry et al 2009 [62] | Retrospective | USA | 29 | AO | 25.9 | 7.4 | 33.3 |
| Geffen et al 2009 [63] | Retrospective | Israel | 25 | AO | 24.0 | 12.0 | 36.0 |
| Klang et al 2010 [64] | Retrospective | Israel | 313 | NCCN & ASCO | 33.5 | 6.4 | 39.9 |
| Ademuyiwa et al 2011 [65] | Retrospective | USA | 276 | AO & NPI | 31.9 | 6.2 | 38.0 |
| Partin et al 2011 [18] | Retrospective | USA | 169 | St Gallen; NCCN & AO | 71.0 | 3.0 | 74.0 |
| Oratz et al 2011 [66] | Prospective | USA | 89 | NCCN & ASCO | 35.0 | n.a. | 35.0 |
| Joh et al 2011 [67] | Retrospective | USA | 154 | NCCN & ASCO | 24.9 | n.a. | 24.9 |
| Haas et al 2011 [29] | Retrospective | USA | 364 | NCCN | 61.8 | 0.3 | 62.1 |
| Hassett et al 2012 [16] | Retrospective | USA | 1,111 | St Gallen & NCCN | 9.6 | 8.0 | 17.6 |
| Eiermann et al 2012 [24] | Prospective | Germany | 366 | n.a. | 21.6 | 10.6 | 33.0 |
| Albanell et al 2012 [26] | Retrospective | Spain | 107 | St Gallen & ESMO | 20.6 | 11.2 | 31.8 |
| Kamal et al 2012 [17] | Retrospective | USA | 186 | St Gallen & NCCN | 10.8 | 8.1 | 18.9 |
| Schneider & Khalil 2012 [28] | Retrospective | USA | 89 | NCCN; ASCO & AO | 39.3 | 4.5 | 43.8 |
| Malo et al 2012 [68] | Retrospective | USA | 118 | NCCN | 42.4 | 0.8 | 43.2 |
| deBoer et al 2013 [23] | Prospective | Australia | 151 | St Gallen | 35.8 | 15.5 | 24.5 |
| Chen et al 2013 [14] | Retrospective | USA | 1,822 | NCCN & ASCO | 23.3 | 3.1 | 26.4 |
| Holt et al 2013 [69] | Prospective | United Kingdom | 142 | AO & NPI | 18.3 | 8.5 | 26.8 |
| Stemmer et al 2013 [70] | Retrospective | Israel | 282 | St Gallen | 45.6 | n.a. | 45.6 |
| Davidson et al 2013 [71] | Prospective | Canada | 150 | n.a. | 20.0 | 15.0 | 35.0 |
| Yamauchi et al 2014 [72] | Prospective | Japan | 124 | NCCN & Japanese | 32.3 | 5.6 | 37.9 |
| Fried & Moskovitz et al 2014 [73] | Retrospective | Israel | 111 | n.a. | 11.7 | 9.9 | 21.6 |
| Toole et al 2014 [15] | Retrospective | USA | 22 | n.a. | 27.3 | 9.1 | 36.4 |
AO:Adjuvant! Online; NCCN: National Comprehensive Cancer Network; ASCO: American Society of Clinical Oncology; NPI: Nottingham Prognostic Index; St Gallen: St Gallen International Consortium; CT: Chemotherapy; HT: Hormonal Therapy
Using recurrence scores resulted in changes in treatment recommendations from initial treatment recommendations in 31.8% (n=2,113) of all patients in the studies. A reduction in the administration of chemotherapy but maintenance of hormonal therapy was observed in the majority of study patients (83.3%; n=1,761). However, addition of chemotherapy to hormonal therapy was found in less than one-fifth of the pooled study patients (16.4%; n=346). These changes from the initial treatment recommendations were statistically significant (paired t(26)=5.82, p<0.001, 2-tailed; Cohen's d=2.0; effect size r=0.71.5; coefficient of determination r2=51.1%) suggesting that treatment recommendations based on use of the Oncotype DX® recurrence scores resulted in less administration of chemotherapy. Reported percentages in changes from initial treatment recommendations about systemic adjuvant chemotherapy varied between less than 20% up to 74% of all pooled study patients [16,17,18].
DISCUSSION
We evaluated the evidence in the peer-reviewed literature for whether: (1) the currently available gene-signatures for early-stage breast cancer are associated with the likelihood of distant recurrence of breast cancer based upon reported adjusted multivariate Cox hazard ratios; and (2) the strength of evidence for influence of the Oncotype DX® RS from the 21-gene expression profiling assay on changes in physician treatment recommendations from initial recommendations.
We found that the four gene signatures evaluated appear to be associated with predicting distant recurrence, and point estimates and their 95% confidence intervals showed some degree of overlap. This supports the suggestion by some authors that perhaps combining one or more of the already developed mulitgene predictors will likely provide more precise prognostic and predictive information about recurrence risk with concomitant therapeutic outcomes and one such attempt has been reported [19-21]. In our analysis, the 21-gene signature Oncotype DX® showed the highest stability in the estimation of likelihood of risk of recurrence. This suggests that the estimation was less variable and/or less susceptible to variations in predictors of recurrence, including tumor genomics (e.g. Ki67, HER2 biomarker status). It further suggests the clinical validity of Oncotype DX® as a predictive and prognostic diagnostic for making treatment decisions for breast cancer [22]. Although other authors have identified these gene signatures as predicting clinical outcome [22], to the best of our knowledge, this study is the first to pool together and statistically analyze standardized adjusted Cox hazard ratios, which are estimations of the probability of 10-year recurrence.
Our findings that treatment recommendations were changed based on using the RS in more than 30% of patients in the pooled analysis of studies are consistent with other individual studies which found treatment recommendations changed in some 24% to 30% for women with node-negative cancer following RS use [23-26].
Our analysis demonstrated that changes in treatment recommendations mostly resulted in proportionally reduced administration of chemotherapy in the majority of cases, and less frequently in addition of chemotherapy. This result is not surprising, since chemotherapy is an accepted standard therapeutic intervention in breast cancer [27]. However, prior individual studies had only found changes in treatment recommendations varied from 9.6 to 71 with a mean of 29.8 [28,29]. Our finding may have possible policy-relevant consequences for clinical utilization, testing costs, and potential future liability cases.
Although several reviews have recently evaluated the literature related to multigene predictors of clinical outcome in breast cancer [30,31,32], these did not specifically statistically pool and analyze the proportional changes in treatment recommendations that were made as a result of using recurrence scores.
Our study suggests that GEP tests, and particularly use of the 21-gene assay, Oncotype DX® are increasingly influencing how breast cancer patients are treated.
Although studies that directly show an association with outcomes following use of multigene predictors are limited, evidence is accumulating that tests such as the 21-gene signature (Oncotype DX®) and the 70-gene signature (MammaPrint®) contribute additional prognostic value beyond conventional clinical indicators [26, 33,34, 70]. Thus, treatment recommendations for adjuvant chemotherapy should include gene expression profiling as an integral part of any decision algorithm [19,35].
It is important for clinicians to be aware of how these multigene predictive assays are increasingly being used in practice [36] and of their limitations. In addition to factors such as risk of distant recurrence that affect treatment, clinicians should also consider potential risks as well as patient quality of life. Since this field, and what constitutes standard of care appears to be continually evolving, it is equally important for clinicians to avail themselves of opportunities to learn about the latest clinical practice guidelines and accepted standards in order to avoid any potential litigation [37] even though it maybe premature to consider litigation at this time.
Since the standard of care for multigene predictors is still fraught with uncertainty, this presents with a “damned if you do, damned if you don't” dilemma for physicians [37]. A physician that fails to utilize such a test and recommends against chemotherapy on other grounds may face a lawsuit by a patient with recurring breast cancer who argues after the-fact that the physician was negligent in not relying on a multigene predictor test. Alternatively, if the physician recommends a multigene predictor assay and no chemotherapy based on the results of that test, the physician may face a lawsuit alleging premature reliance on a test that was not yet subsequently validated [37]. In either case, with the advantage of perfect hindsight, the physician could be portrayed by a skilled trial lawyer as having acted too slowly or too quickly to rely on multigene predictor assays.
While uncertainty and rapid change in medical technologies create an inherent liability risk for physicians, the best way to protect against such liability is for the physician to document in the patient's medical record at the time of decision that the physician was aware of the available and uncertain evidence on the clinical utility of GEP assays, and made an informed professional judgment in the face of such uncertainty.
We recognize that policy regarding the use of multigene predictors should ideally be governed by evidence from prospective randomized clinical trials [22, 38]. Until the results of prospective clinical trials such as the TAILOR-X [39] and the MINDACT [40] become available, systematic assessments and meta-analyses such as this study ought to be useful for providing insight to clinicians, policymakers, and for better understanding potential liability in this burgeoning field.
EXPERT COMMENTARY
Multigene predictors, including gene expression profiling, are being used increasingly for risk stratification to identify early stage breast cancer patients who are most likely to benefit from adjuvant chemotherapy. During the past decade, a number of studies have been published focused on the analytical and clinical validity, and clinical utility of at least 4 multigene predictors, of which the most frequently used in US populations is the 21-gene signature Oncotype DX®. Although incorporation of multigene predictor assays into routine clinical breast cancer practice has not yet been fully realized, gene-expression profiling for prediction and prognosis is increasingly being used in clinical settings, with a consequent interest on the part of clinicians and policymakers towards developing and managing health care systems that incorporate personalized genomic medicine as a standard of medical practice. This has the potential to affect future malpractice litigation risks. This study suggests that multigene predictor assays, and particularly use of the 21-gene assay, are increasingly being used in practice. It appears that this trend towards adoption of multigene predictors and other molecular genomic technologies will continue as more advances in technologies for genomic profiling are made. As what constitutes standard of care continues to evolve, it is important for physicians to document in the patient's medical record at the time of decision that the physician was aware of the available and uncertain evidence on the clinical utility of multigene predictor assays, and made an informed professional judgment in the face of such uncertainty.
FIVE-YEAR VIEW
We believe that as multigene predictors and other genomic technologies continue to evolve, it is likely that the potential for malpratice litigation will increase as well. Historically, there are precedents for increased litigation with increased use and adoption of novel technologies, primarily due to increased patient expectations. Thus, we can expect that in the next 5 to 10 years, as genomic technologies such as multigene predictor assays become more routinely integrated into clinical practice and health systems, physicians will likely be dealing with the first wave of malpractice litigation.
KEY ISSUES.
Multigene predictors, including gene expression profiling, are being used increasingly for risk stratification to identify early stage breast cancer patients who are most likely to benefit from adjuvant chemotherapy.
One consequence of the increased use of GEPs and other molecular companion diagnostics, and the heightened efforts towards their incorporation into routine clinical practice, is the potential for future malpractice litigation.
A systematic search of the literature from Jan 2002 to March 2014 and meta-analyses were performed.
The analysis of Cox hazard ratios demonstrated that gene signatures appear to be associated with predicting distant recurrence.
Point estimates and their 95% confidence intervals showed some degree of overlap between the 4 gene signatures assessed, however, the 21-gene signature Oncotype DX® showed the highest stability in the estimation of likelihood of distant risk of recurrence.
Using recurrence scores resulted in changes in treatment recommendations from initial treatment recommendations in 31.8% of all patients in the studies.
The results of this study may provide insight about the use of multigene predictors in clinical practice for prediction and prognosis in breast cancer, and improved understanding of potential liability issues in the field of personalized genomic medicine.
ACKNOWLEDGEMENT
The authors would like to thank Dr. Douglas Campos-Outcalt for comments on an earlier draft of this article.
This work was supported by grant 5R01HG00614502 from the National Human Genome Research Institute (NHGRI) of the National Institutes of Health (NIH).
Footnotes
Financial & competing interests disclosure
The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
References
- 1.Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin. 2014;64(1):9–29. doi: 10.3322/caac.21208. [DOI] [PubMed] [Google Scholar]
- 2.National Comprehensive Cancer Network Clinical Practice Guidelines in Oncology: Breast Cancer. Version 2. 2011 Mar; doi: 10.6004/jnccn.2004.0021. 2011 update. [DOI] [PubMed] [Google Scholar]
- 3.Zeng YC. Chemotherapy in older women with breast cancer. N Engl J Med. 2009;361(10):1023–1024. doi: 10.1056/NEJMc091265. [DOI] [PubMed] [Google Scholar]
- 4.Colleoni M, Gelber RD. Time to Initiation of Adjuvant Chemotherapy for Early Breast Cancer and Outcome: The Earlier, the Better? J Clin Oncol. 2014;32(8):717–719. doi: 10.1200/JCO.2013.54.3942. [DOI] [PubMed] [Google Scholar]
- 5.National Comprehensive Cancer Network (NCCN) NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines)®. Version 3.2013, 05/03/2013©. National Comprehensive Cancer Network, Inc.; 2013. [Google Scholar]
- 6.Goldhirsch A, Winer EP, Coates AS, et al. 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(9):2206–2223. doi: 10.1093/annonc/mdt303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.National Comprehensive Cancer Network (NCCN) NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines)®. Version I.2014.©. National Comprehensive Cancer Network, Inc.; 2014. [Google Scholar]
- 8.Sinn P, Aulmann S, Wirtz R, et al. Multigene Assays for Classification, Prognosis, and Prediction in Breast Cancer: a Critical Review on the Background and Clinical Utility. Geburtshilfe Frauenheilkd. 2013;73(9):932–940. doi: 10.1055/s-0033-1350831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Marchant GE, Lindor RA. Personalized medicine and genetic malpractice. Genet Med. 2013;15(12):921–922. doi: 10.1038/gim.2013.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Marchant GE, Lindor RA. Prudent precaution in clinical trials of nanomedicines. J Law Med Ethics. 2012;40(4):831–840. doi: 10.1111/j.1748-720X.2012.00711.x. [DOI] [PubMed] [Google Scholar]
- 11.Milburn M, Rosman M, Mylander C, Tafra L. Is oncotype DX recurrence score (RS) of prognostic value once HER2-positive and. low-ER expression patients are removed? Breast J. 2013;19(4):357–364. doi: 10.1111/tbj.12126. [DOI] [PubMed] [Google Scholar]
- 12.Cobleigh MA, Tabesh B, Bitterman P, et al. Tumor gene expression and prognosis in breast cancer patients with 10 or more positive lymph nodes. Clin Cancer Res. 2005;11(24 Pt 1):8623–8631. doi: 10.1158/1078-0432.CCR-05-0735. [DOI] [PubMed] [Google Scholar]
- 13.Bartlett JMS, Bloom KJ, Piper T, et al. Mammostrat as an immunohistochemical multigene assay for prediction of early relapse risk in the tamoxifen versus exemestane adjuvant multicenter trial pathology study. J. Clin. Oncol. 2012;30(36):4477–4484. doi: 10.1200/JCO.2012.42.8896. [DOI] [PubMed] [Google Scholar]
- 14.Chen BC, Dhanda R, Tseng W, Forsyth M, Patt DA. Evaluating Use Characteristics for the Onco type Dx 21-Gene Recurrence Score and Concordance With Chemotherapy Use in Early-Stage Breast Cancer. J Oncol Pract. 2013;9(4):182–187. doi: 10.1200/JOP.2012.000638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Toole MJ, Kidwell KM, Van Poznak C. Breast Cancer : Basic and Clinical Research Oncotype Dx Results in Multiple Primary Breast Cancers. Breast Cancer (Auckl) 2014;8:1–6. doi: 10.4137/BCBCR.S13727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hassett MJ, Silver SM, Hughes ME, et al. Adoption of gene expression profile testing and association with use of chemotherapy among women with breast cancer. J. Clin. Oncol. 2012;30(18):2218–26. doi: 10.1200/JCO.2011.38.5740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kamal AH1, Loprinzi CL, Reynolds C, et al. Breast medical oncologists' use of standard prognostic factors to predict a 21-gene recurrence score. Oncologist. 2011;16(10):1359–66. doi: 10.1634/theoncologist.2011-0048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Partin JF, Mamounas EP. Impact of the 21-gene recurrence score assay compared with standard clinicopathologic guidelines in adjuvant therapy selection for node-negative, estrogen receptor-positive breast cancer. Ann. Surg. Oncol. 2011;18(12):3399–406. doi: 10.1245/s10434-011-1698-z. [DOI] [PubMed] [Google Scholar]
- 19.Arpino G, Generali D, Sapino A, et al. Gene expression profiling in breast cancer: a clinical perspective. Breast. 2013;22(2):109–120. doi: 10.1016/j.breast.2013.01.016. [DOI] [PubMed] [Google Scholar]
- 20.Desmedt C, Haibe-Kains B, Wirapati P, et al. Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin. Cancer Res. 2008;14(16):5158–65. doi: 10.1158/1078-0432.CCR-07-4756. [DOI] [PubMed] [Google Scholar]
- 21.Tang G, Shak S, Paik S, et al. 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(1):133–42. doi: 10.1007/s10549-010-1331-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ishibe N1, Schully S, Freedman A, Ramsey SD. Use of Oncotype DX in Women with Node-Positive Breast Cancer. PLoS Curr. 2011;3:RRN1249. doi: 10.1371/currents.RRN1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.de Boer RH, Baker C, Speakman D, et al. The impact of a genomic assay (Oncotype DX) on adjuvant treatment recommendations in early breast cancer. Med J Aust. 2013;199(3):205–208. doi: 10.5694/mja12.11334. [DOI] [PubMed] [Google Scholar]
- 24.Eiermann W, Rezai M, Kümmel S, et al. The 21-gene recurrence score assay impacts adjuvant therapy recommendations for ER-positive, node-negative and node-positive early breast cancer resulting in a risk-adapted change in chemotherapy use. Ann. Oncol. 2013;24(3):618–24. doi: 10.1093/annonc/mds512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Lo SS, Mumby PB, Norton J, et al. Prospective multicenter study of the impact of the 21-gene recurrence score assay on medical oncologist and patient adjuvant breast cancer treatment selection. J. Clin. Oncol. 2010;28(10):1671–6. doi: 10.1200/JCO.2008.20.2119. [DOI] [PubMed] [Google Scholar]
- 26.Albanell J, González a, Ruiz-Borrego M, et al. Prospective trans GEICAM study of the impact of the 21-gene Recurrence Score assay and traditional clinicopathological factors on adjuvant clinical decision making in women with estrogen receptor-positive (ER+) node-negative breast cancer. Ann. Oncol. 2012;23(3):625–31. doi: 10.1093/annonc/mdr278. [DOI] [PubMed] [Google Scholar]
- 27.Esteva FJ, Hortobagyi GN. Integration of Systemic Chemotherapy in the Management of Primary Breast Cancer. Oncologist. 1998;3(5):300–313. [PubMed] [Google Scholar]
- 28.Schneider JG, Khalil DN. Why does Oncotype DX recurrence score reduce adjuvant chemotherapy use? Breast Cancer Res Treat. 2012;134(3):1125–32. doi: 10.1007/s10549-012-2134-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Haas JS, Liang SY, Hassett MJ, Shiboski S, Elkin EB, Phillips KA. Gene expression profile testing for breast cancer and the use of chemotherapy, serious adverse effects, and costs of care. Breast Cancer Res Treat. 2011;130(2):619–26. doi: 10.1007/s10549-011-1628-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Marchionni L, Wilson RF, Wolff AC. Systematic review: gene expression profiling assays in early-stage breast cancer. Ann Intern Med. 2008;148(5):358–369. doi: 10.7326/0003-4819-148-5-200803040-00208. [DOI] [PubMed] [Google Scholar]
- 31.Kok M, Linn SC, Van Laar RK, et al. Comparison of gene expression profiles predicting progression in breast cancer patients treated with tamoxifen. Breast Cancer Res.Treat. 2009;113(2):275–283. doi: 10.1007/s10549-008-9939-y. [DOI] [PubMed] [Google Scholar]
- 32.Carlson JJ, Roth JA. The impact of the Oncotype Dx breast cancer assay in clinical practice: a systematic review and meta-analysis. Breast Cancer Res Treat. 2013;141(1):13–22. doi: 10.1007/s10549-013-2666-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Mamounas EP, Tang G, Paik S, et al. Prognostic impact of the 21-gene recurrence score on disease-free and overall survival of node positive, ER-positive patients with breast cancer treated with adjuvant chemotherapy: Results from NSABP B-28.. Presented at : 2012 Breast Cancer Symposium; San Francsco, CA, USA. 13 September - 15 September 2012. [Google Scholar]
- 34.Drukker CA, Bueno-de-Mesquita JM, Retèl VP, et al. A prospective evaluation of a breast cancer prognosis signature in the observational RASTER study. Int J Cancer. 2013;133(4):929–936. doi: 10.1002/ijc.28082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Asad J, Jacobson AF, Estabrook A, et al. Does oncotype DX recurrence score affect the management of patients with early-stage breast cancer? Am. J. Surg. 2008;196(4):527–9. doi: 10.1016/j.amjsurg.2008.06.021. [DOI] [PubMed] [Google Scholar]
- 36.Mukherjee A, Rakha EA. Integrating breast cancer genetics into clinical practice. Womens Health (Lond Engl) 2012;8(1):99–112. doi: 10.2217/whe.11.81. [DOI] [PubMed] [Google Scholar]
- 37.Marchant GE, Lindor RA, Campos-Outcalt DE. Physician liability: the next big thing for personalized medicine?. Personalized Med. 2011;8(4):457–467. doi: 10.2217/pme.11.33. [DOI] [PubMed] [Google Scholar]
- 38.Albain KS, Barlow WE, Shak S, 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(1):55–65. doi: 10.1016/S1470-2045(09)70314-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Sparano JA. TAILORx: trial assigning individualized options for treatment (Rx). Clin Breast Cancer. 2006;7(4):347–50. doi: 10.3816/CBC.2006.n.051. [DOI] [PubMed] [Google Scholar]
- 40.Viale G, Slaets L, Bogaerts J, et al. High concordance of protein (by IHC), gene (by FISH; HER2 only), and microarray readout (by TargetPrint) of ER, PgR, and HER2: results from the EORTC 10041/BIG 03-04 MINDACT trial. Ann Oncol. 2014;25(4):816–823. doi: 10.1093/annonc/mdu026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Mook S, Knauer M, Bueno-de-Mesquita JM, et al. Metastatic potential of T1 breast cancer can be predicted by the 70-gene MammaPrint signature. Ann Surg Oncol. 2010;17(5):1406–1413. doi: 10.1245/s10434-009-0902-x. [DOI] [PubMed] [Google Scholar]
- 42.Knauer M, Mook S, Rutgers EJ, et al. The predictive value of the 70-gene signature for adjuvant chemotherapy in early breast cancer. Breast Cancer Res Treat. 2010;120(3):655–661. doi: 10.1007/s10549-010-0814-2. [DOI] [PubMed] [Google Scholar]
- 43.Buyse M, Loi S, van't Veer L, Viale G, 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(17):1183–1192. doi: 10.1093/jnci/djj329. [DOI] [PubMed] [Google Scholar]
- 44.Van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347(25):1999–2009. doi: 10.1056/NEJMoa021967. [DOI] [PubMed] [Google Scholar]
- 45.Bueno-de-Mesquita JM, Linn SC, Keijzer R, et al. Validation of 70-gene prognosis signature in node-negative breast cancer. Breast Cancer Res Treat. 2009;117(3):483–95. doi: 10.1007/s10549-008-0191-2. [DOI] [PubMed] [Google Scholar]
- 46.Mook S, Schmidt MK, Weigelt B, et al. The 70-gene prognosis signature predicts early metastasis in breast cancer patients between 55 and 70 years of age. Ann Oncol. 2010;21(4):717–22. doi: 10.1093/annonc/mdp388. [DOI] [PubMed] [Google Scholar]
- 47.Saghatchian M, Mook S, Pruneri G, et al. Additional prognostic value of the 70-gene signature (MammaPrint(®)) among breast cancer patients with 4-9 positive lymph nodes. Breast. 2013;22(5):682–690. doi: 10.1016/j.breast.2012.12.002. [DOI] [PubMed] [Google Scholar]
- 48.Knauer M, Cardoso F, Wesseling J, et al. Identification of a low-risk subgroup of HER-2-positive breast cancer by the 70-gene prognosis signature. Br J Cancer. 2010;103(12):1788–1793. doi: 10.1038/sj.bjc.6605916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Glas AM, Floore A, Delahaye LJ, et al. Converting a breast cancer microarray signature into a high-throughput diagnostic test. BMC Genomics. 2006;7:278. doi: 10.1186/1471-2164-7-278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mook S, Schmidt MK, Viale G, et al. The 70-gene prognosis-signature predicts disease outcome in breast cancer patients with 1-3 positive lymph nodes in an independent validation study. Breast Cancer Res Treat. 2009;116(2):295–302. doi: 10.1007/s10549-008-0130-2. [DOI] [PubMed] [Google Scholar]
- 51.Ross DT, Kim CY, Tang G, et al. Chemosensitivity and stratification by a five monoclonal antibody immunohistochemistry test in the NSABP B14 and B20 trials. Clin Cancer Res. 2008;14(20):6602–6609. doi: 10.1158/1078-0432.CCR-08-0647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gray RG, Quirke P, Handley K, et al. Validation study of a quantitative multigene reverse transcriptase-polymerase chain reaction assay for assessment of recurrence risk in patients with stage II colon cancer. J Clin Oncol. 2011;29(35):4611–4619. doi: 10.1200/JCO.2010.32.8732. [DOI] [PubMed] [Google Scholar]
- 53.Dowsett M, Cuzick J, Wale C, et al. Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study. J Clin Oncol. 2010;28(11):1829–1834. doi: 10.1200/JCO.2009.24.4798. [DOI] [PubMed] [Google Scholar]
- 54.Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006;24(23):3726–3734. doi: 10.1200/JCO.2005.04.7985. [DOI] [PubMed] [Google Scholar]
- 55.Zhang Y, Sieuwerts AM, McGreevy M, et al. The 76-gene signature defines high-risk patients that benefit from adjuvant tamoxifen therapy. Breast Cancer Res Treat. 2009;116(2):303–309. doi: 10.1007/s10549-008-0183-2. [DOI] [PubMed] [Google Scholar]
- 56.Wang Y, Klijn JG, Zhang Y, et al. Gene-expression profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet. 2005;365(9460):671–679. doi: 10.1016/S0140-6736(05)17947-1. [DOI] [PubMed] [Google Scholar]
- 57.Desmedt C, Piette F, Loi S, Wang Y, Lallemand F, Haibe-Kains B, et al. Strong time dependence of the 76-gene prognostic signature for node-negative breast cancer patients in the TRANSBIG multicenter independent validation series. Clin Cancer Res. 2007;13:3207–3214. doi: 10.1158/1078-0432.CCR-06-2765. [DOI] [PubMed] [Google Scholar]
- 58.Foekens JA, Atkins D, Zhang Y, et al. Multicenter validation of a gene expression-based prognostic signature in lymph node-negative primary breast cancer. J Clin Oncol. 2006;24(11):1665–1671. doi: 10.1200/JCO.2005.03.9115. [DOI] [PubMed] [Google Scholar]
- 59.Prat A, Parker JS, Fan C, et al. Concordance among gene expression-based predictors for ER-positive breast cancer treated with adjuvant tamoxifen. Ann Oncol. 2012;23(11):2866–2873. doi: 10.1093/annonc/mds080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Oratz R, Paul D, Cohn AL, et al. Impact of a commercial reference laboratory test recurrence score on decision making in early-stage breast cancer. J Oncol Pract. 2007;3(4):182–186. doi: 10.1200/JOP.0742001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Erb C, Fox KR, Patel M, et al. Evaluation of practice patterns in the treatment of node-negative, hormone-receptor positive breast cancer patients with the use of the oncotype DX assay at the University of Pennsylvania.. Presented at : 30th Annual San Antonio Breast Cancer Symposium; St. Antonio, Texas. 13 December 2007 - 16 December 2007. [Google Scholar]
- 62.Henry LR, Stojadinovic A, Swain SM, et al. The influence of a gene expression profile on breast cancer decisions. J Surg Oncol. 2009;99(6):319–23. doi: 10.1002/jso.21244. [DOI] [PubMed] [Google Scholar]
- 63.Geffen DB, Amir N, Sion-Vardy N, et al. Stage I breast cancer in a regional oncology practice in Israel 2002-2006: clinicopathologic features, risk estimation and planned therapy of 328 consecutive patients. Breast. 2009;18(5):316–321. doi: 10.1016/j.breast.2009.08.004. [DOI] [PubMed] [Google Scholar]
- 64.Klang SH, Hammerman A, Liebermann N, et al. Economic implications of 21-gene breast cancer risk assay from the perspective of an Israeli-managed health-care organization. Value Health. 2010;13(4):381–387. doi: 10.1111/j.1524-4733.2010.00724.x. [DOI] [PubMed] [Google Scholar]
- 65.Ademuyiwa FO, Miller A, O'Connor T, et al. The effects of oncotype DX recurrence scores on chemotherapy utilization in a multi-institutional breast cancer cohort. Breast Cancer Res Treat. 2011;126(3):797–802. doi: 10.1007/s10549-010-1329-6. [DOI] [PubMed] [Google Scholar]
- 66.Oratz R, Kim B, Chao C, et al. Physician survey of the effect of the 21-gene recurrence score assay results on treatment recommendations for patients with lymph node-positive, estrogen receptor-positive breast cancer. J Oncol Pract. 2011;7(2):94–99. doi: 10.1200/JOP.2010.000046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Joh JE, Esposito NN, Kiluk JV, et al. The effect of Oncotype DX recurrence score on treatment recommendations for patients with estrogen receptor-positive early stage breast cancer and correlation with estimation of recurrence risk by breast cancer specialists. Oncologist. 2011;16(11):1520–1526. doi: 10.1634/theoncologist.2011-0045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Malo TL, Lipkus I, Wilson T, et al. Treatment Choices Based on OncotypeDx in the Breast Oncology Care Setting. J Cancer Epidemiol. 2012;2012:941495. doi: 10.1155/2012/941495. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Holt S, Bertelli G, Humphreys I, et al. A decision impact, decision conflict and economic assessment of routine Oncotype DX testing of 146 women with node-negative or pNImi, ER-positive breast cancer in the U.K. Br J Cancer. 2013;108(11):2250–2258. doi: 10.1038/bjc.2013.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Stemmer SM, Klang SH, Ben-Baruch N, et al. The impact of the 21-gene Recurrence Score assay on clinical decision-making in node-positive (up to 3 positive nodes) estrogen receptor-positive breast cancer patients. Breast Cancer Res Treat. 2013;140(1):83–92. doi: 10.1007/s10549-013-2603-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Davidson JA, Cromwell I, Ellard SL, et al. A prospective clinical utility and pharmacoeconomic study of the impact of the 21-gene Recurrence Score® assay in oestrogen receptor positive node negative breast cancer. Eur J Cancer. 2013;49(11):2469–2475. doi: 10.1016/j.ejca.2013.03.009. [DOI] [PubMed] [Google Scholar]
- 72.Yamauchi H, Nakagawa C2, Takei H, et al. Prospective study of the effect of the 21-gene assay on adjuvant clinical decision-making in Japanese women with estrogen receptor-positive, node-negative, and node-positive breast cancer. Clin Breast Cancer. 2014;14(3):191–197. doi: 10.1016/j.clbc.2013.10.017. [DOI] [PubMed] [Google Scholar]
- 73.Fried G, Moskovitz M, et al. Treatment decisions in estrogen receptor-positive early breast cancer patients with intermediate oncotype DX recurrence score results. Springerplus. 2014;3:71. doi: 10.1186/2193-1801-3-71. [DOI] [PMC free article] [PubMed] [Google Scholar]

