Purpose.—

To update key recommendations of the American Society of Clinical Oncology/College of American Pathologists estrogen receptor (ER) and progesterone receptor (PgR) testing in breast cancer guideline.

Methods.—

A multidisciplinary international Expert Panel was convened to update the clinical practice guideline recommendations informed by a systematic review of the medical literature.

Recommendations.—

The Expert Panel continues to recommend ER testing of invasive breast cancers by validated immunohistochemistry as the standard for predicting which patients may benefit from endocrine therapy, and no other assays are recommended for this purpose. Breast cancer samples with 1% to 100% of tumor nuclei positive should be interpreted as ER positive. However, the Expert Panel acknowledges that there are limited data on endocrine therapy benefit for cancers with 1% to 10% of cells staining ER positive. Samples with these results should be reported using a new reporting category, ER Low Positive, with a recommended comment. A sample is considered ER negative if < 1% or 0% of tumor cell nuclei are immunoreactive. Additional strategies recommended to promote optimal performance, interpretation, and reporting of cases with an initial low to no ER staining result include establishing a laboratory-specific standard operating procedure describing additional steps used by the laboratory to confirm/adjudicate results. The status of controls should be reported for cases with 0% to 10% staining. Similar principles apply to PgR testing, which is used primarily for prognostic purposes in the setting of an ER-positive cancer. Testing of ductal carcinoma in situ (DCIS) for ER is recommended to determine potential benefit of endocrine therapies to reduce risk of future breast cancer, while testing DCIS for PgR is considered optional. Additional information can be found at www.asco.org/breast-cancer-guidelines.

First released in 2010, the American Society of Clinical Oncology (ASCO)/College of American Pathologists (CAP) estrogen receptor (ER) and progesterone receptor (PgR) testing guideline is aimed at improving the analytic performance and diagnostic accuracy of ER and PgR testing and their clinical utility as biomarkers for the management of women with primary breast cancer.1,2  The guideline focuses entirely on immunohistochemical testing, as this reflects the near exclusive use of this approach in contemporary practice. The Expert Panel (Appendix) reconvened to consider evidence for changes in laboratory and clinical practice or the emergence of new data that might require an update in this guideline. The importance of the accurate assessment (protocols and readout) and interpretation of ER and PgR expression is emphasized by more than 1,000,000 women per year worldwide diagnosed with primary breast cancer and tested for these receptors. Studies using contemporary populations note increases in the proportion of breast cancers that are ER positive, with overall rates of between 79% and 84% of breast cancers (with higher ER-positive rates occurring in postmenopausal subpopulations).39  While ER-positive rates are influenced by population-dependent variables (eg, age, race, screening, birth rate, and so on), increased analytic sensitivity of assay protocols due to adherence to previously published guidelines, newer detection methods, more sensitive primary antibodies, and protocol design changes after feedback provided by external quality assessment may also have contributed to this increase.

Utility of ER and PgR Testing and Threshold Setting

The Expert Panel acknowledged that hormone receptor testing in breast cancer currently serves other purposes beyond identification of which patients may benefit from endocrine-specific strategies for breast cancer treatment. These include the following: 1) to assist in classification of breast cancer for the most appropriate overall treatment pathway (with most treatment guidelines centered around ER-positive v ER-negative pathways); 2) to assist in prognostication, such as classification of breast cancer, for the most appropriate overall prognostic stage group (eg, American Joint Committee on Cancer [AJCC] eighth edition prognostic stage groupings)10 ; and 3) as a diagnostic aid in metastatic breast cancer. The Expert Panel acknowledged that a well-performed ER assay should be useful in each of these scenarios. It should be noted, however, that the specific thresholds for a positive versus a negative test result in this guideline are based on the data supporting the optimal threshold to use ER status as a predictive marker for endocrine treatment strategies in breast cancer.

There is unequivocal evidence that patients with cancers devoid of ER expression do not benefit from endocrine treatment.11  The challenge has been and remains defining an ER expression cutoff that best segregates patients who may derive meaningful clinical benefit from endocrine therapy strategies from those who will not. The 2010 guideline recommended that invasive breast cancers be considered positive if at least 1% of cancer nuclei stain positive and that patients with such cancers be considered for endocrine therapy, while such therapy should be withheld from patients with cancers with < 1% staining. It was also noted that it is reasonable for oncologists to discuss the pros and cons of endocrine therapy with patients whose cancers contain low levels of ER by immunohistochemistry (IHC; 1%–10% weakly positive cells) and to make a decision based on the totality of information about the individual case. This recommendation is reaffirmed in this 2019 update (Clinical Question 1).

The utility of PgR testing continues to be largely prognostic in the ER-positive population, but testing using principles similar to those used in ER testing is still recommended for invasive breast cancers.

Current Status of ER and PgR Testing and Focus Areas for Improvement

The Expert Panel examined data on the quality of hormone receptor testing in breast cancer in the years since the 2010 guideline was first published to identify areas where additional guidance might be beneficial. There has been an apparent improvement in the overall quality of hormone receptor testing in breast cancer and improved monitoring of performance. While interlaboratory variability for ER and PgR results has decreased, some variability continues to exist, emphasizing the need for continued publication of antibody- and method-specific results, guidance on best practices, and continued monitoring of pathologist scoring (readout) performance to improve reproducibility and reduce interobserver variation.

Handling of Cases With Low ER Expression

Although the recent literature supports reaffirming the current guideline recommendations overall (Clinical Question 1), there has been increased concern over the proper handling of cases with low ER expression. Such cases with low levels of ER expression are included in ER-positive treatment and prognostic algorithms designed for a majority of cases, which have strong ER expression. Although uncommon (accounting for only 2%–3% of ER-positive cancers), cancers with 1% to 10% cells staining for ER present particular clinical challenges. For example, should a high-grade cancer with 1% to 10% ER expression, 0% PgR expression, and human epidermal growth factor receptor 2 (HER2)−negative results be considered for treatments designed for triple-negative cancers? One of the most common questions asked during the open comment period for the ASCO/CAP HER2 guideline update, which included a question on issues related to the 2010 ER/PgR testing guideline, was what to do clinically with ER Low Positive cancers.

Cases with weak or low ER or PgR staining are also noted to be particular challenges for test reproducibility due to a variety of factors (reproducibility of scoring close to a threshold, particular sensitivity to preanalytic [eg, cold ischemic time, fixation type and time, and so on] and analytic factors [eg, antibody used, methodology], heterogeneity of expression, and lack of standardized low ER−expressing controls). Clinical Question 2 of this update focuses on strategies to promote optimal performance, interpretation, and reporting of all cases, and new recommendations for interpreting and reporting cases with weak or ≤ 10% ER expression are provided.

Given recent advances in alternative testing strategies, the guideline update also addresses the question of whether evolving genomic/molecular and image analysis methods are ready to be incorporated into routine testing for ER and PgR (Clinical Question 3).

All the recommendations apply to patients with invasive breast cancer; however, recommendations on hormone receptor testing in ductal carcinoma in situ (DCIS) are also offered (Clinical Question 4).

This update specifically addresses 4 clinical questions raised after the publication of the 2010 guideline:

  1. What are the optimum quality assurance (QA), tissue handling, scoring system, and reporting for determining potential benefit from endocrine therapy?

  2. What additional strategies can promote optimal performance, interpretation, and reporting of IHC assays, particularly in cases with low ER expression?

  3. Are other ER expression assays acceptable for identifying patients likely to benefit from endocrine therapy?

  4. Should DCIS be routinely tested for hormone receptors?

This ASCO and CAP clinical practice guideline update provides revised recommendations with a comprehensive discussion of the relevant literature for these specific recommendations. The full guideline and additional information are available at www.asco.org/breast-cancer-guidelines; this guideline is also available on the Archives Web site at www.archivesofpathology.org. The complete list of recommendations is in Table 1, including the updated recommendations.

Table 1

Summary of All Recommendations

Summary of All Recommendations
Summary of All Recommendations

Guideline Update Process

This systematic review−based guideline product was developed by a multidisciplinary Expert Panel, which included a patient representative and an ASCO guideline staff member with health research methodology expertise. PubMed and the Cochrane Library were searched for randomized controlled trials, systematic reviews, meta-analyses, and clinical practice guidelines for the period from January 1, 2008, of the previous update through April 30, 2019. The disease and intervention search terms were those that were used for the 2010 guideline. The searches identified 4,897 abstracts, and ultimately, 87 papers met the selection criteria.11,1499  Articles were selected for inclusion in the systematic review of the evidence based on the following criteria:

  • The population included adults with a new diagnosis of breast cancer or a recurrence

  • Studies of ER or PgR testing by IHC

  • Primary end points considered positive and negative predictive values of assays used to accurately determine hormone receptor status, including (but not necessarily limited to): specific assay performance, technique, standardization attempted, QA, proficiency testing, individual or institutional training, or improvement in assay results based on interventions

Articles were excluded from the systematic review if they were (1) meeting abstracts not subsequently published in peer-reviewed journals; (2) editorials, commentaries, letters, news articles, case reports, or narrative reviews; or (3) published in a non-English language.

The Expert Panel met in person at ASCO headquarters to update the guideline. The updated ASCO/CAP guideline was circulated in draft form, reviewed, and approved by the Expert Panel. ASCO's Clinical Practice Guidelines Committee (CPGC) reviewed and approved the final document. For CAP, an independent review panel was assembled to review and approve the guideline. The independent review panel was masked to the Expert Panel and was vetted through the conflict-of-interest process. All funding for the administration of the project was provided by ASCO.

Guideline Disclaimer

The clinical practice guidelines and other guidance published herein are provided by the American Society of Clinical Oncology, Inc. (“ASCO”) and the College of American Pathologists (CAP) to assist providers in clinical decision making. The information therein should not be relied upon as being complete or accurate, nor should it be considered as inclusive of all proper treatments or methods of care or as a statement of the standard of care. With the rapid development of scientific knowledge, new evidence may emerge between the time information is developed and when it is published or read. The information is not continually updated and may not reflect the most recent evidence. The information addresses only the topics specifically identified therein and is not applicable to other interventions, diseases, or stages of diseases. This information does not mandate any particular course of medical care. Further, the information is not intended to substitute for the independent professional judgment of the treating provider, as the information does not account for individual variation among patients. Recommendations reflect high, moderate or low confidence that the recommendation reflects the net effect of a given course of action. The use of words like “must,” “must not,” “should,” and “should not” indicate that a course of action is recommended or not recommended for either most or many patients, but there is latitude for the treating physician to select other courses of action in individual cases. In all cases, the selected course of action should be considered by the treating provider in the context of treating the individual patient. Use of the information is voluntary. ASCO and the CAP provide this information on an “as is” basis, and make no warranty, express or implied, regarding the information. ASCO and the CAP specifically disclaim any warranties of merchantability or fitness for a particular use or purpose. ASCO and the CAP assume no responsibility for any injury or damage to persons or property arising out of or related to any use of this information or for any errors or omissions.

Guideline and Conflicts of Interest

The Expert Panel was assembled in accordance with ASCO's Conflict of Interest Policy Implementation for Clinical Practice Guidelines (“Policy,” found at http://www.asco.org/rwc) as agreed upon with CAP. All members of the Expert Panel completed ASCO's disclosure form, which requires disclosure of financial and other interests, including relationships with commercial entities that are reasonably likely to experience direct regulatory or commercial impact as a result of promulgation of the guideline. Categories for disclosure include employment; leadership; stock or other ownership; honoraria, consulting or advisory role; speaker's bureau; research funding; patents, royalties, other intellectual property; expert testimony; travel, accommodations, expenses; and other relationships. In accordance with the “Policy,” the majority of the members of the Expert Panel did not disclose any relationships constituting a conflict under the “Policy.”

Clinical Question 1

What are the optimum QA, testing conditions, tissue handling, scoring system, and reporting for determining potential benefit from endocrine therapy?

Recommendation 1.1. Optimal Algorithm for ER/PgR testing

Samples with 1% to 100% of tumor nuclei positive for ER or PgR are interpreted as positive. For reporting of ER (not PgR), if 1% to 10% of tumor cell nuclei are immunoreactive, the sample should be reported as ER Low Positive with a recommended comment (Table 2; Figure 1). A sample is considered negative for ER or PgR if < 1% or 0% of tumor cell nuclei are immunoreactive. A sample may be deemed uninterpretable for ER or PgR if the sample is inadequate (insufficient cancer or severe artifacts present, as determined at the discretion of the pathologist), if external and internal controls (if present) do not stain appropriately, or if preanalytic variables have interfered with the assay's accuracy (Figures 1 through 4). Clinicians should be aware of and be able to discuss with patients the limited data on ER−low positive cases and issues with test results that are close to a positive threshold (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Table 2

Additional Recommended Reporting Comments for Specific Scenarios

Additional Recommended Reporting Comments for Specific Scenarios
Additional Recommended Reporting Comments for Specific Scenarios
Figure 1

Recommendations for scoring (readout) and interpretation of immunohistochemistry (IHC) test to determine estrogen receptor (ER) status in breast cancers. For progesterone receptor (PgR) testing, the same overall interpretation principles apply, but the reporting elements are only recommended for ER testing. PgR should be interpreted as either positive (if 1%–100% of cells have nuclear staining) or negative (if < 1% or 0% of cells have nuclear staining), with the overall percentage and intensity of staining reported. (*) Hormone receptor testing should only be done with a validated method and with appropriate laboratory procedures, including ongoing assay monitoring and pathologist competency assessment. (†) If no internal controls are present but external controls are positive, include comment: “No internal controls are present, but external controls are appropriately positive. If needed, testing another specimen that contains internal controls may be warranted for confirmation of ER status.” (‡) For ER Low Positive results, include comment: “The cancer in this sample has a low level (1%–10%) of ER expression by IHC. There are limited data on the overall benefit of endocrine therapies for patients with these results, but they currently suggest possible benefit, so patients are considered eligible for endocrine treatment. There are data that suggest invasive cancers with these results are heterogeneous in both behavior and biology and often have gene expression profiles more similar to ER-negative cancers.” Abbreviation: SOP, standard operating procedure.

Figure 1

Recommendations for scoring (readout) and interpretation of immunohistochemistry (IHC) test to determine estrogen receptor (ER) status in breast cancers. For progesterone receptor (PgR) testing, the same overall interpretation principles apply, but the reporting elements are only recommended for ER testing. PgR should be interpreted as either positive (if 1%–100% of cells have nuclear staining) or negative (if < 1% or 0% of cells have nuclear staining), with the overall percentage and intensity of staining reported. (*) Hormone receptor testing should only be done with a validated method and with appropriate laboratory procedures, including ongoing assay monitoring and pathologist competency assessment. (†) If no internal controls are present but external controls are positive, include comment: “No internal controls are present, but external controls are appropriately positive. If needed, testing another specimen that contains internal controls may be warranted for confirmation of ER status.” (‡) For ER Low Positive results, include comment: “The cancer in this sample has a low level (1%–10%) of ER expression by IHC. There are limited data on the overall benefit of endocrine therapies for patients with these results, but they currently suggest possible benefit, so patients are considered eligible for endocrine treatment. There are data that suggest invasive cancers with these results are heterogeneous in both behavior and biology and often have gene expression profiles more similar to ER-negative cancers.” Abbreviation: SOP, standard operating procedure.

Close modal
Figure 2

Case examples to illustrate stain intensity and percentage interpretation. Examples of invasive cancers with various levels of estrogen receptor (ER) expression. Magnification of slides at (A) ×50 and (B) ×200 from a case that was strongly and uniformly positive (90%–100% cells staining, strong intensity), at (C) ×50 and (D) ×200 from a case with weak-moderate intensity but almost uniform positivity (70%–80% cells staining, weak-moderate intensity), and at (E) ×50 and (F) ×200 from a case that had between 1% and 10% of cancer cells staining with weak intensity (ER Low Positive). This low level of expression is not easily seen on (E) low power but is more readily seen on (F) moderate to higher power. Magnification of slides at (G) ×50 and (H) ×200 from an invasive cancer with no ER expression (0% staining). Cases with nuclear staining in < 1% of total cells in the invasive carcinoma sample are also classified as negative.

Figure 2

Case examples to illustrate stain intensity and percentage interpretation. Examples of invasive cancers with various levels of estrogen receptor (ER) expression. Magnification of slides at (A) ×50 and (B) ×200 from a case that was strongly and uniformly positive (90%–100% cells staining, strong intensity), at (C) ×50 and (D) ×200 from a case with weak-moderate intensity but almost uniform positivity (70%–80% cells staining, weak-moderate intensity), and at (E) ×50 and (F) ×200 from a case that had between 1% and 10% of cancer cells staining with weak intensity (ER Low Positive). This low level of expression is not easily seen on (E) low power but is more readily seen on (F) moderate to higher power. Magnification of slides at (G) ×50 and (H) ×200 from an invasive cancer with no ER expression (0% staining). Cases with nuclear staining in < 1% of total cells in the invasive carcinoma sample are also classified as negative.

Close modal
Figure 3

Internal controls. Examples of internal control analysis in estrogen receptor (ER) stains. (A) ER-negative invasive cancer (×200) with an adjacent internal normal duct control (asterisks) with optimal staining (nuclear staining varying from weak to very strong with alternating clusters of negative cells). (B) Unexpectedly weak stain result (×200) in both the grade 1 invasive cancer and normal duct internal controls (asterisks). This result, although positive, raises concern about possible preanalytic variables affecting the assay or, if these are deemed appropriate, that the level of analytic sensitivity of the assay may be too low (especially to detect ER–low positive results). (C) Case negative for ER expression (×400) both in the invasive cancer and the internal control duct (asterisks). The assay should be repeated and an investigation for potential causes of negative internal controls performed (including both preanalytic and analytic factors). If internal controls remain negative and this issue appears isolated to this sample, the test should be reported as “cannot be determined” (indeterminate; uninterpretable because of negative internal controls, possible preanalytic tissue preservation issues). (D) Invasive cancer with no ER staining (×200) and internal control columnar cell epithelium (asterisks) with uniformly strong nuclear staining (as expected). These results help support that there were no serious issues with the preanalytic or analytic phases. However, such strong positive internal controls are less optimal to evaluate how well the assay is able to detect low levels of ER expression (evaluation of noncolumnar cell, normal duct controls is more optimal, as in panel A).

Figure 3

Internal controls. Examples of internal control analysis in estrogen receptor (ER) stains. (A) ER-negative invasive cancer (×200) with an adjacent internal normal duct control (asterisks) with optimal staining (nuclear staining varying from weak to very strong with alternating clusters of negative cells). (B) Unexpectedly weak stain result (×200) in both the grade 1 invasive cancer and normal duct internal controls (asterisks). This result, although positive, raises concern about possible preanalytic variables affecting the assay or, if these are deemed appropriate, that the level of analytic sensitivity of the assay may be too low (especially to detect ER–low positive results). (C) Case negative for ER expression (×400) both in the invasive cancer and the internal control duct (asterisks). The assay should be repeated and an investigation for potential causes of negative internal controls performed (including both preanalytic and analytic factors). If internal controls remain negative and this issue appears isolated to this sample, the test should be reported as “cannot be determined” (indeterminate; uninterpretable because of negative internal controls, possible preanalytic tissue preservation issues). (D) Invasive cancer with no ER staining (×200) and internal control columnar cell epithelium (asterisks) with uniformly strong nuclear staining (as expected). These results help support that there were no serious issues with the preanalytic or analytic phases. However, such strong positive internal controls are less optimal to evaluate how well the assay is able to detect low levels of ER expression (evaluation of noncolumnar cell, normal duct controls is more optimal, as in panel A).

Close modal
Figure 4

External controls. Optimal external (ideally on-slide) controls for both estrogen receptor (ER) and progesterone receptor (PgR) should include multiple tissues, including ones with expected strong staining, lower limit of detection levels, and negative controls; tonsil and cervix have been used to meet these criteria. (A) Tonsil tissue (×200) with optimal staining for ER to ensure an appropriate low limit of detection; dispersed germinal center cells and the squamous epithelium should be ER positive, but the B cells in the mantle zones should be ER negative. In contrast to ER, no nuclear PgR staining should be seen in tonsillar tissue. Weak-positive PgR staining in tonsil should result in workup to determine if assay drift has occurred. (B) PgR variably staining the basal layer of the squamous mucosa (×200) as expected (this staining should ensure an appropriate low limit of detection for PgR). Stromal cells stain strongly for both ER and PgR. ER should stain the squamous mucosa more uniformly (not just the basal layer), with at least moderate to strong stain intensity. (C) PgR staining (×200) should also be positive in the endocervical columnar epithelial cells (with some variability expected). ER should stain almost all endocervical columnar epithelial cells. Of note, it should be taken into consideration that hormone receptor staining of cervical tissue may be reduced in tissue from postmenopausal women.

Figure 4

External controls. Optimal external (ideally on-slide) controls for both estrogen receptor (ER) and progesterone receptor (PgR) should include multiple tissues, including ones with expected strong staining, lower limit of detection levels, and negative controls; tonsil and cervix have been used to meet these criteria. (A) Tonsil tissue (×200) with optimal staining for ER to ensure an appropriate low limit of detection; dispersed germinal center cells and the squamous epithelium should be ER positive, but the B cells in the mantle zones should be ER negative. In contrast to ER, no nuclear PgR staining should be seen in tonsillar tissue. Weak-positive PgR staining in tonsil should result in workup to determine if assay drift has occurred. (B) PgR variably staining the basal layer of the squamous mucosa (×200) as expected (this staining should ensure an appropriate low limit of detection for PgR). Stromal cells stain strongly for both ER and PgR. ER should stain the squamous mucosa more uniformly (not just the basal layer), with at least moderate to strong stain intensity. (C) PgR staining (×200) should also be positive in the endocervical columnar epithelial cells (with some variability expected). ER should stain almost all endocervical columnar epithelial cells. Of note, it should be taken into consideration that hormone receptor staining of cervical tissue may be reduced in tissue from postmenopausal women.

Close modal

Recommendation 1.2. Optimal Testing Conditions (no change)

Large (preferably multiple) core biopsies of tumor are preferred for testing if they are representative of the tumor (grade and type) at resection. Accession slip and report must include guideline-detailed elements (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 1.3. Optimal Tissue Handling Requirements (no change)

Time from tissue acquisition to fixation should be as short as possible. Samples for ER and PgR testing are fixed in 10% neutral buffered formalin (NBF) for 6 to 72 hours. Samples should be sliced at 5-mm intervals after appropriate gross inspection and margin designation and placed in sufficient volume of NBF to allow adequate tissue penetration. If tumor comes from remote location, it should be bisected through the tumor on removal and sent to the laboratory immersed in a sufficient volume of NBF. Cold ischemia time, fixative type, and time the sample was placed in NBF must be recorded. As in the ASCO/CAP HER2 guideline, use of unstained slides cut more than 6 weeks before analysis is not recommended. Time tissue is removed from patient, time tissue is placed in fixative, duration of fixation, and fixative type must be recorded and noted on accession slip or in report (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 1.4. Optimal Internal Validation Procedures (change anticipated)

This topic is deferred to the forthcoming CAP guideline update of the principles of analytic validation of IHC assays, once available. There should be initial test validation/verification prior to reporting any clinical samples. Prior to that, previously recommended principles apply, as described by Fitzgibbons et al12  and more recently Torlakovic et al13  (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 1.5. Optimal Internal QA Procedures

Ongoing quality control and equipment maintenance are required. Initial and ongoing laboratory personnel training and competency assessment should be performed. Standard operating procedures (SOPs) should be used that include routine use of external control materials with each batch of testing and routine evaluation of internal normal epithelial elements or the inclusion of normal breast sections (or other appropriate control) on each tested slide, wherever possible. External controls should include negative and positive samples as well as samples with lower percentages of ER expression (such as tonsil). On-slide controls are recommended.

Regular, ongoing assay reassessment should be done at least semiannually (as described by Fitzgibbons et al12 ). Revalidation is needed whenever there is a significant change to the test system.13  Ongoing competency assessment and education of pathologists are required (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 1.6. Optimal External Proficiency Assessment

The laboratory performing ER and PgR testing must participate in external proficiency testing or alternative performance assessment as required by its accrediting organization (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 1.7. Optimal Laboratory Accreditation

On-site inspection every other year should be undertaken, with annual requirement for self-inspection (Type: Informal consensus; Evidence quality: Intermediate; Strength of recommendation: Moderate).

Literature Review and Analysis

Data from proficiency testing and quality control programs indicate that an overall improvement in the quality and reproducibility of ER and PgR testing in breast cancer has occurred over time. This is likely the result of improvements in standardization of preanalytic, analytic, and postanalytic factors, as well as increases in antibody sensitivity, allowing the Expert Panel to reaffirm the original 2010 recommendations on specimen handling, optimal testing conditions, and QA.100,101 

Much of the focus of the update review involved reexamining the data on the optimal ER positive threshold and scoring systems to determine potential benefit from endocrine therapy. There are limited new data on this threshold, as most randomized clinical trials addressing the topic took place in the 1990s. There is little argument about the potential benefit of endocrine therapy in patients with cancers with > 10% ER expression or the lack of potential benefit for cancers with < 1% ER expression. However, there are much more limited data on the 2% to 3% of cancers that are low ER expressers (most often defined as 1%–10% ER-positive cells by IHC) and their potential benefit from endocrine therapy. In 2011, a large meta-analysis was published of 20 prior clinical trials with more than 200,000 women-years of follow-up, reporting on the benefit of 5 years of tamoxifen according to ER and PgR levels as measured by ligand-binding assay (LBA).11  More than 50,000 women-years of follow-up were available for women with tumors having <10 fmol ER/mg protein, and no evidence of benefit was apparent. However, patients with cancers with low levels of ER (10–20 fmol ER/mg protein) had their likelihood of recurrence reduced by one third with the addition of approximately 5 years of tamoxifen (risk ratio [RR], 0.67; SE, 0.08). Of note, while benefit increased somewhat with higher ER levels, the proportional effect at the highest ER levels (> 200 fmol/mg) was only slightly better than that at weak ER levels (RR, 0.52; SE, 0.07). Although there are limited data from prospective randomized trials comparing the predictive power of LBAs with the standard IHC methods of ER assessment in routine use, multiple studies support high rates of agreement between these assays.98,99,102104  In most studies, a ≥ 10 fmol/mg threshold corresponded best (albeit imperfectly) with ≥ 1% of cells with nuclear ER staining by IHC, the current recommended IHC cutoff for ER positivity.11,97,105 

Complicating the understanding of low ER-expressing cancers are data indicating that these cancers are a heterogeneous group but often have clinical outcomes and biologic/molecular profiles that are often more similar to those of ER-negative cancers.9197,106  However, none of these retrospective and nonrandomized studies can address the potential benefit of endocrine therapy for at least some patients in the 1% to 10% ER-positive group. The Expert Panel acknowledges the data from these studies provide support that cancers with low ER expression may be biologically distinct from high ER expressers and that the 1% threshold for ER positivity may not uniformly predict differences in prognosis, chemotherapy benefit, or regimen, or define a specific molecular subtype. Most important, low ER expression status has not been validated for these purposes.

Given the relatively low toxicity of endocrine therapy, the desire to minimize false-negative results, and the available (although limited) data supporting potential benefit even in cases with as low as 1% to 10% positivity, the Expert Panel continues to recommend ≥ 1% nuclear ER staining by IHC as the threshold for reporting a positive ER result to predict potential clinical benefit from endocrine therapy treatments. However, cases with 1% to 10% staining should be reported as ER Low Positive, with a recommended comment explaining the more limited clinical data, heterogeneous behavior, and biology of this subgroup of ER-positive cancers. As in 2010, the Expert Panel recommends that oncologists discuss the pros and cons of endocrine therapy with patients whose cancers contain low levels of ER by IHC and base decisions on the totality of information available about an individual case. Laboratories should continue to report both the percentage and intensity of hormone receptor staining in addition to the test interpretation as positive, low positive, or negative. Clinical Question 2 addresses additional steps that should be taken to promote optimal performance and interpretation, especially in the weak or low ER−expressing cases.

Controls

Control tissues are essential for evaluating assay performance. External controls should include negative and positive samples as well as samples with lower percentages of ER expression (such as tonsil). On-slide controls are ideal because each slide is evaluated, and the control stays with the slide. Regardless of the control type, the controls must include samples fixed under conditions similar to those of the test samples and incorporate tissues or cell lines with no ER, low ER, and high ER and be as well standardized as possible.107110  While newer forms of standardized controls are becoming more available, some of which are engineered tissue-like materials with defined quantities of ER or PgR included, long-standing experience has also been good for cervix (as a strong ER-positive/PgR-positive control) and tonsil (for PgR see: https://www.nordiqc.org/epitope.php?id=67 and for ER: https://www.nordiqc.org/downloads/assessments/118_2.pdf).111115  Tonsil has been suggested as an ideal tissue type to include in external controls to monitor the analytic sensitivity for ER and PgR (Figure 4). Dispersed germinal center cells and the squamous epithelium should be ER positive, but the B cells in the mantle zones should be ER negative. In contrast to ER, no nuclear PgR staining should be seen in tonsillar tissue. Weak positive PgR staining in tonsil should result in workup to determine if assay drift has occurred. Tumor tissues with variable levels of expression can be useful as a supplement to tonsil and control tissue with uniform ER/PgR expression (such as cervix); however, it should be noted that tumor tissue can be heterogeneous (creating different staining patterns on a given level), and expression levels may not be as well characterized. If changes in staining results over time or between runs (drift) are noted (especially the staining with low levels of ER expression), the laboratory should undertake a careful analysis of its procedures and any recent changes in test methods (eg, new lot of antibody, change in clone, or modified reagents) prior to issuing results to assess whether revalidation is required.

A guideline update dealing with IHC assay validation is under development by CAP at the time of this publication and should be deferred to once published. Prior to its publication, the Expert Panel recommends applying previously recommended validation principles (as described by Fitzgibbons et al12  and more recently Torlakovic et al13 ).

Do the Same Principles Apply to PgR Testing?

There is substantial evidence for higher rates of clinical response to endocrine therapy in PgR-positive tumors treated neoadjuvantly or in metastatic disease, but randomized trials in the adjuvant setting have revealed no difference in the degree of benefit from adjuvant endocrine treatment according to PgR status.11  The Expert Panel therefore acknowledges that only ER should be used as a predictor of benefit from adjuvant endocrine therapy. Because of this, the Expert Panel discussed whether to continue to recommend routine PgR testing in invasive breast cancers. PgR levels can add prognostic information by helping to stratify outcomes in the ER-positive population, with data supporting that cases with lower or negative PgR expression may have a worse prognosis.8890,116  Used in combination with ER (and other markers), PgR levels by IHC have been used by various tools that estimate prognosis, such as the IHC4 score, Magee equations, nomograms that predict the 21-gene recurrence score results, AJCC eighth edition prognostic stage groupings, and various predictors of response to neoadjuvant therapy.10,73,75,7787 

In addition, PgR may serve as an informal control for samples that test ER negative but PgR positive (especially in the absence of normal internal controls), since there are data suggesting that this phenotype is frequently the result of technical artifact.74  In the 2010 guideline, repeat testing in cases with initial ER-negative/PgR-positive results was suggested (not required), and the Expert Panel continues to support this recommendation (Table 1). Lastly, although controversial as a result category, confirmed ER-negative/PgR-positive samples may represent a rare biologic phenotype that may be offered endocrine therapies, although due to the rarity of this result group, there are limited data to support this.70,71,76 

Given the utility of PgR testing for the above reasons, we continue to recommend routine PgR testing of invasive breast cancers. Many thresholds have been used to differentiate cancers on the basis of PgR expression for prognostic purposes. This may reflect that PgR acts as a more continuous variable for prognosis,72  and in the absence of data consistently supporting alternative thresholds or standards for PgR testing, we recommend using 1% as a positivity threshold for PgR in invasive breast cancers but also continue to recommend reporting the percentage and intensity of cells staining. However, the low positive reporting category and comment recommendation for samples with 1% to 10% ER expression does not apply to PgR. Otherwise, the same general recommendations that apply to ER testing should also apply to PgR testing, including participation in external proficiency testing by the laboratory's accrediting organization. However, laboratory accreditation should primarily be dependent on a passing grade for ER proficiency testing.

Clinical Question 2

What additional strategies can promote optimal performance, interpretation, and reporting of IHC assays, particularly in cases with low to negative ER expression?

Recommendation 2.1

Laboratories should include ongoing quality control using SOPs for test evaluation prior to scoring (readout) and interpretation of any case as defined in the checklist in Figure 1 (Type: Informal consensus; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 2.2

Interpretation of any ER result should include evaluation of the concordance with the histologic findings of each case. Clinicians should also be aware of when results are highly unusual/discordant and work with pathologists to attempt to resolve or explain atypical reported findings; Table 3 is an aid in this process (Type: Informal consensus; Evidence quality: High; Strength of recommendation: Strong).

Table 3

Invasive Breast Cancer Histopathologic Concordance With ER Staining

Invasive Breast Cancer Histopathologic Concordance With ER Staining
Invasive Breast Cancer Histopathologic Concordance With ER Staining

Recommendation 2.3

Laboratories should establish and follow an SOP stating the steps the laboratory takes to confirm or adjudicate ER results for cases with weak stain intensity or ≤ 10% of cells staining; see Supplemental Digital Content Data Supplement 2, Figure 1 (at www.archivesofpathology.org in the May 2020 table of contents) provides an example SOP (Type: Informal consensus; Evidence quality: High; Strength of recommendation: Strong).

Recommendation 2.4

The status of internal controls should be reported for cases with 0% to 10% staining. For cases with these results without internal controls present and with positive external controls, an additional report comment is recommended (Table 2) (Type: Informal consensus; Evidence quality: High; Strength of recommendation: Strong).

Literature Review and Analysis

Recommendations 2.1 and 2.2 re-emphasize elements of the original 2010 guideline. Recommendation 2.3 is a new focus of this update because of concerns about test validity and reproducibility for cases with weak-intensity, low-level, or negative ER staining. The updated recommendations focus on increased standardization in the workup of these weak, low, or negative ER cases with the development of a specific SOP to confirm or adjudicate the result (Figure 1). This is noted to already be standard best practice in many laboratories.

Figure 1 reviews initial steps in the evaluation of any ER IHC and includes the quality control checklist. Cases with moderate-strong stain intensity and > 10% of cells staining are considered to have robust, reportable results as long as they are considered concordant with histology (Table 3) and no checklist issues are identified. For cases with weak stain intensity or ≤ 10% of nuclei staining, additional steps should be taken to confirm or adjudicate the validity of the results, and correlation with histology should be performed. Steps to consider including in an SOP are shown in Figure 1, and an example of a more detailed SOP for these purposes is available as Supplemental Digital Content Data Supplement 2, Figure 1. Because of previously identified factors involved in false-negative results such as negative or absent internal controls, evaluation of controls is considered an essential part of this process.118  If internal controls are negative, or there are no internal controls and the external positive controls do not have appropriate staining, the assay has failed and needs to be troubleshot. In addition, correlation with any prior patient-specific ER results on a breast cancer would be considered relevant. There are data to support that second reviews and digital quantitative image analysis reads can be used to improve reproducibility and accuracy in a pathologist's scoring (readout) and interpretation, so these can be useful components of an SOP for these cases; however, the Expert Panel acknowledges that current data on these topics are not specific enough to distinguish ER–low positive from ER-negative cases.6169,119121  Additional comments to include in reports for samples of invasive carcinoma that are ER−low positive (1%–10%) or cancers (either invasive or DCIS) with ≤ 10% staining without internal controls present (but positive external controls) are listed in Table 2.

Clinical Question 3

Are other ER expression assays acceptable for identifying patients likely to benefit from endocrine therapy?

Recommendation 3

Validated IHC is the recommended standard test for predicting benefit from endocrine therapy. No other assay types are recommended as the primary screening test for this purpose (Type: Evidence based; Evidence quality: High; Strength of recommendation: Strong).

Literature Review and Analysis

The Expert Panel reviewed the existing evidence and concluded that data are insufficient at this time to recommend newer methods of ER testing as alternatives to IHC for the purposes of determining ER status or selecting which patients are likely to benefit from endocrine therapy. One issue that was apparent was the lack of data from randomized clinical trials using these assays and platforms to select patients for treatment with endocrine therapy versus observation.

However, the Expert Panel recognizes that there are limited avenues for validation of new assays and platforms, as these types of prospective clinical trials are not likely to be conducted. While there are multiple studies that compare messenger RNA (mRNA) with IHC with relatively good agreement, the Expert Panel agreed that this was insufficient to recommend the assays.

Some panel-based gene-expression assays, like Oncotype DX (Genomic Health, Redwood City, CA), have already been incorporated into standard treatment algorithms for IHC ER-positive cancers as a tool to assess the likelihood of clinical benefit offered by chemotherapy when added to endocrine therapy.122125  Assays like Oncotype DX, Mammaprint (Agendia, Irvine, CA), the Prosignia Breast Cancer Prognostic Gene Signature Assay (PAM-50; Prosignia NanoString Technologies, Seattle, WA), EndoPredict (Myriad Genetics, Salt Lake City, UT), and the Breast Cancer Index (Biotheranostics, San Diego, CA) also offer prognostic information regarding the risk of recurrence in patients treated with endocrine therapy, and they have improved the ability to understand the biologic behavior of ER-positive breast cancer as defined by standard IHC measures. In most cases, studies of these new assays established their clinical validity regarding their prognostic utility (outcome after a therapy) but were limited in their predictive utility in identifying patients expected to benefit specifically from endocrine treatment.126,127 

Some of the limited data on benefit from endocrine treatment in relation to ER mRNA expression come from a retrospective study of the NSABP B-14 trial of tamoxifen versus no endocrine therapy. This study showed that in a selected population of patients who were ER positive by LBA, higher ESR1 expression by the Oncotype DX assay was the strongest linear predictor of tamoxifen benefit, with a significant interaction with treatment.60  However, specific ER mRNA expression thresholds to predict potential benefit from endocrine therapy as a screening test for all breast cancers were not developed in this study, since ER−negative cases by LBA were not enrolled in NSABP B-14.

Therefore, while new methods of ER testing may offer some advantages over IHC methods (such as producing a quantitative, highly reproducible result), data on their ability to predict endocrine therapy benefit for all cancers as an initial screening test are limited. In addition, a disadvantage of most current mRNA-based methods is that intermixed noncancer tissues can contribute to test results, which may particularly affect cases close to the positive threshold (eg, an IHC ER–low positive result testing ER negative by quantitative mRNA due to dilution by intermixed noncancer ER-negative tissue). There are very limited data on patients with tumors that are deemed ER−low positive by IHC and tested with newer alternative assays. Test comparison studies suggest that cases that have low levels of ER expression by IHC (but ≥ 1%) are more frequently deemed ER negative by mRNA assays and have variable ESR1 expression across IHC categories (in one study, 24% of cases were considered ER positive by mRNA expression if IHC 1%–9% and 92% if IHC > 10%).59,106  Therefore, ER expression of ≥ 1% by IHC remains the current standard to identify patients who could benefit from endocrine therapy in breast cancer, and a negative quantitative mRNA ER result (eg, on Oncotype DX testing) should not negate an IHC-positive result.

Clinical Question 4

Should DCIS be routinely tested for hormone receptors?

Recommendation 4

ER testing in cases of newly diagnosed DCIS (without associated invasion) is recommended to determine potential benefit of endocrine therapies to reduce risk of future breast cancer. PgR testing is considered optional (Type: Evidence based; Evidence quality: Intermediate; Strength of recommendation: Moderate).

Literature Review and Analysis

In 2010, the Expert Panel acknowledged that newly diagnosed DCIS cases (in the absence of invasion) were commonly being tested for ER and PgR based largely on early but unpublished results from the NSABP B-24 trial.128  Because of the limited data available at the time, the Expert Panel did not make a formal recommendation, leaving it up to patients and their physicians to decide on testing. Subsequently, in 2012, the subset analysis of the NSABP B-24 clinical trial was published comparing tamoxifen versus placebo after lumpectomy and radiation therapy; these trial data showed a significant reduction in relative risk of subsequent breast cancer restricted to patients with ER-positive DCIS at 10 years of follow-up (hazard ratio [HR], 0.49; P = .001).58  In the UK/ANZ randomized clinical trial examining endocrine therapy in DCIS (v no endocrine therapy), long-term follow-up data showed that tamoxifen reduced the incidence of all new breast events in excised DCIS treated with radiation therapy. However, these cases were untested for ER.57,129  In another phase III clinical trial, women with intraepithelial neoplasia, including ER-positive DCIS, were randomly assigned to receive low-dose tamoxifen or placebo. After a median follow-up of 5.1 years, tamoxifen reduced the incidence of new DCIS or invasive breast cancer (HR, 0.48; 95% CI, 0.26 to 0.92).130  Retrospective data from single-institution studies also appear to support a higher risk of recurrence in patients with ER-positive DCIS who were not treated with endocrine therapy.55,56  However, it should be acknowledged that there are currently no data indicating that endocrine-based therapy in the setting of newly diagnosed DCIS has a significant impact on overall survival. Therefore, the decision to use endocrine therapy will depend on individual patient goals and discussion with their clinical care team, but patients should be aware of primary risk reduction options based on the ER status of their DCIS.

Based on the current evidence, the Expert Panel now recommends ER testing in DCIS to guide discussions about adjuvant endocrine therapy. The ER status of newly diagnosed DCIS should be reported when no invasive cancer is present.

Data on whether PgR testing in DCIS adds predictive or prognostic value beyond that of ER alone are currently lacking. In the NSAPB B-24 trial, ER alone was more predictive than combined ER and PgR statuses or PgR status of DCIS for tamoxifen benefit, as patients with ER-positive/PgR-negative DCIS still received benefit, although subsets were small. However, contrary to the prognostic value seen for PgR testing in invasive cancers, studies have not shown significant differences in outcome between ER-positive/PgR-positive and ER-positive/PgR-negative statuses in patients with DCIS.55,56  Given that there are no data currently supporting the prognostic or predictive value of PgR testing in DCIS independent of ER, the Expert Panel considers PgR testing of DCIS to be optional.

The draft recommendations were released to the public for open comment from April 15 through April 29, 2019. Response categories of “Agree” and “Disagree” were captured for every proposed recommendation, with 163 written comments received.

More than 80% of the 163 respondents agreed to 8 of the 10 questions pertaining to the recommendations and 2 questions fell below an 80% agreement rate. Expert Panel members reviewed comments from all sources and determined to revise the recommendations that did not receive at least 80% agreement for clarity. All changes were incorporated prior to ASCO CPGC and CAP review and approval.

More information, including a supplement with evidence tables, slide sets, and clinical tools and resources, is available at www.asco.org/breast-cancer-guidelines. The Methodology Manual (available at www.asco.org/guideline-methodology) provides additional information about the methods used to develop this guideline. Patient information is available at www.cancer.net.

American Society of Clinical Oncology (ASCO) clinical practice guidelines provide recommendations, with comprehensive review and analyses of the relevant literature for each recommendation. Additional information, including a supplement with additional evidence tables, slide sets, clinical tools and resources, and links to patient information at www.cancer.net, is available at www.asco.org/breast-cancer-guidelines.

Disclosures provided by the authors and data availability statement (if applicable) are available with this article at DOI https://doi.org/10.1200/JCO.19.02309. Also see page 17.

Conception and design: Kimberly H. Allison, M. Elizabeth H. Hammond, Mitchell Dowsett, Lisa A. Carey, Daniel F. Hayes, David L. Rimm, W. Fraser Symmans, Leticia Varella, Giuseppe Viale, Tracey F. Weisberg, Lisa M. McShane, Antonio C. Wolff

Administrative support: Shannon E. McKernin, Antonio C. Wolff

Collection and assembly of data: Kimberly H. Allison, M. Elizabeth H. Hammond, Shannon E. McKernin, Sunil R. Lakhani, Charles M. Perou, Meredith M. Regan, David L. Rimm, Leticia Varella, Tracey F. Weisberg, Lisa M. McShane, Antonio C. Wolff

Data analysis and interpretation: Kimberly H. Allison, M. Elizabeth H. Hammond, Mitchell Dowsett, Shannon E. McKernin, Lisa A. Carey, Patrick L. Fitzgibbons, Sunil R. Lakhani, Mariana Chavez-MacGregor, Jane Perlmutter, Charles M. Perou, Meredith M. Regan, David L. Rimm, Emina E. Torlakovic, Leticia Varella, Lisa M. McShane, Antonio C. Wolff

Manuscript writing: All authors

Final approval of manuscript: All authors

Accountable for all aspects of the work: All authors

THE BOTTOM LINE—Estrogen and Progesterone Receptor Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Guideline Update

THE BOTTOM LINE—Estrogen and Progesterone Receptor Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Guideline Update
THE BOTTOM LINE—Estrogen and Progesterone Receptor Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Guideline Update

The Expert Panel wishes to thank Carey Anders, MD, Zeina Nahleh, MD, the CAP Independent Review Panel, and the ASCO Clinical Practice Guidelines Committee for their thoughtful reviews and insightful comments on this guideline.

1
Hammond
ME
,
Hayes
DF
,
Dowsett
M
, et al:
American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer (unabridged version)
.
Arch Pathol Lab Med
134
:
e48
e72
,
2010
2
Hammond
ME
,
Hayes
DF
,
Dowsett
M
, et al:
American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer
.
J Clin Oncol
28
:
2784
2795
,
2010
3
Hwang
KT
,
Kim
J
,
Jung
J
, et al:
Impact of breast cancer subtypes on prognosis of women with operable invasive breast cancer: A population-based study using SEER database
.
Clin Cancer Res
25
:
1970
1979
,
2019
4
Dodson
A
,
Parry
S
,
Ibrahim
M
, et al:
Breast cancer biomarkers in clinical testing: analysis of a UK national external quality assessment scheme for immunocytochemistry and in situ hybridisation database containing results from 199 300 patients
.
J Pathol Clin Res
4
:
262
273
,
2018
5
Anderson
WF
,
Katki
HA
,
Rosenberg
PS
:
Incidence of breast cancer in the United States: Current and future trends
.
J Natl Cancer Inst
103
:
1397
1402
,
2011
6
Sharpe
KH
,
McClements
P
,
Clark
DI
, et al:
Reduced risk of oestrogen receptor positive breast cancer among peri- and post-menopausal women in Scotland following a striking decrease in use of hormone replacement therapy
.
Eur J Cancer
46
:
937
943
,
2010
7
Anderson
WF
,
Rosenberg
PS
,
Petito
L
, et al:
Divergent estrogen receptor-positive and -negative breast cancer trends and etiologic heterogeneity in Denmark
.
Int J Cancer
133
:
2201
2206
,
2013
8
Mullooly
M
,
Murphy
J
,
Gierach
GL
, et al:
Divergent oestrogen receptor-specific breast cancer trends in Ireland (2004–2013): Amassing data from independent Western populations provide etiologic clues
.
Eur J Cancer
86
:
326
333
,
2017
9
Rosenberg
PS
,
Barker
KA
,
Anderson
WF
:
Estrogen receptor status and the future burden of invasive and in situ breast cancers in the United States
.
J Natl Cancer Inst
107
:
djv159
,
2015
10
Hortobagyi
GN
,
Connolly
JL
,
D'Orsi
CJ
, et al:
Breast
,
in
Amin
MB
,
Edge
S
,
Greene
F
, et al
(
eds
):
AJCC Cancer Staging Manual, 8th ed
.
Chicago, IL
,
American College of Surgeons
,
2018
,
pp
589
637
11
Davies
C
,
Godwin
J
,
Gray
R
, et al:
Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: Patient-level meta-analysis of randomised trials
.
Lancet
378
:
771
784
,
2011
12
Fitzgibbons
PL
,
Murphy
DA
,
Hammond
ME
, et al:
Recommendations for validating estrogen and progesterone receptor immunohistochemistry assays
.
Arch Pathol Lab Med
134
:
930
935
,
2010
13
Torlakovic
EE
,
Cheung
CC
,
D'Arrigo
C
, et al:
Evolution of quality assurance for clinical immunohistochemistry in the era of precision medicine. Part 3: Technical validation of immunohistochemistry (IHC) assays in clinical IHC laboratories
.
Appl Immunohistochem Mol Morphol
25
:
151
159
,
2017
14
Ravaioli
S
,
Tumedei
MM
,
Foca
F
, et al:
Androgen and oestrogen receptors as potential prognostic markers for patients with ductal carcinoma in situ treated with surgery and radiotherapy
.
Int J Exp Pathol
98
:
289
295
,
2017
15
Lin
Y
,
Hatem
J
,
Wang
J
, et al:
Tissue microarray-based immunohistochemical study can significantly underestimate the expression of HER2 and progesterone receptor in ductal carcinoma in situ of the breast
.
Biotech Histochem
86
:
345
350
,
2011
16
Aitken
SJ
,
Thomas
JS
,
Langdon
SP
, et al:
Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nodal metastases
.
Ann Oncol
21
:
1254
1261
,
2010
17
Tuominen
VJ
,
Ruotoistenmäki
S
,
Viitanen
A
, et al:
ImmunoRatio: A publicly available Web application for quantitative image analysis of estrogen receptor (ER), progesterone receptor (PR), and Ki-67
.
Breast Cancer Res
12
:
R56
,
2010
18
Stodkowska
J
,
Filas
V
,
Buszkiewicz
E
, et al:
Study on breast carcinoma Her2/neu and hormonal receptors status assessed by automated images analysis systems: ACIS III (Dako) and ScanScope (Aperio)
.
Folia Histochem Cytobiol
48
:
19
25
,
2010
19
Nassar
A
,
Cohen
C
,
Agersborg
SS
, et al:
A multisite performance study comparing the reading of immunohistochemical slides on a computer monitor with conventional manual microscopy for estrogen and progesterone receptor analysis
.
Am J Clin Pathol
135
:
461
467
,
2011
20
Ali
HR
,
Irwin
M
,
Morris
L
, et al:
Astronomical algorithms for automated analysis of tissue protein expression in breast cancer
.
Br J Cancer
108
:
602
612
,
2013
21
Stålhammar
G
,
Fuentes Martinez
N
,
Lippert
M
, et al:
Digital image analysis outperforms manual biomarker assessment in breast cancer
.
Mod Pathol
29
:
318
329
,
2016
22
Liu
WL
,
Wang
LW
,
Chen
JM
, et al:
Application of multispectral imaging in quantitative immunohistochemistry study of breast cancer: A comparative study
.
Tumour Biol
37
:
5013
5024
,
2016
23
Gertych
A
,
Mohan
S
,
Maclary
S
, et al:
Effects of tissue decalcification on the quantification of breast cancer biomarkers by digital image analysis
.
Diagn Pathol
9
:
213
,
2014
24
Nielsen
TO
,
Parker
JS
,
Leung
S
, et al:
A comparison of PAM50 intrinsic subtyping with immunohistochemistry and clinical prognostic factors in tamoxifen-treated estrogen receptor-positive breast cancer
.
Clin Cancer Res
16
:
5222
5232
,
2010
25
Müller
BM
,
Kronenwett
R
,
Hennig
G
, et al:
Quantitative determination of estrogen receptor, progesterone receptor, and HER2 mRNA in formalin-fixed paraffin-embedded tissue: A new option for predictive biomarker assessment in breast cancer
.
Diagn Mol Pathol
20
:
1
10
,
2011
26
Maeda
I
,
Kubota
M
,
Ohta
J
, et al:
Effectiveness of computer-aided diagnosis (CADx) of breast pathology using immunohistochemistry results of core needle biopsy samples for synaptophysin, oestrogen receptor and CK14/p63 for classification of epithelial proliferative lesions of the breast
.
J Clin Pathol
70
:
1057
1062
,
2017
27
Tramm
T
,
Hennig
G
,
Kyndi
M
, et al:
Reliable PCR quantitation of estrogen, progesterone and ERBB2 receptor mRNA from formalin-fixed, paraffin-embedded tissue is independent of prior macro-dissection
.
Virchows Arch
463
:
775
786
,
2013
28
Cheang
MC
,
Martin
M
,
Nielsen
TO
, et al:
Defining breast cancer intrinsic subtypes by quantitative receptor expression
.
Oncologist
20
:
474
482
,
2015
29
Bastien
RR
,
Rodríguez-Lescure
Á
,
Ebbert
MT
, et al:
PAM50 breast cancer subtyping by RT-qPCR and concordance with standard clinical molecular markers
.
BMC Med Genomics
5
:
44
,
2012
30
Wirtz
RM
,
Sihto
H
,
Isola
J
, et al:
Biological subtyping of early breast cancer: A study comparing RT-qPCR with immunohistochemistry
.
Breast Cancer Res Treat
157
:
437
446
,
2016
31
Sheffield
BS
,
Kos
Z
,
Asleh-Aburaya
K
, et al:
Molecular subtype profiling of invasive breast cancers weakly positive for estrogen receptor
.
Breast Cancer Res Treat
155
:
483
490
,
2016
32
Laible
M
,
Schlombs
K
,
Kaiser
K
, et al:
Technical validation of an RT-qPCR in vitro diagnostic test system for the determination of breast cancer molecular subtypes by quantification of ERBB2, ESR1, PGR and MKI67 mRNA levels from formalin-fixed paraffin-embedded breast tumor specimens
.
BMC Cancer
16
:
398
,
2016
33
Wu
NC
,
Wong
W
,
Ho
KE
, et al:
Comparison of central laboratory assessments of ER, PR, HER2, and Ki67 by lHC/FISH and the corresponding mRNAs (ESR1, PGR, ERBB2, and MKi67) by RT-qPCR on an automated, broadly deployed diagnostic platform
.
Breast Cancer Res Treat
172
:
327
338
,
2018
34
Varga
Z
,
Lebeau
A
,
Bu
H
, et al:
An international reproducibility study validating quantitative determination of ERBB2, ESR1, PGR, and MKI67 mRNA in breast cancer using Mamma Typer
.
Breast Cancer Res
19
:
55
,
2017
35
Hyeon
J
,
Cho
SY
,
Hong
ME
, et al:
NanoString nCounter approach in breast cancer: A comparative analysis with quantitative real-time polymerase chain reaction, in situ hybridization, and immunohistochemistry
.
J Breast Cancer
20
:
286
296
,
2017
36
Cai
H
,
Guo
W
,
Zhang
S
, et al:
A qualitative transcriptional signature to reclassify estrogen receptor status of breast cancer patients
.
Breast Cancer Res Treat
170
:
271
277
,
2018
37
Wilson
TR
,
Xiao
Y
,
Spoerke
JM
, et al:
Development of a robust RNA-based classifier to accurately determine ER, PR, and HER2 status in breast cancer clinical samples
.
Breast Cancer Res Treat
148
:
315
325
,
2014
38
Welsh
AW
,
Moeder
CB
,
Kumar
S
, et al:
Standardization of estrogen receptor measurement in breast cancer suggests false-negative results are a function of threshold intensity rather than percentage of positive cells
.
J Clin Oncol
29
:
2978
2984
,
2011
39
Karn
T
,
Metzler
D
,
Ruckhäberle
E
, et al:
Data-driven derivation of cutoffs from a pool of 3,030 Affymetrix arrays to stratify distinct clinical types of breast cancer
.
Breast Cancer Res Treat
120
:
567
579
,
2010
40
Bordeaux
JM
,
Cheng
H
,
Welsh
AW
, et al:
Quantitative in situ measurement of estrogen receptor mRNA predicts response to tamoxifen
.
PLoS One
7
:
e36559
,
2012
41
Wesseling
J
,
Tinterri
C
,
Sapino
A
, et al:
An international study comparing conventional versus mRNA level testing (TargetPrint) for ER, PR, and HER2 status of breast cancer
.
Virchows Arch
469
:
297
304
,
2016
42
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
25
:
816
823
,
2014
43
Dekker
TJ
,
ter Borg
S
,
Hooijer
GK
, et al:
Quality assessment of estrogen receptor and progesterone receptor testing in breast cancer using a tissue microarray-based approach
.
Breast Cancer Res Treat
152
:
247
252
,
2015
[erratum: Breast Cancer Res Treat 152:253, 2015]
44
Zarrella
ER
,
Coulter
M
,
Welsh
AW
, et al:
Automated measurement of estrogen receptor in breast cancer: A comparison of fluorescent and chromogenic methods of measurement
.
Lab Invest
96
:
1016
1025
,
2016
45
Viale
G
,
Slaets
L
,
de Snoo
FA
, et al:
Discordant assessment of tumor biomarkers by histopathological and molecular assays in the EORTC randomized controlled 10041/BIG 03-04 MINDACT trial breast cancer: Intratumoral heterogeneity and DCIS or normal tissue components are unlikely to be the cause of discordance
.
Breast Cancer Res Treat
155
:
463
469
,
2016
46
Viale
G
,
de Snoo
FA
,
Slaets
L
, et al:
Immunohistochemical versus molecular (BluePrint and MammaPrint) subtyping of breast carcinoma: Outcome results from the EORTC 10041/BIG 3-04 MINDACT trial
.
Breast Cancer Res Treat
167
:
123
131
,
2018
47
Reisenbichler
ES
,
Lester
SC
,
Richardson
AL
, et al:
Interobserver concordance in implementing the 2010 ASCO/CAP recommendations for reporting ER in breast carcinomas: A demonstration of the difficulties of consistently reporting low levels of ER expression by manual quantification
.
Am J Clin Pathol
140
:
487
494
,
2013
48
Cserni
G
,
Francz
M
,
Kálmán
E
, et al:
Estrogen receptor negative and progesterone receptor positive breast carcinomas: How frequent are they?
Pathol Oncol Res
17
:
663
668
,
2011
49
Bae
SY
,
Kim
S
,
Lee
JH
, et al:
Poor prognosis of single hormone receptor-positive breast cancer: Similar outcome as triple-negative breast cancer
.
BMC Cancer
15
:
138
,
2015
50
Zhang
Z
,
Wang
J
,
Skinner
KA
, et al:
Pathological features and clinical outcomes of breast cancer according to levels of oestrogen receptor expression
.
Histopathology
65
:
508
516
,
2014
51
Landmann
A
,
Farrugia
DJ
,
Zhu
L
, et al:
Low estrogen receptor (ER)-positive breast cancer and neoadjuvant systemic chemotherapy: Is response similar to typical ER-positive or ER-negative disease?
Am J Clin Pathol
150
:
34
42
,
2018
52
Spring
LM
,
Gupta
A
,
Reynolds
KL
, et al:
Neoadjuvant endocrine therapy for estrogen receptor-positive breast cancer: A systematic review and meta-analysis
.
JAMA Oncol
2
:
1477
1486
,
2016
53
Ejlertsen
B
,
Aldridge
J
,
Nielsen
KV
, et al:
Prognostic and predictive role of ESR1 status for postmenopausal patients with endocrine-responsive early breast cancer in the Danish cohort of the BIG 1-98 trial
.
Ann Oncol
23
:
1138
1144
,
2012
54
Bui
MM
,
Riben
MW
,
Allison
KH
, et al:
Quantitative image analysis of human epidermal growth factor receptor 2 immunohistochemistry for breast cancer: Guideline from the College of American Pathologists
.
Arch Pathol Lab Med
143
:
1180
1195
,
2019
55
Hwang
KT
,
Kim
EK
,
Jung
SH
, et al:
Tamoxifen therapy improves overall survival in luminal A subtype of ductal carcinoma in situ: A study based on nationwide Korean Breast Cancer Registry database
.
Breast Cancer Res Treat
169
:
311
322
,
2018
56
Chaudhary
LN
,
Jawa
Z
,
Hanif
A
, et al:
Does progesterone receptor matter in the risk of recurrence for patients with ductal carcinoma in situ
?
WMJ
117
:
62
67
,
2018
57
Cuzick
J
,
Sestak
I
,
Pinder
SE
, et al:
Effect of tamoxifen and radiotherapy in women with locally excised ductal carcinoma in situ: Long-term results from the UK/ANZ DCIS trial
.
Lancet Oncol
12
:
21
29
,
2011
58
Allred
DC
,
Anderson
SJ
,
Palk
S
, et al:
Adjuvant tamoxifen reduces subsequent breast cancer in women with estrogen receptor–positive ductal carcinoma in situ: A study based on NSABP protocol B-24
.
J Clin Oncol
30
:
1268
1273
,
2012
59
Kraus
JA
,
Dabbs
DJ
,
Beriwal
S
, et al:
Semi-quantitative immunohistochemical assay versus oncotype DX qRT-PCR assay for estrogen and progesterone receptors: An independent quality assurance study
.
Mod Pathol
25
:
869
876
,
2012
60
Kim
C
,
Tang
G
,
Pogue-Geile
KL
, et al:
Estrogen receptor (ESR1) mRNA expression and benefit from tamoxifen in the treatment and prevention of estrogen receptor–positive breast cancer
.
J Clin Oncol
29
:
4160
4167
,
2011
61
Rimm
DL
,
Leung
SCY
,
McShane
LM
, et al:
An international multicenter study to evaluate reproducibility of automated scoring for assessment of Ki67 in breast cancer
.
Mod Pathol
32
:
59
69
,
2019
62
Jorns
JM
,
Healy
P
,
Zhao
L
:
Review of estrogen receptor, progesterone receptor, and HER-2/neu immunohistochemistry impacts on treatment for a small subset of breast cancer patients transferring care to another institution
.
Arch Pathol Lab Med
137
:
1660
1663
,
2013
63
Engelberg
JA
,
Retallack
H
,
Balassanian
R
, et al:
“Score the Core” Web-based pathologist training tool improves the accuracy of breast cancer IHC4 scoring
.
Hum Pathol
46
:
1694
1704
,
2015
64
Elmore
JG
,
Tosteson
AN
,
Pepe
MS
, et al:
Evaluation of 12 strategies for obtaining second opinions to improve interpretation of breast histopathology: Simulation study
.
BMJ
353
:
i3069
,
2016
65
Tosteson
ANA
,
Yang
Q
,
Nelson
HD
, et al:
Second opinion strategies in breast pathology: A decision analysis addressing over-treatment, under-treatment, and care costs
.
Breast Cancer Res Treat
167
:
195
203
,
2018
66
Lloyd
MC
,
Allam-Nandyala
P
,
Purohit
CN
, et al:
Using image analysis as a tool for assessment of prognostic and predictive biomarkers for breast cancer: How reliable is it?
J Pathol Inform
1
:
29
,
2010
67
Barnes
M
,
Srinivas
C
,
Bai
I
, et al:
Whole tumor section quantitative image analysis maximizes between-pathologists' reproducibility for clinical immunohistochemistry-based biomarkers
.
Lab Invest
97
:
1508
1515
,
2017
68
Ahern
TP
,
Beck
AH
,
Rosner
BA
, et al:
Continuous measurement of breast tumour hormone receptor expression: A comparison of two computational pathology platforms
.
J Clin Pathol
70
:
428
434
,
2017
69
Peck
M
,
Moffat
D
,
Latham
B
, et al:
Review of diagnostic error in anatomical pathology and the role and value of second opinions in error prevention
.
J Clin Pathol
71
:
995
1000
,
2018
70
Kuroda
H
,
Muroi
N
,
Hayashi
M
, et al:
Oestrogen receptor-negative/progesterone receptor-positive phenotype of invasive breast carcinoma in Japan: Re-evaluated using immunohistochemical staining
.
Breast Cancer
26
:
249
254
,
2019
71
Knoop
AS
,
Lænkholm
AV
,
Jensen
MB
, et al:
Estrogen receptor, progesterone receptor, HER2 status and Ki67 index and responsiveness to adjuvant tamoxifen in postmenopausal high-risk breast cancer patients enrolled in the DBCG 77C trial
.
Eur J Cancer
50
:
1412
1421
,
2014
72
Dowsett
M
,
Allred
C
,
Knox
J
, et al:
Relationship between quantitative estrogen and progesterone receptor expression and human epidermal growth factor receptor 2 (HER-2) status with recurrence in the Arimidex, Tamoxifen, Alone or in Combination trial
.
J Clin Oncol
26
:
1059
1065
,
2008
73
Weiss
A
,
Chavez-MacGregor
M
,
Lichtensztajn
DY
, et al:
Validation study of the American Joint Committee on Cancer Eighth Edition prognostic stage compared with the anatomic stage in breast cancer
.
JAMA Oncol
4
:
203
209
,
2018
74
Foley
NM
,
Coll
JM
,
Lowery
AJ
, et al:
Re-appraisal of estrogen receptor negative/progesterone receptor positive (ER−/PR+) breast cancer phenotype: True subtype or technical artefact?
Pathol Oncol Res
24
:
881
884
,
2018
75
Allison
KH
,
Kandalaft
PL
,
Sitlani
CM
, et al:
Routine pathologic parameters can predict Oncotype DX recurrence scores in subsets of ER positive patients: Who does not always need testing?
Breast Cancer Res Treat
131
:
413
424
,
2012
76
Ahmed
SS
,
Thike
AA
,
Zhang
K
, et al:
Clinicopathological characteristics of oestrogen receptor negative, progesterone receptor positive breast cancers: Re-evaluating subsets within this group
.
J Clin Pathol
70
:
320
326
,
2017
77
Turner
BM
,
Skinner
KA
,
Tang
P
, et al:
Use of modified Magee equations and histologic criteria to predict the Oncotype DX recurrence score
.
Mod Pathol
28
:
921
931
,
2015
78
Klein
ME
,
Dabbs
DJ
,
Shuai
Y
, et al:
Prediction of the Oncotype DX recurrence score: Use of pathology-generated equations derived by linear regression analysis
.
Mod Pathol
26
:
658
664
,
2013
79
Farrugia
DJ
,
Landmann
A
,
Zhu
L
, et al:
Magee equation 3 predicts pathologic response to neoadjuvant systemic chemotherapy in estrogen receptor positive, HER2 negative/equivocal breast tumors
.
Mod Pathol
30
:
1078
1085
,
2017
80
Bhargava
R
,
Clark
BZ
,
Dabbs
DJ
:
Breast cancers with Magee equation score of less than 18, or 18–25 and mitosis score of 1, do not require Oncotype DX testing: A value study
.
Am J Clin Pathol
151
:
316
323
,
2019
81
Yeo
B
,
Zabaglo
L
,
Hills
M
, et al:
Clinical utility of the IHC4+C score in oestrogen receptor-positive early breast cancer: A prospective decision impact study
.
Br J Cancer
113
:
390
395
,
2015
82
Lee
SB
,
Kim
J
,
Sohn
G
, et al:
A nomogram for predicting the Oncotype DX recurrence score in women with T1-3N0-1miM0 hormone receptor–positive, human epidermal growth factor 2 (HER2)–negative breast cancer
.
Cancer Res Treat
51
:
1073
1085
,
2019
83
Cuzick
J
,
Dowsett
M
,
Pineda
S
, et al:
Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and comparison with the Genomic Health recurrence score in early breast cancer
.
J Clin Oncol
29
:
4273
4278
,
2011
84
Tan
W
,
Luo
W
,
Jia
W
, et al:
A combination of Nottingham prognostic index and IHC4 score predicts pathological complete response of neoadjuvant chemotherapy in estrogen receptor positive breast cancer
.
Oncotarget
7
:
87312
87322
,
2016
85
Sheri
A
,
Smith
IE
,
Hills
M
, et al:
Relationship between IHC4 score and response to neo-adjuvant chemotherapy in estrogen receptor-positive breast cancer
.
Breast Cancer Res Treat
164
:
395
400
,
2017
86
Lakhanpal
R
,
Sestak
I
,
Shadbolt
B
, et al:
IHC4 score plus clinical treatment score predicts locoregional recurrence in early breast cancer
.
Breast
29
:
147
152
,
2016
87
Kim
HS
,
Umbricht
CB
,
Illei
PB
, et al:
Optimizing the use of gene expression profiling in early-stage breast cancer
.
J Clin Oncol
34
:
4390
4397
,
2016
88
Regan
MM
,
Francis
PA
,
Pagani
O
, et al:
Absolute benefit of adjuvant endocrine therapies for premenopausal women with hormone receptor-positive, human epidermal growth factor receptor 2–negative early breast cancer: TEXT and SOFT trials
.
J Clin Oncol
34
:
2221
2231
,
2016
89
Albert
JM
,
Gonzalez-Angulo
AM
,
Guray
M
, et al:
Patients with only 1 positive hormone receptor have increased locoregional recurrence compared with patients with estrogen receptor-positive progesterone receptor-positive disease in very early stage breast cancer
.
Cancer
117
:
1595
1601
,
2011
90
Ahn
SG
,
Yoon
CI
,
Lee
JH
, et al:
Low PR in ER(+)/HER2(−) breast cancer: High rates of TP53 mutation and high SUV
.
Endocr Relat Cancer
doi:
[epub ahead of print on November 1, 2018]
91
Raghav
KP
,
Hernandez-Aya
LF
,
Lei
X
, et al:
Impact of low estrogen/progesterone receptor expression on survival outcomes in breast cancers previously classified as triple negative breast cancers
.
Cancer
118
:
1498
1506
,
2012
92
Honma
N
,
Horii
R
,
Iwase
T
, et al:
Proportion of estrogen or progesterone receptor expressing cells in breast cancers and response to endocrine therapy
.
Breast
23
:
754
762
,
2014
93
Chen
T
,
Zhang
N
,
Moran
MS
, et al:
Borderline ER-positive primary breast cancer gains no significant survival benefit from endocrine therapy: A systematic review and meta-analysis
.
Clin Breast Cancer
18
:
1
8
,
2018
94
Balduzzi
A
,
Bagnardi
V
,
Rotmensz
N
, et al:
Survival outcomes in breast cancer patients with low estrogen/progesterone receptor expression
.
Clin Breast Cancer
14
:
258
264
,
2014
95
Gloyeske
NC
,
Dabbs
DJ
,
Bhargava
R
:
Low ER+ breast cancer: Is this a distinct group?
Am J Clin Pathol
141
:
697
701
,
2014
96
Deyarmin
B
,
Kane
JL
,
Valente
AL
, et al:
Effect of ASCO/CAP guidelines for determining ER status on molecular subtype
.
Ann Surg Oncol
20
:
87
93
,
2013
97
Yi
M
,
Huo
L
,
Koenig
KB
, et al:
Which threshold for ER positivity? A retrospective study based on 9639 patients
.
Ann Oncol
25
:
1004
1011
,
2014
98
Khoshnoud
MR
,
Löfdahl
B
,
Fohlin
H
, et al:
Immunohistochemistry compared to cytosol assays for determination of estrogen receptor and prediction of the long-term effect of adjuvant tamoxifen
.
Breast Cancer Res Treat
126
:
421
430
,
2011
99
Badve
SS
,
Baehner
FL
,
Gray
RP
, et al:
Estrogen- and progesterone-receptor status in ECOG 2197: Comparison of immunohistochemistry by local and central laboratories and quantitative reverse transcription polymerase chain reaction by central laboratory
.
J Clin Oncol
26
:
2473
2481
,
2008
[Erratum: J Clin Oncol 26:3472, 2008]
100
Cheang
MC
,
Treaba
DO
,
Speers
CH
, et al:
Immunohistochemical detection using the new rabbit monoclonal antibody SP1 of estrogen receptor in breast cancer is superior to mouse monoclonal antibody 1D5 in predicting survival
.
J Clin Oncol
24
:
5637
5644
,
2006
101
Dowsett
M
:
Estrogen receptor: Methodology matters
.
J Clin Oncol
24
:
5626
5628
,
2006
102
Harvey
JM
,
Clark
GM
,
Osborne
CK
, et al:
Estrogen receptor status by immunohistochemistry is superior to the ligand-binding assay for predicting response to adjuvant endocrine therapy in breast cancer
.
J Clin Oncol
17
:
1474
1481
,
1999
103
Fisher
ER
,
Anderson
S
,
Dean
S
, et al:
Solving the dilemma of the immunohistochemical and other methods used for scoring estrogen receptor and progesterone receptor in patients with invasive breast carcinoma
.
Cancer
103
:
164
173
,
2005
104
Molino
A
,
Micciolo
R
,
Turazza
M
, et al:
Prognostic significance of estrogen receptors in 405 primary breast cancers: A comparison of immunohistochemical and biochemical methods
.
Breast Cancer Res Treat
45
:
241
249
,
1997
105
Bouchard-Fortier
A
,
Provencher
L
,
Blanchette
C
, et al:
Prognostic and predictive value of low estrogen receptor expression in breast cancer
.
Curr Oncol
24
:
e106
e114
,
2017
106
Iwamoto
T
,
Booser
D
,
Valero
V
, et al:
Estrogen receptor (ER) mRNA and ER-related gene expression in breast cancers that are 1% to 10% ER-positive by immunohistochemistry
.
J Clin Oncol
30
:
729
734
,
2012
107
Pérez
T
,
Makrestsov
N
,
Garatt
J
, et al:
Modeling Canadian quality control test program for steroid hormone receptors in breast cancer: Diagnostic accuracy study
.
Appl Immunohistochem Mol Morphol
24
:
679
687
,
2016
108
Torlakovic
EE
,
Nielsen
S
,
Vyberg
M
, et al:
Getting controls under control: The time is now for immunohistochemistry
.
J Clin Pathol
68
:
879
882
,
2015
109
Torlakovic
EE
,
Nielsen
S
,
Francis
G
, et al:
Standardization of positive controls in diagnostic immunohistochemistry: Recommendations from the International Ad Hoc Expert Committee
.
Appl Immunohistochem Mol Morphol
23
:
1
18
,
2015
110
Cheung
CC
,
Taylor
CR
,
Torlakovic
EE
:
An audit of failed immunohistochemical slides in a clinical laboratory: The role of on-slide controls
.
Appl Immunohistochem Mol Morphol
25
:
308
312
,
2017
111
Bogen
SA
:
A root cause analysis into the high error rate in clinical immunohistochemistry
.
Appl Immunohistochem Mol Morphol
27
:
329
338
,
2019
112
Vani
K
,
Sompuram
SR
,
Schaedle
AK
, et al:
The importance of epitope density in selecting a sensitive positive IHC control
.
J Histochem Cytochem
65
:
463
477
,
2017
113
Sompuram
SR
,
Vani
K
,
Tracey
B
, et al:
Standardizing immunohistochemistry: A new reference control for detecting staining problems
.
J Histochem Cytochem
63
:
681
690
,
2015
114
Vani
K
,
Sompuram
SR
,
Naber
SP
, et al:
Levey-Jennings analysis uncovers unsuspected causes of immunohistochemistry stain variability
.
Appl Immunohistochem Mol Morphol
24
:
688
694
,
2016
115
Sompuram
SR
,
Vani
K
,
Schaedle
AK
, et al:
Selecting an optimal positive IHC control for verifying antigen retrieval
.
J Histochem Cytochem
67
:
275
289
,
2019
116
Prat
A
,
Cheang
MC
,
Martín
M
, et al:
Prognostic significance of progesterone receptor–positive tumor cells within immunohistochemically defined luminal A breast cancer
.
J Clin Oncol
31
:
203
209
,
2013
117
Reference deleted
.
118
Cameron
MA
:
Commission of Inquiry on Hormone Receptor Testing
.
St John's, Newfoundland, Canada
,
Government of Newfoundland and Labrador
,
2009
119
Aeffner
F
,
Wilson
K
,
Martin
NT
, et al:
The gold standard paradox in digital image analysis: Manual versus automated scoring as ground truth
.
Arch Pathol Lab Med
141
:
1267
1275
,
2017
120
Khazai
L
,
Middleton
LP
,
Goktepe
N
, et al:
Breast pathology second review identifies clinically significant discrepancies in over 10% of patients
.
J Surg Oncol
111
:
192
197
,
2015
121
Allen
TC
:
Second opinions: Pathologists' preventive medicine
.
Arch Pathol Lab Med
137
:
310
311
,
2013
122
Sparano
JA
,
Gray
RJ
,
Makower
DF
, et al:
Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer
.
N Engl J Med
379
:
111
121
,
2018
123
Sparano
JA
,
Gray
RJ
,
Makower
DF
, et al:
Prospective validation of a 21-gene expression assay in breast cancer
.
N Engl J Med
373
:
2005
2014
,
2015
124
Dowsett
M
,
Turner
N
:
Estimating risk of recurrence for early breast cancer: Integrating clinical and genomic risk
.
J Clin Oncol
37
:
689
692
,
2019
125
Andre
F
,
Ismaila
N
,
Henry
NL
, et al:
Use of biomarkers to guide decisions on adjuvant systemic therapy for women with early-stage invasive breast cancer: ASCO clinical practice guideline update—Integration of results from TAILORx
.
J Clin Oncol
37
:
1956
1964
,
2019
126
Krop
I
,
Ismaila
N
,
Andre
F
, et al:
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 focused update
.
J Clin Oncol
35
:
2838
2847
,
2017
127
Harris
LN
,
Ismaila
N
,
McShane
LM
, et al:
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
.
J Clin Oncol
34
:
1134
1150
,
2016
128
Allred
DC
,
Bryant
J
,
Land
S
, et al:
Estrogen receptor expression as a predictive marker of the effectiveness of tamoxifen in the treatment of DCIS: Findings from NSABP Protocol B-24
.
Breast Cancer Res Treat
76
:
S36
,
2002
129
Houghton
J
,
George
WD
,
Cuzick
J
, et al:
Radiotherapy and tamoxifen in women with completely excised ductal carcinoma in situ of the breast in the UK, Australia, and New Zealand: Randomised controlled trial
.
Lancet
362
:
95
102
,
2003
130
DeCensi
A
,
Puntoni
M
,
Guerrieri-Gonzaga
A
, et al:
Randomized placebo controlled trial of low-dose tamoxifen to prevent local and contralateral recurrence in breast intraepithelial neoplasia
.
J Clin Oncol
37
:
1629
1637
,
2019
131
Wolff
AC
,
Hammond
MEH
,
Allison
KH
, et al:
Human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline focused update
.
Arch Pathol Lab Med
142
:
1364
1382
,
2018
132
Burstein
HJ
,
Lacchetti
C
,
Anderson
H
, et al:
Adjuvant endocrine therapy for women with hormone receptor–positive breast cancer: ASCO clinical practice guideline focused update
.
J Clin Oncol
37
:
423
438
,
2019
133
Giordano
SH
,
Temin
S
,
Chandarlapaty
S
, et al:
Systemic therapy for patients with advanced human epidermal growth factor receptor 2–positive breast cancer: ASCO clinical practice guideline update
.
J Clin Oncol
36
:
2736
2740
,
2018
APPENDIX

Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Clinical Practice Guideline Update Expert Panel Membership

Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Clinical Practice Guideline Update Expert Panel Membership
Estrogen and Progesterone Receptor Testing in Breast Cancer: ASCO/CAP Clinical Practice Guideline Update Expert Panel Membership

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Estrogen and Progesterone Receptor Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Guideline Update

The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted.

Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/site/ifc.

Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

Kimberly H. Allison

Honoraria: Genentech

Consulting or Advisory Role: Mammotome

Expert Testimony: Kaiser Permanente

Mitchell Dowsett

Honoraria: Myriad Genetics

Consulting or Advisory Role: GTx, Radius Health, Orion Pharma GmbH

Research Funding: Pfizer (Inst), Radius Health (Inst)

Travel, Accommodations, Expenses: Pfizer, Myriad Genetics

Other Relationship: Institute of Cancer Research

Lisa A. Carey

Research Funding: Innocrin Pharma (Inst), Syndax (Inst), Immunomedics (Inst), Novartis (Inst)

Patents, Royalties, Other Intellectual Property: Royalty-sharing agreement, investorship interest in licensed intellectual property to startup company, Falcon Therapeutics, which is designing neural stem cell-based therapy for glioblastoma multiforme (I)

Patrick L. Fitzgibbons

Stock and Other Ownership Interests: Johnson & Johnson, Pfizer, Medtronic, Merck

Daniel F. Hayes

Stock and Other Ownership Interests: OncImmune, InBiomotion

Consulting or Advisory Role: Cepheid, Freenome, Cellworks

Consulting or Advisory Role: CVS Caremark Breast Cancer Expert Panel, Agendia

Research Funding: AstraZeneca (Inst), Pfizer (Inst), Merrimack (Inst), Menarini Silicon Biosystems (Inst)

Patents, Royalties, Other Intellectual Property: Royalties from licensed technology; Diagnosis and treatment of breast cancer, patent No. US 8,790,878 B2, date of patent July 29, 2014, applicant proprietor University of Michigan, Daniel F. Hayes designated as inventor/co-inventor; circulating tumor cell capturing techniques and devices, patent No. US 8,951,484 B2, date of patent February 10, 2015, applicant proprietor University of Michigan, Daniel F. Hayes designated as inventor/co-inventor; method for predicting progression-free and overall survival at each follow-up time point during therapy of metastatic breast cancer patients using circulating tumor cells, patent No. 05725638.0-1223-US2005008602

Travel, Accommodations, Expenses: Menarini Silicon Biosystems

Other Relationship: Menarini

Sunil R. Lakhani

Consulting or Advisory Role: Suburban Hospital (Inst)

Travel, Accommodations, Expenses: Ventana Medical Systems

Mariana Chavez-MacGregor

Employment: MD Anderson Physician's Network

Honoraria: Pfizer, Eisai

Consulting or Advisory Role: Roche/Genentech

Research Funding: Novartis (Inst)

Expert Testimony: Abbott Laboratories, Pfizer

Travel, Accommodations, Expenses: Pfizer

Charles M. Perou

Leadership: GeneCentric

Stock and Other Ownership Interests: Bioclassifier, GeneCentric

Consulting or Advisory Role: Bioclassifier, GeneCentric, G1 Therapeutics, Ions Pharmaceuticals

Patents, Royalties, Other Intellectual Property: Royalties from PAM50 breast cancer gene patent application and from lung gene signature patent

Travel, Accommodations, Expenses: Takeda, Chugai Pharma

Meredith M. Regan

Consulting or Advisory Role: Ipsen (Inst)

Research Funding: Veridex (Inst), OncoGenex (Inst), Pfizer (Inst), Ipsen (Inst), Novartis (Inst), Merck (Inst), Ferring (Inst), Celgene (Inst), AstraZeneca (Inst), Pierre Fabre (Inst), Bayer (Inst), Bristol-Myers Squibb (Inst), Roche (Inst), Astellas Pharma (Inst), Medivation (Inst), Janssen (Inst), Millennium Pharmaceuticals (Inst), Sanofi (Inst), Sotio (Inst), Dendreon (Inst), TerSera (Inst)

Travel, Accommodations, Expenses: Bristol-Myers Squibb

David L. Rimm

Stock and Other Ownership Interests: Pixel Gear

Honoraria: Amgen, Bristol-Myers Squibb, Ventana Medical Systems

Consulting or Advisory Role: Biocept, Perkin Elmer, Bristol-Myers Squibb, AstraZeneca, Agendia, Ultivue, Merck, Daiichi Sankyo, GlaxoSmithKline, Konica Minolta, NanoString Technologies, NextCure, Cell Signaling Technology, Roche, Paige AI, Cepheid

Research Funding: Cepheid (Inst), Perkin Elmer (Inst), AstraZeneca/MedImmune (Inst), NextCure (Inst), Lilly (Inst), NanoString Technologies (Inst), Navigate Biopharma (Inst), Ultivue (Inst)

Patents, Royalties, Other Intellectual Property: Rarecyte circulating tumor cells; quantitative immunofluorescence (AQUA) (Inst)

Travel, Accommodations, Expenses: Ventana Medical Systems, Genentech, NextCure

W. Fraser Symmans

Stock and Other Ownership Interests: ISIS Pharmaceuticals, Nuvera Biosciences, Delphi Diagnostics

Consulting or Advisory Role: Merck, Almac Diagnostics

Travel, Accommodations, Expenses: Luminex, Merck

Emina E. Torlakovic

Honoraria: Janssen Latin America Oncology, Leica Biosystems, Janssen

Consulting or Advisory Role: Merck, Pfizer, Millipore, Bristol-Myers Squibb, Merck Sharp & Dohme, AstraZeneca, Janssen Oncology, Roche

Research Funding: AstraZeneca (Inst), Roche (Inst), Bristol-Myers Squibb, Merck, Pfizer (Inst)

Giuseppe Viale

Honoraria: MSD Oncology, Pfizer

Consulting or Advisory Role: Dako, Roche/Genentech, Astellas Pharma, Novartis, Bayer

Speakers Bureau: Roche/Genentech

Research Funding: Roche/Genentech, Ventana Medical Systems (Inst), Dako/Agilent Technologies (Inst)

Travel, Accommodations, Expenses: Roche, Celgene

Tracey F. Weisberg

Honoraria: Invitae

Research Funding: NanoString Technologies (Inst)

Antonio C. Wolff

Consulting or Advisory Role: Ionis Pharmaceuticals

Research Funding: Myriad Genetics (Inst), Pfizer (Inst), Biomarin (Inst), Celldex (Inst)

Patents, Royalties, Other Intellectual Property: Antonio Wolff has been named as inventor on one or more issued patents or pending patent applications relating to methylation in breast cancer and has assigned his rights to Johns Hopkins University (JHU) and participates in a royalty-sharing agreement with JHU

No other potential conflicts of interest were reported.

Author notes

Author support information (if applicable) appears at the end of this article.

Supplemental digital content is available for this article at www.archivesofpathology.org in the May 2020 table of contents.

K.H.A. and A.C.W. were Expert Panel co-chairs; M.E.H.H., M.D., and L.M.M. were members of the Steering Group with the co-chairs.

Competing Interests

This guideline was developed through a collaboration between the College of American Pathologists and the American Society of Clinical Oncology and has been published jointly by invitation and consent in both the Archives of Pathology & Laboratory Medicine and the Journal of Clinical Oncology. It has been edited in accordance with style standards established at the Journal of Clinical Oncology. Copyright 2019 College of American Pathologists and the American Society of Clinical Oncology. All rights reserved. No part of this document may be reproduced or transmitted in any form or by any means, electronic or mechanical, including photocopy, recording, or any information storage and retrieval system, without written permission by the College of American Pathologists or the American Society of Clinical Oncology.

Supplementary data