Context.—

Neoadjuvant systemic therapy refers to the use of systemic agent(s) for malignancy prior to surgical treatment and has recently emerged as an option for most breast cancer patients eligible for adjuvant systemic therapy. Consequently, treated breast carcinomas have become routine specimens in pathology practices. A standard protocol has not yet been universally adopted for the evaluation and reporting of these specimens. The American Joint Committee on Cancer staging system recognizes the challenges in staging breast carcinomas after neoadjuvant treatment and provides important data points but does not currently provide detailed guidance in estimating the residual tumor burden in the breast and lymph nodes. The Residual Cancer Burden system is the only Web-based system that quantifies treatment response as a continuous variable using residual tumor burden in the breast and the lymph nodes.

Objective.—

To provide clarifications and guidance for evaluation and reporting of postneoadjuvant breast specimens, discuss issues with the current staging and reporting systems, and provide specific suggestions for future modifications to the American Joint Committee on Cancer system and the Residual Cancer Burden calculator.

Data Sources.—

English-language literature on the subject and the data from the I-SPY 2, a multicenter, adaptive randomization phase 2 neoadjuvant platform trial for early-stage, high-risk breast cancer patients.

Conclusions.—

This article highlights challenges in the pathologic evaluation and reporting of treated breast carcinomas and provides recommendations and clarifications for pathologists and clinicians. It also provides specific recommendations for staging and discusses future directions.

Neoadjuvant systemic therapy (NST) in the treatment of breast cancer involves the administration of chemotherapy (NCT) or endocrine therapy prior to definitive surgery. Initially, NCT was implemented as a standard treatment for inflammatory and locally advanced inoperable breast cancers, but its use has broadened to include operable and early-stage breast cancers.13  Currently, NCT is considered as a treatment option for any breast cancer patient who is eligible for adjuvant chemotherapy. A measure of the success of NCT is pathologic complete response (pCR), defined as the absence of residual invasive cancer in the breast and in the lymph nodes after completion of NCT.4  Additionally, pCR can be used as a short-term intermediate endpoint for treatment efficacy in clinical trials. Typically, rates of pCR are highest in triple-negative breast carcinomas (TNBCs) (estrogen receptor [ER], progesterone receptor [PR], and HER2), as well as in HER2+ tumors, and lowest in low-grade, hormone receptor (HR)+ tumors.5  The US Food and Drug Administration (FDA)6  has issued guidance for the use of pCR to support accelerated drug approval. Although pooled clinical trial analysis supports a strong prognostic association between pCR and disease-free survival, observed differences in the rate of pCR between study treatment arms were not established as a trial-level predictor of survival benefit.7  However, there have been recent examples of trial-level prediction with contemporary treatments for specific breast cancer subtypes.8,9 

Multiple clinical trials have shown that the overall and disease-free survival rates for NST and adjuvant therapy are similar.1017  Despite the lack of survival benefit from the timing of chemotherapy relative to surgery, neoadjuvant therapy provides unique advantages over adjuvant therapy: (1) it enables more women to be eligible for breast-conserving surgery2,3,10,12,13,18,19; (2) it has the potential to downstage axillary disease to avoid axillary lymph node dissection; (3) it offers prognostic information based on the degree of response to treatment7,17,20,21; (4) pCR is most predictive of outcome in TNBC, HR/HER2+, and high-grade HR+/HER2 tumors7,22; (5) early assessment of drug efficacy can potentially facilitate changes in the therapeutic regimen and determine the use of additional therapy in patients who have a high volume of residual disease after NST2325 ; and lastly, (6) NCT can be used as a valuable research tool to understand the biology of treatment response in vivo and to develop predictive markers.26 

As an increasing number of breast cancer patients are treated with NCT, pathologic evaluation of posttreatment specimens has become critical to determine an accurate prognosis. Multiple classification systems have been developed to assess treatment response, but these differ with respect to the number of response categories, determination of tumor size and cellularity, inclusion of in situ carcinoma, and assessment of response in lymph nodes.4,2735  Despite the differences, tumor response in each system has been shown to correlate with outcome. The Residual Cancer Burden (RCB) system is unique among classification systems in that it uses the combination of residual tumor size and cellularity in the breast and responses in the nodes as a continuous variable.30  The current American Joint Committee on Cancer (AJCC) system recognizes the need to address residual carcinomas after NST differently from untreated tumors.36  However, posttreatment AJCC stage does not incorporate measures of response beyond tumor size or offer specific guidelines to address the difficult scenarios.

In the NCT setting, interpretation of the basic parameters such as tumor size, grade, and lymph node involvement can be challenging for pathologists because of therapy-related changes. Furthermore, the lack of clear guidelines on estimating residual tumor after NCT and the failure to adopt a classification system for universal use have created inconsistencies in the reporting of residual tumor burden in the breast and lymph nodes.3739  This article discusses the challenges in the pathologic examination of specimens after NCT, compares some of the available classification systems to highlight the issues and offer suggestions for improvement, recommends the essential elements to be included in the pathology report, and provides direction to maximize the benefits to be gained from future NCT trials.

Gross and Microscopic Changes in Primary Tumor

Accurate identification of the original tumor site in the breast specimen is the most important step for proper classification of treatment response (Figure 1). The general term fibrous tumor bed or tumor bed area refers to the grossly or radiologically identifiable area of breast parenchyma where the tumor was located before initiation of systemic therapy (Figure 2). It is relatively easy to identify residual tumor when there is minimal or no response to NCT. Tumors, particularly smaller ones that show complete or near-complete resolution on clinical examination and imaging studies, are difficult to visualize on gross examination. Even larger tumors with a pronounced response to treatment can be difficult to delineate in a background of dense breast tissue. As the gross findings are typically subtle, radiologic markers and careful palpation of the specimen slices are important to identify tumor bed area(s). Indeed, access to specimen radiography greatly improves the accuracy and confidence in pathologic evaluation after neoadjuvant treatment (Figure 3). Residual calcification patterns can also help to identify tumor location in the specimen radiograph. However, it is important to remember that calcifications are not always associated with invasive carcinoma, and the extent of calcification may not correlate with the disease extent. Lacking these, one must rely on a detailed description of the pretreatment tumor location (eg, quadrant, distance from nipple) or sutures placed by the surgeon or the microscopic proof of the tumor bed to ensure the prior tumor site was removed.

Figure 1

Schematic diagram demonstrates the different types of treatment responses seen in breast carcinomas after neoadjuvant chemotherapy.

Figure 1

Schematic diagram demonstrates the different types of treatment responses seen in breast carcinomas after neoadjuvant chemotherapy.

Close modal
Figure 2

Grossly identifiable fibrous tumor bed after neoadjuvant chemotherapy (arrows).

Figure 3. Specimen radiograph of a partial mastectomy with 2 separate tumors and corresponding biopsy clips (arrows).

Figure 4. A posttreatment partial mastectomy specimen serially sectioned and annotated to show the origin of sections in relation to the biopsy clip and radioactive seed used for localization of the tumor bed.

Figure 5. Low-power appearance of a tumor bed (outlined) characterized by pale vascularized stroma without glandular structures, surrounded by normal breast lobules. The arrows indicate the presence of prior core biopsy site lined by macrophages (hematoxylin-eosin, original magnification ×20).

Figure 6. Tumor bed of a high-grade mucinous carcinoma with complete response to neoadjuvant chemotherapy reveals only extracellular mucin without tumor cells (hematoxylin-eosin, original magnification ×40).

Figure 7. Cellular changes in residual tumor after chemotherapy demonstrate pleomorphic and bizarre nuclei but inconspicuous mitoses (hematoxylin-eosin, original magnification ×100).

Figure 2

Grossly identifiable fibrous tumor bed after neoadjuvant chemotherapy (arrows).

Figure 3. Specimen radiograph of a partial mastectomy with 2 separate tumors and corresponding biopsy clips (arrows).

Figure 4. A posttreatment partial mastectomy specimen serially sectioned and annotated to show the origin of sections in relation to the biopsy clip and radioactive seed used for localization of the tumor bed.

Figure 5. Low-power appearance of a tumor bed (outlined) characterized by pale vascularized stroma without glandular structures, surrounded by normal breast lobules. The arrows indicate the presence of prior core biopsy site lined by macrophages (hematoxylin-eosin, original magnification ×20).

Figure 6. Tumor bed of a high-grade mucinous carcinoma with complete response to neoadjuvant chemotherapy reveals only extracellular mucin without tumor cells (hematoxylin-eosin, original magnification ×40).

Figure 7. Cellular changes in residual tumor after chemotherapy demonstrate pleomorphic and bizarre nuclei but inconspicuous mitoses (hematoxylin-eosin, original magnification ×100).

Close modal

It is important for the grossing pathologist (or pathology assistant) to have the information regarding the number, size, and location of tumor(s) and biopsy clips and any involvement of skin, chest wall, or nipple before treatment, and any documented response to therapy on clinical notes and imaging studies. As with any resection specimen, when the residual tumor or tumor bed is identified, the size of the tumor bed area and any grossly recognizable residual tumor should be documented, at least in 2 greatest dimensions, along with its morphologic characteristics and relationship to the surgical margins.

Although there are no strict guidelines regarding the extent to which tumor bed should be sampled, it is recommended that if there is an obvious tumor bed on gross examination that is large (>3 cm), one section per centimeter of the pretreatment carcinoma size is reasonable. If the residual tumor bed is small (≤3 cm), the entire fibrous area should be submitted for pathologic examination. To determine the dimensions and cellularity of the residual invasive cancer, it is necessary to map the tumor bed and correlate the microscopic sections with gross findings. An important objective is to map out a complete cross section of the tumor bed along its longest axis, because residual carcinoma could be present in any region of the tumor bed (Figure 4). Any additional sampling should be based on other gross and imaging findings in the specimen, as well as pretreatment tumor characteristics and response to treatment from clinical and imaging reports. The RCB system offers a prescriptive protocol with recommendations on tumor sampling on its Web site (http://www.mdanderson.org/breastcancer_RCB).

Microscopically, the tumor bed is characterized by an irregular area of vascularized fibrous stroma that either lacks or has scant normal ducts and lobules (Figure 5). The stroma may exhibit varying degrees of edema, myxoid change, fibrosis with or without elastosis, residual calcification, and inflammatory cells. The inflammatory response typically contains lymphocytes, plasma cells, mast cells, macrophages, and hemosiderin-laden macrophages, sometimes in loose aggregates or large sheets. As with calcifications, extracellular mucin associated with carcinoma does not resolve in the breast or in the metastatic sites after NCT (Figure 6). It is important to confirm the presence of tumor bed microscopically when there is a complete response to therapy. Although some clinicians may find it useful for comparison, the size of the tumor bed is not a reliable indicator of the pretreatment tumor size, as a significant response to treatment shrinks the overall area of a tumor.

Therapy-related changes in residual tumor, if present, are not specific to any agent or regimen. The most common therapy-related change is decrease in cellularity. Additional changes seen in residual carcinoma after NCT include increased cytoplasm, cytoplasmic vacuolization and eosinophilia, pleomorphic and bizarre nuclei, and decreased mitotic activity (Figure 7). Prominent stromal retraction artifacts around the tumor nests are common and should not be misinterpreted as lymphatic or vascular invasion. Residual single tumor cells in cases with near-complete response can be difficult to discern from inflammatory cells on routine hematoxylin-eosin stain. In such cases, immunohistochemical (IHC) stains for cytokeratin AE1/3 can be used to delineate the distribution of tumor cells. Residual ductal carcinoma in situ (DCIS) trapped in fibrous stroma can be mistaken for invasive carcinoma (Figure 8). Occasionally, lymphovascular invasion (LVI) can be the only residual disease after chemotherapy and should not be mistaken for residual DCIS.40  Many of the challenges listed above can be resolved by IHC for epithelial, myoepithelial, and endothelial markers.

Figure 8

Residual ductal carcinoma in situ trapped in fibroblastic stroma mimics invasive carcinoma. Immunostain for smooth muscle myosin reveals intact myoepithelial cells around the distorted in situ carcinoma (inset) (hematoxylin-eosin, original magnification ×40; original magnification ×40 [inset]).

Figure 9. Tumor bed with single nodule of residual tumor (hematoxylin-eosin, original magnification ×20).

Figure 10. Partial response to therapy. A, Nodules of residual tumor (outlined). B, Tumor bed with multiple clusters of tumor cells (outlined) (hematoxylin-eosin, original magnification ×20).

Figure 11. Lymph node with complete response to chemotherapy shows a large area of node replaced by fibrosis and aggregates of macrophages (hematoxylin-eosin, original magnification ×20).

Figure 12. A lymph node after neoadjuvant chemotherapy is completely replaced by fibrosis with scattered single tumor cells, better visible on immunostain for cytokeratin AE1/3 (hematoxylin-eosin, original magnification ×40; original magnification ×40 [inset]).

Figure 8

Residual ductal carcinoma in situ trapped in fibroblastic stroma mimics invasive carcinoma. Immunostain for smooth muscle myosin reveals intact myoepithelial cells around the distorted in situ carcinoma (inset) (hematoxylin-eosin, original magnification ×40; original magnification ×40 [inset]).

Figure 9. Tumor bed with single nodule of residual tumor (hematoxylin-eosin, original magnification ×20).

Figure 10. Partial response to therapy. A, Nodules of residual tumor (outlined). B, Tumor bed with multiple clusters of tumor cells (outlined) (hematoxylin-eosin, original magnification ×20).

Figure 11. Lymph node with complete response to chemotherapy shows a large area of node replaced by fibrosis and aggregates of macrophages (hematoxylin-eosin, original magnification ×20).

Figure 12. A lymph node after neoadjuvant chemotherapy is completely replaced by fibrosis with scattered single tumor cells, better visible on immunostain for cytokeratin AE1/3 (hematoxylin-eosin, original magnification ×40; original magnification ×40 [inset]).

Close modal

Determining residual tumor size can be difficult after treatment. It is easier to determine the size of the residual carcinoma when the tumor shrinks concentrically, leaving one contiguous focus, an uncommon scenario after NCT (Figure 9). Most treated tumors with moderate to marked response to therapy leave scattered nodules of residual tumor, multiple clusters, or single tumor cells in the tumor bed (Figure 10, A and B).41  For a detailed discussion on how to measure residual tumor size for the AJCC and RCB systems, please refer to the corresponding sections.

Gross and Microscopic Changes in Lymph Nodes

It may be difficult to identify 10 or more lymph nodes from an axillary dissection because of nodal atrophy and fibrosis induced by chemotherapy.42  Therefore, axillary fat should be carefully searched for lymph nodes, whether it is from an axillary dissection or sentinel lymph node (SLN) biopsy, and all identified nodes without grossly visible tumor should be sliced at 2- to 3-mm intervals perpendicular to the long axis of the node and submitted entirely.43  In instances where very few nodes are found on palpation, submitting fibrotic areas and the tissue around the vessels may reveal additional small lymph nodes.

In most cases, the response in nodal metastases mirrors the response observed in the primary tumor (Figure 1).44  Nodal metastases completely eradicated by treatment are replaced by fibrous scars or sheets of macrophages (Figure 11). It is rare to have a large subcapsular scar in axillary lymph nodes of patients who undergo primary surgery for breast carcinoma. Therefore, it is important to document the presence of scars or other findings in negative nodes that most likely represent treated metastasis. It is possible that a prior micrometastasis or smaller metastasis may resolve entirely without leaving a fibrous scar. Residual disease in metastatic nodes with partial response to NCT is often distributed either as single cells or as clusters of tumor cells surrounded by fibrosis. It is recommended that pathologists document any treatment effect in residual metastases. Please see the pertinent section below for measuring the size of metastases after treatment.

SLN Biopsy After NCT

Because a large proportion of patients with TNBC and HER2+ cancer achieve pCR in both breast and axilla, many centers are performing SLN surgery to avoid the morbidity of axillary dissection.4547  It should be emphasized to the clinical team to place a biopsy clip in the lymph node at the time of fine-needle aspiration or image-guided core needle biopsy of a clinically suspicious or positive node (or prior to initiation of systemic therapy if not placed at the time of aspiration or core biopsy). This will ensure removal of the biopsied positive node or suspicious node at the time of definitive surgery. It is important for pathologists to document in the pathology report whether the lymph node with marker clip was removed or not at the time of surgery.

Intraoperative evaluation of SLNs is quite challenging because of fibrosis and treatment-related changes. In an NCT setting, using touch imprints only for intraoperative evaluation of lymph nodes can lead to a higher false-negative rate. Gimbergues et al48  reported lower sensitivity and negative predictive value in detecting tumor cells in the SLN using touch imprints after NCT. The authors concluded that the lower sensitivity was related to a higher proportion of cases with micrometastasis and isolated tumor cells (ITCs) after NCT. Although frozen sections may reduce the overall number of false-negative results, the rate may still be significantly high in lobular carcinomas, smaller metastases, and ITCs.

Routine use of cytokeratin to detect metastasis is not currently recommended for SLNs after NCT. Pathologists should use their judgment to determine if a case would benefit from cytokeratin stain. Some of the clinical scenarios where cytokeratin stain may be useful include prior positive node from a lobular carcinoma or HR+ tumor and any node with scattered single tumor cells embedded in therapy-related fibro-inflammatory changes or any atypical cell that is difficult to characterize as tumor versus nontumor (Figure 12). Given the challenges of identifying residual lymph node metastasis in the NCT setting, pathologists should have a low threshold for using IHC to decrease the chance of missing metastasis.

ER, PR, and HER2

Given the high concordance rate of ER, PR, and HER2 status between core biopsies and resections (>90%), most treatment-naïve breast cancers are not retested at excision.49  In the NCT setting, however, clinically actionable alterations in ER and HER2 have been reported in 3% to 18% and 1% to 7% of residual tumors, respectively.5052  Reasons implicated in discrepant biomarker status between pretreatment and posttreatment tumors include preanalytic factors (eg, cold ischemia time), intratumoral heterogeneity, unsampled tumor, and response to targeted therapy, that is, conversion of HER2+ tumor to HER2 status following HER2-targeted therapies (Figure 13).

Figure 13

Schematic diagram demonstrates various reasons for discrepant biomarkers between pretreatment and posttreatment tumor specimens.

Figure 13

Schematic diagram demonstrates various reasons for discrepant biomarkers between pretreatment and posttreatment tumor specimens.

Close modal

Current practice data indicate considerable variation in retesting tumor biomarkers after neoadjuvant therapy, with only 65% of surveyed academic pathology practices in the United States reporting routine biomarker retesting.53  Repeat testing of HER2-positive carcinomas is particularly controversial because of the potential impact on patient management with HER2-targeted therapies. Although the current College of American Pathologists (CAP) guidelines for ER/PR testing54  and HER2 testing55  in breast cancer do not specifically address this issue, other groups such as the Breast International Group and North American Breast Cancer Group have released recommendations for biomarker retesting in the post-NCT setting.56  For routine clinical practice, we recommend that retesting of ER, PR, and HER2 be considered based on the pretreatment biopsy testing results, pathologic features of the residual tumor, and whether repeat testing will result in a change in treatment. Example scenarios for consideration of repeat testing include residual tumor(s) with intratumoral heterogeneity or lack of response to targeted therapy; pretreatment tumor biopsy with negative or equivocal biomarker results; heterogeneous, unusual, or unanticipated results; and undersampled or marginally sufficient sample of pretreatment tumor (Figure 14).

Figure 14

Recommendations for retesting of tumor markers on residual carcinoma after systemic chemotherapy.

Figure 14

Recommendations for retesting of tumor markers on residual carcinoma after systemic chemotherapy.

Close modal

Proliferation Index (Ki-67)

IHC assessment of Ki-67 expression is a validated surrogate of proliferation in many tumor types, including breast cancer, and has both prognostic and predictive value. Although intralaboratory reproducibility has been shown to be high,57,58  concerns about low interlaboratory reproducibility and lack of standard protocol on testing and scoring have prevented universal adoption of Ki-67 testing in breast cancer. Nevertheless, many laboratories have incorporated Ki-67 testing as part of their routine tumor profile testing.

Posttreatment Ki-67 index has been shown to be of prognostic value for both neoadjuvant endocrine therapy and chemotherapy in ER+/HER2 disease.59  In the NCT setting, change in the Ki-67 index after treatment can be useful to measure partial responses and for consideration of additional adjuvant therapy.60  However, not all laboratories currently perform Ki-67 testing of breast cancers before or after NCT. Nonetheless, some clinical trials use centralized Ki-67 testing, including a recently FDA-approved Ki-67 assay as a companion diagnostic test for ER+/HER2 breast cancer treated with abemaciclib.61  While there remains a need for standardization of Ki-67 testing and scoring, pathologists should take the initiative to optimize the assay in their own laboratory or use an automated system for routine clinical use.

Tumor-Infiltrating Lymphocytes and Immune-Related Biomarkers

Immune profiling has rapidly expanded prognostic and predictive immune-related biomarkers within the tumoral microenvironment. Tumor-infiltrating lymphocytes (TILs) have been shown to be a reproducible biomarker affecting the prognosis and response to treatment in breast cancer.6268  In the era of targeted immunotherapy (eg, pembrolizumab for PD-L1+ TNBC),8,69  there is an increasing clinical demand for assessment of TILs and immune-related biomarkers in pretreatment biopsy and posttreatment residual tumors. In a study, the combination of RCB and TILs was a better predictor of outcome than TILs alone.70  For example, TILs were strongly prognostic in TNBC only in the RCB-II class.71  The International Immuno-Oncology Biomarker Working Group on Breast Cancer has issued a guideline for assessing TILs in the residual carcinoma after NCT.72  However, evaluating TILs in breast cancer remains challenging, particularly in pretreatment core biopsies, because of tumor heterogeneity and concerns about interobserver reproducibility.73  Currently, quantitative assessment of TILs in pretreatment tumor biopsy or posttreatment residual tumor is not a standard practice among pathologists.

Multiple methods have been proposed to assess the degree of response to NCT to provide prognostic information, all of which require some degree of residual tumor quantification. Some classification methods, such as those proposed by the National Surgical Adjuvant Breast and Bowel Project B-18,4  Penault-Llorca et al,27  Chevallier et al,28  and Pinder et al,29  use broad categories such as complete (or near-complete), partial, or no response. These methods may29  or may not4,27,28  include lymph node status in the assessment. Although the prognosis of patients with tumors achieving complete or near-complete pathologic response is clearly better than that of nonresponders, it is important to identify assessment methods that can further stratify the prognosis of patients with varying degrees of partial response (Table 1). Methods proposed by Sataloff et al,32  Smith et al,33  and Bonadonna et al35  have added additional degrees of response based on the percentage of residual tumor cells. To correct for variation in the cellularity of different pretreatment tumors, the Miller-Payne system31  proposes comparison of the posttreatment and pretreatment tumor cellularity, rather than posttreatment cellularity assessment alone. Although this controls for tumor heterogeneity, it does not include lymph node involvement as a prognostic factor in the assessment.

Table 1

Classification Systems That Included Both Breast and Lymph Nodes to Categorize Treatment Responses

Classification Systems That Included Both Breast and Lymph Nodes to Categorize Treatment Responses
Classification Systems That Included Both Breast and Lymph Nodes to Categorize Treatment Responses

The Residual Disease in Breast and Nodes classification system proposed by Chollet et al34  is one of the most encompassing formulas, including the size of the residual tumor, tumor grade, and nodal involvement to place patients into 4 risk categories. Although most available post-NCT assessment methods are similar to one another and provide prognostic value, RCB is the only Web-based formula to include measurements of tumor in both the breast and lymph nodes with the inclusion of tumor cellularity in the breast to quantify response to NCT as a continuous variable.30  The distributions of RCB have been compared in the randomized treatment arms from clinical trials to evaluate overall treatment efficacy.74  Additionally, external validation of the RCB system to demonstrate generalizable prognostic performance within each subtype of breast cancer after NCT is underway.

RCB System

The RCB system is a method to quantify the extent of residual disease in the resection specimen of patients who received NCT.30  It measures residual disease as a continuous variable, combining the input from the primary tumor bed and axillary lymph nodes. The discussions below are meant to clarify some of the questions that were shared by the working group from the feedback obtained from various pathologists who are asked to use the RCB system or are familiar with the reporting of the RCB system.

For the cross-sectional dimensions of the primary tumor bed, the principle is to provide measurements of the 2 largest dimensions of the tumor bed occupied by residual invasive cancer, irrespective of the cellularity or the distribution of the invasive tumor cells within that area. These dimensions are determined by correlating gross and microscopic examination of the specimen, with microscopic determination of the tumor bed used if they are discordant. Hence, there is considerable value in mapping the specimen indicating where the sections were taken (Figure 4). For patients who have multifocal or multicentric tumors (similar in morphology or in tumor profile), measurements of the largest residual tumor should be used to report RCB. In instances where multiple tumors with different morphology and/or tumor profiles are identified, one should calculate RCB separately for each one.

The overall cancer cellularity in the RCB formula refers to the proportion of cancer in the tumor bed area (as percentage residual cancer cellularity). If the tumor bed spans several tissue blocks, cellularity is estimated for each block in order to estimate the average cellularity for the entire residual tumor area. LVI is interpreted as residual invasive cancer in the RCB system.

Cellularity assessments are typically identified to the nearest 10%, with additional selections of 1% and 5% for low cellularity. But one can enter any value, and for tumors with very low cellularity scattered intermittently over a large area, that value may be less than 1%. The usual misunderstanding is to make estimates only in foci of the tumor bed that contain significant residual tumor. The estimates should represent the average across the entire area of residual invasive cancer, not the most concentrated foci.

Challenges With the Current RCB System and Suggested Revisions

One of the challenges in the RCB system is that the calculator requires 2 levels of information (the percentage of the area that is invasive cancer, and the percentage of the cancer that is in situ) to calculate the percentage of the area that is invasive cancer. Many pathologists find this breakdown to be redundant and difficult to ascertain, as they are able to confidently determine in one visual step the proportion of invasive component in an area. Additionally, pathologists are not clear on when not to include the in situ component in the calculation (for example, cases where residual DCIS extends beyond invasive carcinoma and classic lobular carcinoma in situ). Because the inclusion of residual DCIS does not affect the final RCB class or index, the classification of pCR, and, importantly, the outcome of patients, the working group recommends simplifying the RCB calculator to minimize errors (Table 2).

Table 2

Proposed Changes to the Current Residual Cancer Burden (RCB) Calculator

Proposed Changes to the Current Residual Cancer Burden (RCB) Calculator
Proposed Changes to the Current Residual Cancer Burden (RCB) Calculator

Current AJCC Staging System for Reporting of Treated Tumors

First introduced in 1959, the AJCC staging system, currently in its 8th edition, defines the anatomic TNM staging classification based on primary tumor (T), regional lymph nodes (N), and distant metastases (M).36,75  In the 8th edition, the AJCC, for the first time, further defined prognostic stage groups by integrating anatomic stage data with other prognostic factors such as histologic grade, and ER, PR, and HER2 status for breast carcinoma. The 8th edition staging manual establishes postneoadjuvant therapy clinical ycTNM classification (defined by clinical and radiographic findings) and pathologic ypTNM classification (determined by microscopic examination of the primary tumor, any lymph nodes, and any metastatic foci).75,76 

Multiple prospective clinical trials have shown that a pCR is associated with significantly improved survival. Presence of residual in situ carcinoma after NCT in the absence of invasive cancer (classified as ypTis) constitutes pCR by AJCC. Clinical trials using this definition have shown no difference in overall survival whether residual DCIS is present or not in the tumor bed.77  When reporting pCR, a grossly identifiable tumor bed confirmed by microscopic examination is sufficient to document pCR. However, the current AJCC staging manual does not provide clear guidance on the pT staging of non-pCR cases. Below we discuss some of the common issues in the current AJCC staging of treated tumors and suggest modifications to consider for the 9th edition to improve clarity and consistency in reporting residual carcinoma after NCT.

Challenges With the Current AJCC Staging and Suggested Revisions

Most non-pCR cases have residual invasive carcinoma interspersed by uneven areas of fibrous stroma without tumor cells. Per AJCC staging guidelines, posttherapy tumor size is based on the largest contiguous focus with a modifier “m” if multiple foci are present. We suggest that the modifier m be reserved for clearly multifocal and multicentric tumors (similar to untreated tumors); for example, the modifier m should be reserved when residual tumor is present in each pretreated tumor.

The definition of the pT category is derived from the largest diameter of contiguous invasive cancer by the AJCC system. Although this is certainly reasonable for measuring untreated carcinomas, there are considerable challenges when this approach is applied to variably distributed residual tumors with uneven cell density after NCT. There is subjectivity regarding how strictly to apply the term contiguous, but pathologists should probably be lenient in measuring the largest tumor dimension by including small patches of fibrosis between tumor nests, in addition to describing the total volume of tumor, that is, the number of tumor foci in the tumor bed, as suggested by the AJCC staging manual.75 

The AJCC 8th edition affirms that the presence of lymphatic or vascular tumor emboli without residual invasive carcinoma in the breast precludes posttreatment classification as a pCR.75  However, AJCC has not designated any specific ypT classification for this setting. These cases could be classified as ypT(LVI), with a note describing the extent of the lymphatic-vascular channel involvement.

The AJCC ypN staging uses the same categories used for untreated tumors (pN), and is based on the largest contiguous focus of residual tumor in the lymph node (fibrosis between metastatic tumor foci is not included). This method, similar to the T categorization, may underestimate the residual tumor burden in lymph nodes. The method for measuring residual node metastasis should be similar to measuring primary tumor diameter.

Currently, ITCs are not included in the positive lymph node count. Scant or isolated residual tumor cells (less than 0.2 mm or less than 200 cells) in the lymph nodes, whether associated with treatment effect (fibrosis) or not, are currently staged as ypN0(i+), which may be mistaken as a complete nodal response. The presence of ITCs in the NCT setting may not have the same clinical significance compared with untreated tumors.78,79  Therefore, in order to avoid understaging and undertreatment, nodes with ITCs should not be classified as ypN0 (i+) but as ypN1(i+).

Currently, reporting of extranodal extension is not mandatory in the AJCC system; however, a standardized reporting of extranodal extension should be a part of risk stratification for both adjuvant and neoadjuvant cases, as extranodal extension may be a marker of poor prognosis.8082 

Table 3 lists the elements that should be included in the pathology report of resected specimens of treated breast carcinomas. It is mandatory for pathologists to use a synoptic format to report breast carcinomas, both untreated and treated, as recommended by the CAP. In Table 4, we have included the portion of the current CAP protocol83  that is applicable for the reporting of treated breast cancer specimens and proposes revisions to a future version for better categorization of treatment responses. In addition, we suggest that the CAP breast cancer protocol should include a Web-based classification system such as RCB for a standardized assessment of tumor burden, hence allowing comparison of the predictive value afforded by different systems.

Table 3

Elements to Be Included in the Pathology Report of Treated Breast Carcinomas

Elements to Be Included in the Pathology Report of Treated Breast Carcinomas
Elements to Be Included in the Pathology Report of Treated Breast Carcinomas
Table 4

Proposed Revision to the College of American Pathologists (CAP) Breast Cancer Checklist

Proposed Revision to the College of American Pathologists (CAP) Breast Cancer Checklist
Proposed Revision to the College of American Pathologists (CAP) Breast Cancer Checklist

One of the future steps that has been tried is the evaluation of treatment response by image-guided core biopsy during NCT to determine whether a specific chemotherapy regimen is effective.84  This approach would also allow medical oncologists to modify the therapeutic regimen for nonresponders while informing a response-based decision for surgical treatment. However, the timing of the biopsy will need to be determined by future studies and may vary for different tumor types and treatment regimens.

Currently, a standardized post-NCT breast carcinoma reporting system is not available. The goal should be to maximize the clinical utility of the reported information while keeping the process as efficient as possible, thereby allowing for generalized adoption of the system. This approach would also provide extensive population data available for further studies in a relatively short time frame. To achieve this goal, every pathology practice should implement a standard operating procedure for collecting, handling, and interpreting pathology specimens treated with NST. All pathologists participating in neoadjuvant trials should receive formal training as recommended by the FDA guidance document.6 

Finally, artificial intelligence has recently shown promising utility in applications in medicine and pathology. Incorporating machine learning to evaluate treated breast cancers to standardize reporting and search for prognostic and predictive factors will be an important next step in breast cancer pathology.

In summary, a pressing need is to develop a postneoadjuvant synoptic report format relevant to reporting residual disease after NST. As discussed earlier, the current CAP synoptic report for invasive breast cancer and the RCB system require modifications to address the specific challenges posed by neoadjuvant treatment. To refine a patient's prognostic risk, any future modifications to predictive models, whether the AJCC system or RCB calculator, should be based on pretreatment tumor characteristics and patient outcomes based on the posttreatment response category.

Context of Research

The I-SPY 2 Trial Pathology Working Group comprises pathologists working across national trial sites. I-SPY 2 is a multicenter, adaptive randomization phase 2 neoadjuvant platform trial for early-stage, high-risk breast cancer patients. A subcommittee of the I-SPY 2 Pathology Working Group was formed by a group of pathologists with extensive experience in examining and reporting on breast and lymph node specimens after NCT. This subcommittee met virtually (once a month for a year and quarterly thereafter) to compile and compare reporting practices across sites and discuss the issues and controversies related to therapy before providing recommendations.

1.
Mougalian
SS
,
Soulos
PR
,
Killelea
BK
, et al.
Use of neoadjuvant chemotherapy for patients with stage I to III breast cancer in the United States
.
Cancer
.
2015
;
121
(
15
):
2544
2552
.
2.
Vugts
G
,
Maaskant-Braat
AJ
,
Nieuwenhuijzen
GA
,
Roumen
RM
,
Luiten
EJ
,
Voogd
AC
.
Patterns of care in the administration of neo-adjuvant chemotherapy for breast cancer: a population-based study
.
Breast J
.
2016
;
22
(
3
):
316
321
.
3.
Early Breast Cancer Trialists' Collaborative Group
.
Long-term outcomes for neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of individual patient data from ten randomised trials
.
Lancet Oncol
.
2018
;
19
(
1
):
27
39
.
4.
Fisher
ER
,
Wang
J
,
Bryant
J
,
Fisher
B
,
Mamounas
E
,
Wolmark
N.
Pathobiology of preoperative chemotherapy: findings from the National Surgical Adjuvant Breast and Bowel (NSABP) protocol B-18
.
Cancer
.
2002
;
95
(
4
):
681
695
.
5.
Haque
W
,
Verma
V
,
Hatch
S
,
Klimberg
VS
,
Butler
EB
,
Teh
BS
.
Response rates and pathologic complete response by breast cancer molecular subtype following neoadjuvant chemotherapy
.
Breast Cancer Res Treat
.
2018
;
170
(
3
):
559
567
.
6.
United States Food and Drug Administration
.
Pathologic complete response in neoadjuvant treatment of high-risk early-stage breast cancer: use as an endpoint to support accelerated approval.
https://www.fda.gov/regulatory-information/search-fda-guidance-documents/pathologic-complete-response-neoadjuvant-treatment-high-risk-early-stage-breast-cancer-use-endpoint. Published
2014
.
Updated August 24, 2018. Accessed December 18, 2019.
7.
Cortazar
P
,
Zhang
L
,
Untch
M
, et al.
Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis
.
Lancet
.
2014
;
384
(
9938
):
164
172
.
8.
Schmid
P
,
Cortes
J
,
Pusztai
L
, et al.
Pembrolizumab for early triple-negative breast cancer
.
N Engl J Med
.
2020
;
382
(
9
):
810
821
.
9.
Loibl
S
,
O'Shaughnessy
J
,
Untch
M
, et al.
Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): a randomised, phase 3 trial
.
Lancet Oncol
.
2018
;
19
(
4
):
497
509
.
10.
Gianni
L
,
Baselga
J
,
Eiermann
W
, et al.
Phase III trial evaluating the addition of paclitaxel to doxorubicin followed by cyclophosphamide, methotrexate, and fluorouracil, as adjuvant or primary systemic therapy: European Cooperative Trial in Operable Breast Cancer
.
J Clin Oncol
.
2009
;
27
(
15
):
2474
2481
.
11.
Mauriac
L
,
MacGrogan
G
,
Avril
A
, et al.
Neoadjuvant chemotherapy for operable breast carcinoma larger than 3 cm: a unicentre randomized trial with a 124-month median follow-up. Institut Bergonie Bordeaux Groupe Sein (IBBGS)
.
Ann Oncol
.
1999
;
10
(
1
):
47
52
.
12.
van der Hage
JA
,
van de Velde
CJ
,
Julien
JP
,
Tubiana-Hulin
M
,
Vandervelden
C
,
Duchateau
L.
Preoperative chemotherapy in primary operable breast cancer: results from the European Organization for Research and Treatment of Cancer trial 10902
.
J Clin Oncol
.
2001
;
19
(
22
):
4224
4237
.
13.
Wolmark
N
,
Wang
J
,
Mamounas
E
,
Bryant
J
,
Fisher
B.
Preoperative chemotherapy in patients with operable breast cancer: nine-year results from National Surgical Adjuvant Breast and Bowel Project B-18
.
J Natl Cancer Inst Monogr
.
2001
(30
):
96
102
.
14.
Mauri
D
,
Pavlidis
N
,
Ioannidis
JP
.
Neoadjuvant versus adjuvant systemic treatment in breast cancer: a meta-analysis
.
J Natl Cancer Inst
.
2005
;
97
(
3
):
188
194
.
15.
Scholl
SM
,
Asselain
B
,
Palangie
T
, et al.
Neoadjuvant chemotherapy in operable breast cancer
.
Eur J Cancer
.
1991
;
27
(
12
):
1668
1671
.
16.
Fisher
B
,
Bryant
J
,
Wolmark
N
, et al.
Effect of preoperative chemotherapy on the outcome of women with operable breast cancer
.
J Clin Oncol
.
1998
;
16
(
8
):
2672
2685
.
17.
Rastogi
P
,
Anderson
SJ
,
Bear
HD
, et al.
Preoperative chemotherapy: updates of National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27
.
J Clin Oncol
.
2008
;
26
(
5
):
778
785
.
18.
Powles
TJ
,
Hickish
TF
,
Makris
A
, et al.
Randomized trial of chemoendocrine therapy started before or after surgery for treatment of primary breast cancer
.
J Clin Oncol
.
1995
;
13
(
3
):
547
552
.
19.
Mieog
JS
,
van der Hage
JA
,
van de Velde
CJ
.
Neoadjuvant chemotherapy for operable breast cancer
.
Br J Surg
.
2007
;
94
(
10
):
1189
1200
.
20.
Symmans
WF
,
Wei
C
,
Gould
R
, et al.
Long-term prognostic risk after neoadjuvant chemotherapy associated with residual cancer burden and breast cancer subtype
.
J Clin Oncol
.
2017
;
35
(
10
):
1049
1060
.
21.
Kuerer
HM
,
Newman
LA
,
Smith
TL
, et al.
Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy
.
J Clin Oncol
.
1999
;
17
(
2
):
460
469
.
22.
von Minckwitz
G
,
Untch
M
,
Blohmer
JU
, et al.
Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes
.
J Clin Oncol
.
2012
;
30
(
15
):
1796
1804
.
23.
von Minckwitz
G
,
Huang
CS
,
Mano
MS
, et al.
Trastuzumab emtansine for residual invasive HER2-positive breast cancer
.
N Engl J Med
.
2019
;
380
(
7
):
617
628
.
24.
Masuda
N
,
Lee
SJ
,
Ohtani
S
, et al.
Adjuvant capecitabine for breast cancer after preoperative chemotherapy
.
N Engl J Med
.
2017
;
376
(
22
):
2147
2159
.
25.
Johnston
SRD
,
Harbeck
N
,
Hegg
R
, et al.
Abemaciclib combined with endocrine therapy for the adjuvant treatment of HR+, HER2, node-positive, high-risk, early breast cancer (monarchE)
.
J Clin Oncol
.
2020
;
38
(
34
):
3987
3998
.
26.
Selli
C
,
Sims
AH
.
Neoadjuvant therapy for breast cancer as a model for translational research
.
Breast Cancer (Auckl)
.
2019
;
13
:
1178223419829072
.
27.
Penault-Llorca
F
,
Abrial
C
,
Raoelfils
I
, et al.
Comparison of the prognostic significance of Chevallier and Sataloff's pathologic classifications after neoadjuvant chemotherapy of operable breast cancer
.
Hum Pathol
.
2008
;
39
(
8
):
1221
1228
.
28.
Chevallier
B
,
Roche
H
,
Olivier
JP
,
Chollet
P
,
Hurteloup
P.
Inflammatory breast cancer: pilot study of intensive induction chemotherapy (FEC-HD) results in a high histologic response rate
.
Am J Clin Oncol
.
1993
;
16
(
3
):
223
228
.
29.
Pinder
SE
,
Provenzano
E
,
Earl
H
,
Ellis
IO
.
Laboratory handling and histology reporting of breast specimens from patients who have received neoadjuvant chemotherapy
.
Histopathology
.
2007
;
50
(
4
):
409
417
.
30.
Symmans
WF
,
Peintinger
F
,
Hatzis
C
, et al.
Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy
.
J Clin Oncol
.
2007
;
25
(
28
):
4414
4422
.
31.
Ogston
KN
,
Miller
ID
,
Payne
S
, et al.
A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival
.
Breast
.
2003
;
12
(
5
):
320
327
.
32.
Sataloff
DM
,
Mason
BA
,
Prestipino
AJ
,
Seinige
UL
,
Lieber
CP
,
Baloch
Z.
Pathologic response to induction chemotherapy in locally advanced carcinoma of the breast: a determinant of outcome
.
J Am Coll Surg
.
1995
;
180
(
3
):
297
306
.
33.
Smith
IC
,
Heys
SD
,
Hutcheon
AW
, et al.
Neoadjuvant chemotherapy in breast cancer: significantly enhanced response with docetaxel
.
J Clin Oncol
.
2002
;
20
(
6
):
1456
1466
.
34.
Chollet
P
,
Abrial
C
,
Durando
X
, et al.
A new prognostic classification after primary chemotherapy for breast cancer: residual disease in breast and nodes (RDBN)
.
Cancer J
.
2008
;
14
(
2
):
128
132
.
35.
Bonadonna
G
,
Veronesi
U
,
Brambilla
C
, et al.
Primary chemotherapy to avoid mastectomy in tumors with diameters of three centimeters or more
.
J Natl Cancer Inst
.
1990
;
82
(
19
):
1539
1545
.
36.
Hortobagyi
GN
,
Connoly
JL
,
D'Orsi
CJ
, et al.
Breast
.
In:
AJCC Cancer Staging Manual, 8th ed
.
Cham, Switzerland
:
Springer;
2017
:
589
636
.
37.
Sahoo
S
,
Lester
SC
.
Pathology considerations in patients treated with neoadjuvant chemotherapy
.
Surg Pathol Clin
.
2012
;
5
(
3
):
749
774
.
38.
Mittendorf
EA
,
Vila
J
,
Tucker
SL
, et al.
The Neo-Bioscore update for staging breast cancer treated with neoadjuvant chemotherapy: incorporation of prognostic biologic factors into staging after treatment
.
JAMA Oncol
.
2016
;
2
(
7
):
929
936
.
39.
Han
R
,
Regpala
S
,
Slodkowska
E
, et al.
Lack of standardization in the processing and reporting of post-neoadjuvant breast cancer specimens
.
Arch Pathol Lab Med
.
2020
;
144
(
10
):
1262
1270
.
40.
Sharkey
FE
,
Addington
SL
,
Fowler
LJ
,
Page
CP
,
Cruz
AB
.
Effects of preoperative chemotherapy on the morphology of resectable breast carcinoma
.
Mod Pathol
.
1996
;
9
(
9
):
893
900
.
41.
Pastorello
RG
,
Laws
A
,
Grossmith
S
, et al.
Clinico-pathologic predictors of patterns of residual disease following neoadjuvant chemotherapy for breast cancer
.
Mod Pathol
.
2021
;
34
(
5
):
875
882
.
42.
Donnelly
J
,
Parham
DM
,
Hickish
T
,
Chan
HY
,
Skene
AI
.
Axillary lymph node scarring and the association with tumour response following neoadjuvant chemoendocrine therapy for breast cancer
.
Breast
.
2001
;
10
(
1
):
61
66
.
43.
Czaja
R
,
Wu
R
,
Jorns
JM
.
Breast sentinel lymph node frozen section practice: an enterprise audit as a guide for moving forward
.
Arch Pathol Lab Med
.
2021
;
145
(
8
):
1018
1024
.
44.
Samiei
S
,
van Nijnatten
TJA
,
de Munck
L
, et al.
Correlation between pathologic complete response in the breast and absence of axillary lymph node metastases after neoadjuvant systemic therapy
.
Ann Surg
.
2020
;
271
(
3
):
574
580
.
45.
Tee
SR
,
Devane
LA
,
Evoy
D
, et al.
Meta-analysis of sentinel lymph node biopsy after neoadjuvant chemotherapy in patients with initial biopsy-proven node-positive breast cancer
.
Br J Surg
.
2018
;
105
(
12
):
1541
1552
.
46.
Caudle
AS
,
Yang
WT
,
Krishnamurthy
S
, et al.
Improved axillary evaluation following neoadjuvant therapy for patients with node-positive breast cancer using selective evaluation of clipped nodes: implementation of targeted axillary dissection
.
J Clin Oncol
.
2016
;
34
(
10
):
1072
1078
.
47.
Mamtani
A
,
Barrio
A
,
King
TA
, et al.
How often does neoadjuvant chemotherapy avoid axillary dissection in patients with histologically confirmed nodal metastases?: results of a prospective study
.
Ann Surg Oncol
.
2016
;
23
:
3467
3474
.
48.
Gimbergues
P
,
Dauplat
MM
,
Durando
X
, et al.
Intraoperative imprint cytology examination of sentinel lymph nodes after neoadjuvant chemotherapy in breast cancer patients
.
Ann Surg Oncol
.
2010
;
17
(
8
):
2132
2137
.
49.
Dekker
TJ
,
Smit
VT
,
Hooijer
GK
, et al.
Reliability of core needle biopsy for determining ER and HER2 status in breast cancer
.
Ann Oncol
.
2013
;
24
(
4
):
931
937
.
50.
Xian
Z
,
Quinones
AK
,
Tozbikian
G
,
Zynger
DL
.
Breast cancer biomarkers before and after neoadjuvant chemotherapy: does repeat testing impact therapeutic management?
Hum Pathol
.
2017
;
62
:
215
221
.
51.
Gahlaut
R
,
Bennett
A
,
Fatayer
H
, et al.
Effect of neoadjuvant chemotherapy on breast cancer phenotype, ER/PR and HER2 expression—implications for the practising oncologist
.
Eur J Cancer
.
2016
;
60
:
40
48
.
52.
Zhang
N
,
Moran
MS
,
Huo
Q
,
Haffty
BG
,
Yang
Q.
The hormonal receptor status in breast cancer can be altered by neoadjuvant chemotherapy: a meta-analysis
.
Cancer Invest
.
2011
;
29
(
9
):
594
598
.
53.
Lanjewar
S
,
Patil
P
,
Fineberg
S.
Pathologic reporting practices for breast cancer specimens after neoadjuvant chemotherapy—a survey of pathologists in academic institutions across the United States
.
Mod Pathol
.
2020
;
33
(
1
):
91
98
.
54.
Allison
KH
,
Hammond
MEH
,
Dowsett
M
, et al.
Estrogen and progesterone receptor testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists guideline update
.
Arch Pathol Lab Med
.
2020
;
144
(
5
):
545
563
.
55.
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
.
J Clin Oncol
.
2018
;
36
(
20
):
2105
2122
.
56.
Provenzano
E
,
Bossuyt
V
,
Viale
G
, et al.
Standardization of pathologic evaluation and reporting of postneoadjuvant specimens in clinical trials of breast cancer: recommendations from an international working group
.
Mod Pathol
.
2015
;
28
(
9
):
1185
1201
.
57.
Polley
MY
,
Leung
SC
,
McShane
LM
, et al.
An international Ki67 reproducibility study
.
J Natl Cancer Inst
.
2013
;
105
(
24
):
1897
1906
.
58.
Focke
CM
,
Burger
H
,
van Diest
PJ
, et al.
Interlaboratory variability of Ki67 staining in breast cancer
.
Eur J Cancer
.
2017
;
84
:
219
227
.
59.
Ellis
MJ
,
Suman
VJ
,
Hoog
J
, et al.
Ki67 proliferation index as a tool for chemotherapy decisions during and after neoadjuvant aromatase inhibitor treatment of breast cancer: results from the American College of Surgeons oncology group Z1031 trial (alliance)
.
J Clin Oncol
.
2017
;
35
(
10
):
1061
1069
.
60.
Cabrera-Galeana
P
,
Munoz-Montano
W
,
Lara-Medina
F
, et al.
Ki67 changes identify worse outcomes in residual breast cancer tumors after neoadjuvant chemotherapy
.
Oncologist
.
2018
;
23
(
6
):
670
678
.
61.
Royce
M
,
Osgood
C
,
Mulkey
F
, et al.
FDA approval summary: abemaciclib with endocrine therapy for high-risk early breast cancer. J Clin Oncol. 2022:JCO2102742.
62.
Dieci
MV
,
Criscitiello
C
,
Goubar
A
, et al.
Prognostic value of tumor-infiltrating lymphocytes on residual disease after primary chemotherapy for triple-negative breast cancer: a retrospective multicenter study
.
Ann Oncol
.
2014
;
25
(
3
):
611
618
.
63.
Loi
S
,
Dushyanthen
S
,
Beavis
PA
, et al.
RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors
.
Clin Cancer Res
.
2016
;
22
(
6
):
1499
1509
.
64.
Hamy
AS
,
Pierga
JY
,
Sabaila
A
, et al.
Stromal lymphocyte infiltration after neoadjuvant chemotherapy is associated with aggressive residual disease and lower disease-free survival in HER2-positive breast cancer
.
Ann Oncol
.
2017
;
28
(
9
):
2233
2240
.
65.
Pelekanou
V
,
Barlow
WE
,
Nahleh
ZA
, et al.
Tumor-infiltrating lymphocytes and PD-L1 expression in pre- and posttreatment breast cancers in the SWOG S0800 phase II neoadjuvant chemotherapy trial
.
Mol Cancer Ther
.
2018
;
17
(
6
):
1324
1331
.
66.
Li
X
,
Warren
S
,
Pelekanou
V
, et al.
Immune profiling of pre- and post-treatment breast cancer tissues from the SWOG S0800 neoadjuvant trial
.
J Immunother Cancer
.
2019
;
7
(
1
):
88
.
67.
Pelekanou
V
,
Carvajal-Hausdorf
DE
,
Altan
M
, et al.
Effect of neoadjuvant chemotherapy on tumor-infiltrating lymphocytes and PD-L1 expression in breast cancer and its clinical significance
.
Breast Cancer Res
.
2017
;
19
(
1
):
91
.
68.
Dieci
MV
,
Miglietta
F
,
Guarneri
V.
Immune infiltrates in breast cancer: recent updates and clinical implications
.
Cells
.
2021
;
10
(
2
):
223
.
69.
Cortes
J
,
Cescon
DW
,
Rugo
HS
, et al.
Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (KEYNOTE-355): a randomised, placebo-controlled, double-blind, phase 3 clinical trial
.
Lancet
.
2020
;
396
(
10265
):
1817
1828
.
70.
Asano
Y
,
Kashiwagi
S
,
Goto
W
, et al.
Prediction of survival after neoadjuvant chemotherapy for breast cancer by evaluation of tumor-infiltrating lymphocytes and residual cancer burden
.
BMC Cancer
.
2017
;
17
(
1
):
888
.
71.
Luen
SJ
,
Salgado
R
,
Dieci
MV
, et al.
Prognostic implications of residual disease tumor-infiltrating lymphocytes and residual cancer burden in triple-negative breast cancer patients after neoadjuvant chemotherapy
.
Ann Oncol
.
2019
;
30
(
2
):
236
242
.
72.
Dieci
MV
,
Radosevic-Robin
N
,
Fineberg
S
, et al.
Update on tumor-infiltrating lymphocytes (TILs) in breast cancer, including recommendations to assess TILs in residual disease after neoadjuvant therapy and in carcinoma in situ: a report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer
.
Semin Cancer Biol
.
2018
;
52
(
pt 2
):
16
25
.
73.
Khoury
T
,
Peng
X
,
Yan
L
,
Wang
D
,
Nagrale
V.
Tumor-infiltrating lymphocytes in breast cancer: evaluating interobserver variability, heterogeneity, and fidelity of scoring core biopsies
.
Am J Clin Pathol
.
2018
;
150
(
5
):
441
450
.
74.
Symmans
WF
,
Yau
C
,
Chen
YY
, et al.
Assessment of residual cancer burden and event-free survival in neoadjuvant treatment for high-risk breast cancer: an analysis of data from the I-SPY2 randomized clinical trial
.
JAMA Oncol
.
2021
;
7
(
11
):
1654
1663
.
75.
Amin
MB
,
Edge
SB
,
Greene
FL
, et al.
AJCC Cancer Staging Manual.
Springer International Publishing;
2016
.
76.
Giuliano
AE
,
Connolly
JL
,
Edge
SB
, et al.
Breast cancer—major changes in the American Joint Committee on Cancer eighth edition cancer staging manual
.
CA Cancer J Clin
.
2017
;
67
(
4
):
290
303
.
77.
Mazouni
C
,
Peintinger
F
,
Wan-Kau
S
, et al.
Residual ductal carcinoma in situ in patients with complete eradication of invasive breast cancer after neoadjuvant chemotherapy does not adversely affect patient outcome
.
J Clin Oncol
.
2007
;
25
(
19
):
2650
2655
.
78.
Rubio
IT
.
Sentinel lymph node metastasis after neoadjuvant treatment in breast cancer: any size matters?
World J Clin Oncol
.
2015
;
6
(
6
):
202
206
.
79.
Boileau
JF
,
Poirier
B
,
Basik
M
, et al.
Sentinel node biopsy after neoadjuvant chemotherapy in biopsy-proven node-positive breast cancer: the SN FNAC study
.
J Clin Oncol
.
2015
;
33
(
3
):
258
264
.
80.
Nottegar
A
,
Veronese
N
,
Senthil
M
, et al.
Extra-nodal extension of sentinel lymph node metastasis is a marker of poor prognosis in breast cancer patients: a systematic review and an exploratory meta-analysis
.
Eur J Surg Oncol
.
2016
;
42
(
7
):
919
925
.
81.
Choi
AH
,
Blount
S
,
Perez
MN
, et al.
Size of extranodal extension on sentinel lymph node dissection in the American College of Surgeons Oncology group Z0011 trial era
.
JAMA Surg
.
2015
;
150
(
12
):
1141
1148
.
82.
Barrio
AV
,
Downs-Canner
S
,
Edelweiss
M
, et al.
Microscopic extracapsular extension in sentinel lymph nodes does not mandate axillary dissection in Z0011-eligible patients
.
Ann Surg Oncol
.
2020
;
27
(
5
):
1617
1624
.
83.
College of American Pathologists
.
Protocol for the examination of resection specimens from patients with invasive carcinoma of the breast. Version 4.5.0.1.
https://documents.cap.org/protocols/Breast.Invasive_4.5.0.1.REL_CAPCP.pdf. Published
November
2021
.
Accessed November 11, 2021.
84.
Tasoulis
MK
,
Lee
HB
,
Yang
W
, et al.
Accuracy of post-neoadjuvant chemotherapy image-guided breast biopsy to predict residual cancer
.
JAMA Surg
.
2020
;
155
(
12
):
e204103
.

Author notes

Symmans is a founder, scientific advisor, and shareholder of Delphi Diagnostics and has intellectual property in the company. Symmans has a patent issued for a method to measure residual cancer burden. The other authors have no relevant financial interest in the products or companies described in this article.