You are on page 1of 13

Review

Breast specimen handling and reporting in the post-

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
neoadjuvant setting: challenges and advances
Miralem Mrkonjic,1,2 Hal K Berman,1,2 Susan J Done,1,2 Bruce Youngson,1,2
Anna Marie Mulligan1,2

1
Laboratory Medicine Program, Summary agents in the clinical trials setting.9 The United
University Health Network, Neoadjuvant systemic therapy is becoming more States Food and Drug Administration (FDA) has
Toronto, Ontario, Canada
2
Laboratory Medicine and commonly used in patients with earlier stages of breast recommended complete pathological response as
Pathobiology, University of cancer. To assess tumour response to neoadjuvant an end-point for accelerated approval of new agents
Toronto, Toronto, Ontario, chemotherapy, pathological evaluation is the gold for neoadjuvant treatment of high-risk early stage
Canada standard. Depending on the treatment response, the breast cancer11 12; however, improved event-free
pathological examination of these specimens can be survival still remains the end-point for full FDA
Correspondence to quite challenging. However, a uniform approach to approval.13
Dr Anna Marie Mulligan,
Laboratory Medicine Program, evaluate post-neoadjuvant-treated breast specimens has Response to neoadjuvant systemic therapy is
University Health Network, been lacking. Furthermore, there is no single universally an excellent predictor of outcome,13 and achieve-
Toronto, ON M5G 2C4, Canada; ​ accepted or endorsed classification system for assessing ment of pathological complete response (pCR) is
annamarie.​mulligan@​uhn.​ca treatment response in this setting. Recent initiatives a prognostic predictor of long-term outcome with
have attempted to create a standardised protocol for significantly better disease-free survival and overall
Received 27 November 2018
Accepted 28 November 2018 evaluation of post-neoadjuvant breast specimens. survival.14 15 In addition, poor response to neoad-
This review outlines the necessary information that juvant treatment provides opportunities to patients
should be collected prior to macroscopic examination for supplementary or alternative postsurgical ther-
of these specimens, the recommended and most apies.16 It is important to note that complete clin-
pragmatic approach to tissue sampling for microscopic ical tumour regression does not correlate well with
examination, describes the macroscopic and microscopic complete pathological response. Between 60% and
features of post-therapy breast specimens, summarises 80% of patients considered to have had a complete
two commonly used systems for classifying treatment clinical response have residual microscopic disease,
response and outlines the critical variables that should while 20% of patients with clinically suspected
be included in the final pathology report. residual disease have complete pathological
response.17 Therefore, thorough macroscopic and
microscopic assessment by the pathologist remains
the gold standard for determining complete treat-
Introduction ment response.
Neoadjuvant therapy refers to the treatment of Responses to preoperative systemic therapies are
patients before surgical removal of carcinoma.1 variable and assessment of these specimens pose
In patients with invasive breast carcinoma, this significant challenges to pathologists. Centralised
is usually some combination of chemotherapy, reviews of pathology reports reveal huge varia-
hormone receptor blockade and human epidermal tion in handling and reporting between centres
growth factor-2 (HER2) receptor monoclonal anti- resulting in substantial differences in the patho-
body, but may also include radiation therapy. Tradi- logical complete response rates.18–20 As such,
tionally, neoadjuvant systemic therapy has been there is a need for a standardised and universally
used for either downsizing carcinomas to facilitate accepted protocol for pathological evaluation and
►► http://​​dx.​​doi.​​org/​​10.​​1136/​ breast-conserving surgery or for downsizing previ- reporting. A recent multidisciplinary initiative by
jclinpath-​2018-​205592 ously unresectable locally advanced breast carci- the Breast International Group–North American
►► http://​​dx.​​doi.​​org/​​10.​1136/​ nomas.2 3 Supported by evidence from multiple Breast Cancer Group (BIG-NABCG) aimed to
jclinpath-​2018-​205595 randomised trials showing comparable rates of
►► http://​​dx.​​doi.​​org/​​10.​1136/​
produce standardised guidelines for evaluation and
jclinpath-​2018-​205596 survival or overall disease progression between reporting of post-neoadjuvant breast specimens.13
►► http://​​dx.​​doi.​​org/​​10.​1136/​ neoadjuvant and adjuvant systemic chemotherapy In this review, we will outline the challenges faced
jclinpath-​2018-​205634 regimens, neoadjuvant therapies are increasingly by pathologists evaluating post-neoadjuvant breast
being offered as an alternative to adjuvant therapy, specimens, summarise the most practical guide-
particularly in women with early stage high-risk lines for macroscopic and microscopic evaluation,
breast cancer.4–10 While more frequent local recur- and present the most salient features for adequate
© Author(s) (or their
employer(s)) 2019. No rence rates associated with neoadjuvant chemo- reporting of these specimens.
commercial re-use. See rights therapy in the setting of breast-conserving surgery
and permissions. Published have been reported, this does not impact overall
by BMJ. survival.10 Furthermore, the neoadjuvant setting Prior to gross examination
To cite: Mrkonjic M, provides an opportunity to assess tumour response Detailed clinical and radiological information is
Berman HK, Done SJ, to different systemic chemotherapeutic agents and essential for these specimens as the tumour site(s)
et al. J Clin Pathol provides an ideal setting to study biomarkers of can be difficult to appreciate on macroscopic exam-
2019;72:120–132. responsiveness and resistance to novel therapeutic ination. Several initiatives have recommended
120   Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598
Review

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
Figure 1  Breast MRI with examples of radiological neoadjuvant treatment response. Patient before (A) and after (B) neoadjuvant therapy. Post-
therapy breast MRI demonstrates enhancement around the radiological clip site without evidence of residual carcinoma. Patient before (C) and after
(D) neoadjuvant therapy. Post-therapy breast MRI demonstrates minimal therapeutic response.

that all relevant clinical information accompany the specimen Diagnostic imaging
requisition forms and have provided examples of requisition Pre-treatment imaging should be performed for clinical staging
templates.2 13 Alternatively, review of the documentation in the at the time of diagnosis. A detailed description of the tumour
electronic patient record is required. Multidisciplinary collabo- dimensions and location is required with an o’clock radius and
ration and communication is essential as neoadjuvant therapy distance from the nipple being superior to the quadrant loca-
creates challenges for all involved disciplines.16 tion.13 This applies to all additional tumour foci as well as the
relationships between additional foci to the main tumour mass.
Pathology Clip placement at the time of diagnostic biopsy is strongly
If the image-guided core-needle biopsy was performed at an recommended even if mastectomy is planned.21 Radiological
outside institution, and the slides are not available for review, the clips can subsequently be used to localise the tumour bed(s) in
clinical documents should contain the original pathology report cases with complete radiological response (figure 1). Presence of
or a summary including the following: histological tumour calcifications associated with tumour should be noted as they can
type, preliminary tumour grade, biomarker status and any other be subsequently used to localise the lesion post-therapy.13
parameters used to inform neoadjuvant treatment decisions.13 As the presence of nodal metastasis is a key prognostic indi-
Similarly, original pathology reports for all fine-needle aspi- cator and also plays a major role in treatment planning,22–25 the
rates of radiologically suspicious axillary lymph nodes should axillary lymph nodes should be assessed and the report should
be included. describe the number and dimensions of the most concerning

Figure 2  Spectrum of macroscopic changes observed in multiple post-neoadjuvant breast specimens. (A) Focal haemorrhagic area with no residual
tumour mass or tumour bed, (B) fibrous breast tissue without grossly visible tumour mass, (C) small focus of residual carcinoma within fibrous tumour
bed (black asterisk), (D) well-circumscribed firm solid mass (red circle) and (E) spiculated hard tumour mass.
Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598 121
Review

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
Figure 3  Example of specimen mapping for microscopic evaluation. Lumpectomy specimen was sectioned into four slices, oriented from medial
to lateral. A firm fibrous mass was identified in tissue slices 2, 3 and focally in 4. Black frames indicate tissue sampled: full-face of the largest cross-
sectional area of mass with respective margins and skin in slice 3, additional sections of mass and margins in slice 2, adjacent areas with medial and
lateral margins in tissue slices 1 and 4, respectively. Orientation markers: S, superior; I, inferior; A, anterior; P, posterior.

lymph nodes. The National Comprehensive Cancer Network negative for metastases.26 32 Removal of positive axillary lymph
guidelines recommend pathological confirmation of all clini- node(s) with a sentinel lymph node biopsy before neoadjuvant
cally palpable lymph nodes using ultrasound-guided sampling therapy is discouraged as it prevents the assessment of treatment
methods.26 Axillary ultrasound combined with fine-needle aspi- response in the lymph nodes. On the other hand, post-treatment
ration (FNA) of abnormal lymph nodes have high sensitivity and lymph node assessment is strongly recommended, as the treat-
specificity for detecting lymph node metastases.22 Response to ment response in the lymph nodes is a major prognostic vari-
neoadjuvant therapy in the lymph nodes may be so pronounced able.33 Emerging evidence suggests that, in a neoadjuvant setting,
that any involved lymph node may appear unremarkable on sentinel lymph node biopsy may be an appropriate alternative to
subsequent imaging. Therefore, if an axillary lymph node was axillary node dissection in clinically node-positive patients.34–37
sampled by FNA, clip placement for future localisation of that Previously clipped lymph nodes are identified as sentinel lymph
lymph node is strongly recommended as it improves accuracy of nodes in 75% of cases and intraoperative localisation or patho-
sentinel lymph node biopsy.26 27 logical assessment is necessary to ensure that the clipped lymph
Post-treatment imaging should reflect the clinical response to nodes have been resected.32 Removal of all sentinel lymph nodes
therapy as well as the dimensions of the tumour bed and the rela- and clipped lymph nodes has been found to reduce the false
tionship to the radiological clip. Clinical response to therapy of negative rates.27 Axillary lymph node dissection is recommended
any abnormal axillary lymph node should also be noted. As clips if the clipped node has not been resected or if residual disease is
can migrate post-insertion, their position relative to the tumour identified within the sentinel lymph node.32
mass, tumour bed and positive lymph node should be reported.
Gross examination and sampling of resection
Medical oncology specimens
The type and duration of presurgical chemotherapy should be Lumpectomy and mastectomy specimens
available in the clinical record. Patient participation in clinical Macroscopic examination of neoadjuvant surgical specimens and
trials should be highlighted and noted on the requisition form identification of tumour bed can be particularly challenging for
especially if tissue sampling and processing requires modifica- pathologists. Tumour response to neoadjuvant therapy can be
tions to the standard institutional protocol. very dramatic with residual tumour bed resembling unremark-
able fibrous breast tissue (figure 2). Accompanying calcifications,
Surgery fat necrosis or haemorrhage may be present. Prior needle core
Surgical resection is based on preoperative imaging, and all detect- biopsy reaction site, if present, is useful, to identify the loca-
able residual disease should be excised with clear margins.28–30 tion of the tumour bed. Identification of radiological clips,
The optimal margin width in the neoadjuvant setting is unknown macroscopically or with specimen radiographs, is helpful and
with a recent study demonstrating no difference in disease-free may be essential. Clinical judgement should be used regarding
survival and overall survival for margins <1 mm versus >1 mm.31 tissue collection for research in this setting. Regardless of spec-
For cases with complete radiological response, the tumour bed imen size/type, careful mapping using specimen photography or
with radiological clip(s) should be removed. Any intraoperative diagrams is necessary to allow reconstruction of the specimen
localisation techniques should be noted and careful specimen and to facilitate correlation of macroscopic and microscopic
orientation is imperative and applies to all additional speci- findings (figure 3).18
mens, such as margin revisions. Sentinel lymph node biopsy is Specimen preparation and processing protocols do not differ in
the standard approach for axillary staging in patients with clini- the neoadjuvant setting. Lumpectomy specimens are inked fresh
cally negative lymph nodes22 with recommended resection of at according to the institutional protocols and sliced at 3 mm to 5
least two sentinel lymph nodes.32 Completion of axillary lymph mm intervals to ensure formalin penetration. Small specimens
node dissection is not required if the sentinel lymph nodes are (<5 cm in greatest diameter or <30 g) can be submitted in their
122 Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598
Review

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.

Figure 4  Spectrum of microscopic changes in the breast post-neoadjuvant therapy. (A) Normal breast with atrophic terminal duct lobular unit
and stromal fibrosis. Note prominent myoepithelial cell layer and nuclear hyperchomasia of epithelial cells. (B) Fibrous tumour bed without residual
carcinoma and absence of normal breast parenchyma. Higher magnification of fibrous stroma (insert). (C) Tumour bed with chronic inflammation,
foamy macrophages, giant cells and cholesterol clefts without evidence of residual carcinoma. (D) Clip-reaction site in the tumour bed without
evidence of residual carcinoma. (E) Scattered small nests of tumour cells in tumour bed (black asterisk), representing a residual tumour cellularity of
approximately 1%. (F) The only focus of residual carcinoma is identified adjacent to a clip-site reaction (black asterisk) highlighting the importance of
identifying the radiological clip. Higher magnification (insert) demonstrates invasive carcinoma. (G) Heterogeneous response to neoadjuvant therapy.
Tumour area with near-complete treatment response (white asterisk) adjacent to an area with minimal treatment response (black asterisk). Overall,
this represents an average residual tumour cellularity of 30%. (H) Minimal response to neoadjuvant therapy with residual carcinoma within fibrotic
stroma and an average residual tumour cellularity of 50%. (I) Prominent retraction artefact surrounding the nests of tumour cells mimicking lymphatic
vascular invasion (LVI). (J) Residual invasive ductal carcinoma cells with foamy cytoplasm mimicking histiocytes. Insert demonstrates CAM 5.2
immunohistochemical staining in these cells, confirming their epithelial nature. (K) Single filing of residual invasive ductal carcinoma cells. (L) The only
residual carcinoma that can be identified is present as lymphovascular tumour emboli (LVI).

Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598 123


Review

Figure 5  Spectrum of microscopic changes in axillary lymph nodes post-neoadjuvant therapy. (A) Atrophic lymph node demonstrating sinus J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
histiocytosis and haemosiderin-laden macrophages with no evidence of metastatic carcinoma or scarring. (B) Lymph node with a small subcapsular
scar and no evidence of residual metastatic carcinoma. (C) Lymph node with a prominent fibrous scar and foamy macrophages. Fibrous scar extends
beyond the lymph node capsule suggesting pre-treatment extranodal extension or pre-treatment tissue sampling (fine-needle aspiration or core
biopsy). (D) Lymph node completely replaced by a fibrous scar and without evidence of residual carcinoma. Immunohistochemistry for keratins may
be useful to rule out isolated tumour cells. (E) Isolated tumour cells marked with a white asterisk identified in subcapsular metastatic tumour bed. (F)
Micrometastatic carcinoma (white asterisk) with minimal scarring. (G) Macrometastatic carcinoma with a background of fibrosis indicating partial
response of metastatic carcinoma to neoadjuvant therapy. (H) Macrometastatic carcinoma without any evidence of treatment response.

entirety.13 18 Residual tumour mass, tumour bed and radiolog- Neoadjuvant mastectomy specimens are inked fresh according
ical clip site should be noted and measured. Tumour bed grossly to the institutional protocols and sliced at 0.5 cm to 1.0 cm inter-
extending to the resection margins should be documented. With vals to ensure formalin penetration. Grossly identified tumour(s),
larger lumpectomy specimens, targeted representative sections tumour bed(s) and clip site(s) should be documented. Sampling
are taken with careful and accurate representation of the tumour should cover the pre-treatment size of the tumour and tissue just
bed, surrounding adjacent tissue and resection margins. beyond the visible lesion should be sampled for evaluation of
124 Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598
Review
microscopic extent.13 In line with BIG-NABCG recommenda- more prominent (figure 4). Occasional atypical epithelial cells

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
tion, when residual carcinoma is grossly identified, one full-face with large hyperchromatic nuclei, visible nucleoli and vacuo-
cross-section of the slice with the largest tumour area should be lated cytoplasm may be seen and should not be mistaken for
submitted for microscopic assessment. For large tumours, five non-treatment-related significant epithelial atypia.
representative sections of such a full-face are submitted. Several Chemotherapy-induced changes in breast carcinoma result
such full-face sections 1 cm apart may be submitted to deter- in stromal fibrosis, oedema and fibromyxoid changes with
mine the full extent of the tumour. This approach is sufficient to chronic inflammatory infiltrate, haemosiderin-laden macro-
assess the tumour size and cellularity for both the American Joint phages, foreign body giant cells, and cholesterol clefts with
Committee on Cancer (AJCC) staging and the residual cancer a total absence of normal breast ducts and TDLUs (figure 4).
burden (RCB) calculations.16 25 38–40 Haemosiderin-laden macrophages, foreign body giant cells and
Residual tumour and tumour bed may be difficult to identify in fat necrosis are also present in biopsy site reactions and should
patients with a good response to neoadjuvant therapy. Correla- not be confused with treatment response. Tumour response to
tion with preoperative imaging should be used to help localise preoperative systemic therapy is highly variable with hetero-
the tumour site and, if possible, specimen radiographs should be geneous change in tumour size and cellularity ranging from
used to locate the clip site and/or calcifications, if known to be complete resolution, concentric contraction, scattered patterns
associated with tumour prior to treatment. The pre-treatment of residual disease, to minimal response. Even in the absence of a
size of the tumour will determine the extent of the sampling. significant decrease in tumour size, carcinomas are typically less
A systematic approach is preferred over exhaustive sampling cellular post-chemotherapy and the distribution of cells varies
of fibrotic areas or blind submission of an arbitrary number of from uniform to haphazard.1 Residual cancer cells, if present, are
blocks. This is achieved by sampling of the correct area best seen as infiltrating cords, nests and singly dispersed individual
identified by careful clinical and radiological correlation.16 cells resembling lobular carcinoma or histiocytes (figure 4).43
BIG-NABCG recommends the following approach when no Difficult cases may require immunohistochemistry to distinguish
residual carcinoma is identified: one full-face (or five represen- between residual tumour cells and histiocytes (figure 4). Most
tative sections of a full-face for large tumours) of the cross-sec- common cytomorphological changes induced by chemotherapy
tion of the pre-treatment area of involvement per 1 cm (1–2 cm include nuclear hyperchromasia, nuclear pleomorphism, abun-
for large tumours) of pre-treatment size up to a maximum of 25 dant hypereosinophilic cytoplasm and cytoplasmic vacuolisation.
blocks.13 16 Conversely, the US FDA recommends at least one Apart from decreased cellularity, most carcinomas do not change
block per centimetre of the pre-treatment tumour size, or at least in appearance and overall histological grade.1 Some tumours
10 blocks in total, whichever is greater.12 16 The Royal College of may appear to be of higher nuclear grade because of cytomor-
Pathologists (UK), however, does not make any distinct recom- phological changes induced by chemotherapy17 45 46; however,
mendations for specimen sampling in the neoadjuvant setting.41 this is usually offset by decreased tumour cell mitotic activity.
In the case of multifocal disease, each tumour should be handled Nevertheless, actual change in tumour histological grade can
in the same manner and sections documenting breast tissue in only be assessed by comparison of the post-treatment tumour
between the tumours should be submitted. Additional sections to the pre-treatment tumour in the needle core biopsy, if one is
may have to be taken in cases where microscopic findings do not available. Even then, tumour heterogeneity has to be considered.
correlate with the gross impression. Change in the histological grade of cancers post-chemotherapy
has not been correlated with clinical outcome and its prognostic
significance is unknown.1 It is important to note that pre-treat-
Axillary contents
ment and post-treatment tumour histological grade, tumour size
As with breast specimens, preparation and processing protocols
and lymphovascular invasion (LVI) are correlated with distant
for axillary contents/sentinel lymph nodes do not differ in the
disease-free survival.45
neoadjuvant from the non-neoadjuvant setting. All identified
Ductal carcinoma in situ (DCIS) and lymphovascular
lymph nodes should be sectioned at 2 mm intervals in the long
tumour emboli are relatively resistant to treatment compared
axis of the lymph node and, in the absence of gross evidence of
with carcinoma invading the stroma and may represent the
disease, entirely submitted for microscopic examination. Serial
only residual disease (figure 4).46 Residual tumour nests
slicing at 2 mm intervals results in detection of most macro-
may show marked retraction artefact in the fibrous stroma
metastases.42 Care should be taken to identify and document
mimicking lymphovascular invasion and immunohistochem-
radiological clips placed following prior FNA procedures and
istry may be useful to distinguish the two (figure 4). DCIS can
correlation with specimen radiographs may be helpful in these
show a variable response to therapy from minimal alteration
cases.32 When no gross lymph nodes are identified, any tissue
to reduced distension and replacement of neoplastic cells by
that may represent a lymph node should be submitted for micro-
macrophages.47 In some cases, only ducts with rare residual
scopic examination.18 All grossly involved lymph nodes should
neoplastic cells with marked nuclear atypia remain after treat-
be documented along with grossly evident extranodal disease.
ment. In these cases, it can be particularly challenging to
Representative sections of grossly involved lymph nodes demon-
distinguish foci of residual DCIS from benign ducts with treat-
strating the largest focus of metastatic carcinoma and extranodal
ment artefact.
extension are appropriate.16
Margins can be difficult to evaluate in the post-neoadjuvant
setting when there has been marked treatment response. In such
Microscopic examination cases, the extent of the tumour bed can be difficult to appreciate
Breast tissue specimens on radiological imaging, intraoperatively by the surgeon and even
Chemotherapy-induced changes in normal breast tissue are well macroscopically by the pathologist, and it may not be possible to
documented. These include significant atrophy of the terminal obtain an excision of the entire tumour bed surrounded by a
ductal lobular units (TDLUs).43 44 Lobular acini appear reduced rim of normal tissue.1 In patients with a pCR, the significance
with attenuation of lobular/ductal epithelium, intralobular of tumour bed at the margin is unclear. However, in cases with
sclerosis and, as a result, the myoepithelial cell layer becomes scattered residual foci of invasive carcinoma, presence of tumour
Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598 125
Review
bed at the resection margin in breast-conserving specimens may

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
Table 1  Summary elements to be included in the post-neoadjuvant
predict for residual carcinoma in the breast.1
pathology report
Breast specimen
Axillary lymph nodes
Gross examination Comments
Axillary lymph nodes may become small and atrophic following
1. Time and date of tissue excision, placement Fixation and cold ischaemic
neoadjuvant therapy with depletion of lymphocytes and with
of tissue in formalin and duration of fixation in time are important if biomarker
prominent sinus histiocytosis (figure 5).43 Lymph node metastases formalin testing needs to be reported
with complete response to neoadjuvant therapy show hyaline 2. Procedure type
stromal scars, mucin pools, aggregates of histiocytes and haemo-
3. Tissue labelling, orientation by the surgeon and
siderin without viable tumour cells (figure 5). In some cases, a subsequent painting of the resection margins
diagnosis of pCR in lymph nodes cannot be definitively made as 4. Specimen dimensions: breast tissue, skin, nipple Depends on the procedure type
metastases can resolve without a scar or with minimal scarring and areola
(figure 5).1 Changes related to previous FNA can be identical 5. Description of any grossly visible lesion with Appearance, texture,
to regressed metastasis and cannot be reliably distinguished.48 dimensions and distances to the resection margins demarcation, relationship to
Partial response to therapy, on the other hand, is character- other lesions if present
ised by small clusters and individual tumour cells set in hyaline 6. If no visible gross lesion, description of fibrous
stromal fibrosis (figure 5). It is important to note any treatment parenchyma, radiological clip or biopsy site
effect in positive lymph nodes as these patients have better reaction
disease-free survival and lower relapse rates than patients with 7. Description of the uninvolved breast tissue Fatty or fibrous
positive lymph nodes without any treatment effects (figure 5).1 49 8. Specimen photography or diagrams of the
It is also recommended to report any histological findings in the sectioned breast tissue with gross lesion(s)
lymph nodes containing radiological clips.32 Overall, the pres- 9. Note if specimen X-rays were performed and
ence of residual lymph node disease confers a worse prognosis reported
regardless of the tumour response in the breast.50 A summary of 10. Mapping of all tissue sections submitted for With the aid of photograph or
microscopic examination diagram
all elements to be included in the post-neoadjuvant pathology
Microscopic examination Comments
report is provided in table 1.
While the AJCC 8th edition guidelines remain the same 1. Specimen, procedure and neoadjuvant setting
for assessing the metastatic involvement of the lymph nodes 2. Type of residual carcinoma If present
regardless of the treatment status, it is imperative to note that 3. Histological grade of residual tumour
the significance of lymph node metastases drastically changes 4. Dimensions:
post-chemotherapy. Post-treatment isolated tumour cells (ITCs) ►► Size of largest contiguous focus of invasive
carcinoma for AJCC staging (include span and
and micrometastases predict worse survival compared with the
focality of residual carcinoma)
same findings in the non-neoadjuvant setting.17 Furthermore, ►► Size of largest cross section of residual invasive
survival in patients with lymph node metastases measuring <1 carcinoma (“residual tumour bed”) in 2
mm and <2 mm (micrometastases) is similar to that of patients dimensions for RCB score
with macrometastases in the post-neoadjuvant setting.17 5. Tumour cellularity (%) Average cellularity across the
Molecular assays for assessment of sentinel lymph nodes are largest cross-section containing
discouraged by the BIG-NABCG as they do not allow evaluation residual carcinoma (including
of treatment response in the lymph node and the assays have not invasive and in situ disease)
been calibrated for detection of ITCs.18 51 6. Percentage of cancer within tumour bed that
represents in situ disease
7. Lymphovascular invasion
Complete pathological response and residual 8. Confluent tumour necrosis
disease classification systems 9. Distance to closest margins Include tumour bed at margins
Until recently, various definitions for complete pathological 10. Treatment effect Describe presence of treatment
response have been applied in neoadjuvant clinical trials in breast effect, biopsy site reaction, clip-
cancer, which hindered any cross-trial data interpretation.12 18 site reaction
The US FDA defines pCR as ypT ypN0 and ypTis ypN0.12 52 11. Other findings Including calcifications
Similarly, the AJCC 8th edition states that any residual inva- 12. Biomarker status If required
sive carcinoma in the breast or lymph nodes precludes classi-
Axillary lymph nodes
fication as pCR.40 This includes the presence of tumour within
lymphatic or vascular spaces (LVI) as well as isolated tumour Gross examination Comments
cells in lymph nodes.25 Conversely, the presence of in situ carci- 1. Tissue labelling, orientation by the surgeon and Proper precautions and disposal
localisation technique of radioactive material
noma after treatment (DCIS or LCIS) in the absence of residual
2. Specimen dimensions: for the overall specimen, Note any grossly identified
invasive disease constitutes a pCR.25 Mounting data with a
for each lymph node identified, and for the extranodal extension
strong consensus exists to include absence of invasive disease greatest metastatic focus identified
in the breast and the lymph nodes as a standard definition of
3. Description of lymph nodes Appearance of sectioned lymph
pCR.52–60 Patients with residual carcinoma that is present only in nodes
the lymph nodes have considerably inferior prognoses.52–60 On 4. Presence or absence of clip(s) Correlate with specimen X-ray
the other hand, the significance of in situ carcinoma is unclear Microscopic examination Comment
and patients with ypTis ypN0 have comparable survival with
1.Type of lymph nodes examined: sentinel and
ypT0 ypN0 patients.52 61
non-sentinel
Depending on the type and duration of neoadjuvant chemo-
therapy, different rates of pCR have been reported between breast Continued

126 Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598


Review
The AJCC ypT category is based on the largest contiguous

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
Table 1  Continued
focus of residual invasive carcinoma (figure 6). Treatment-re-
Axillary lymph nodes lated fibrosis adjacent to residual disease is not included when
2. Number of lymph nodes identified and involved measuring the maximum dimension of the tumour. Cases with
by the metastatic carcinoma scattered tumour nests that were part of a single tumour mass
3. Dimensions: before treatment will result in systemic artificial downstaging by
►► Size of single largest continuous metastatic this system.13 For this reason, AJCC also recommends a descrip-
focus for AJCC staging tion of the extent of residual carcinoma to be included in the
►► Size of the largest extent of lymph node
involvement by tumour cells including
pathology report as this may assist the clinician in estimating
intervening fibrosis for RCB score the extent of residual disease. Where possible, inclusion of the
4. Presence of extranodal extension pre-treatment clinical tumour stage (cT) category should be docu-
5. Treatment effect: number of lymph nodes with Including FNA/biopsy site
mented. When the only residual cancer in the breast is present
possible treatment effect including negative lymph reaction and clip-site reaction as lymphovascular tumour emboli (LVI), the tumour is classified
nodes as ypT0; however, as noted above, pCR cannot be assigned in
AJCC, American Joint Committee on Cancer; FNA, fine-needle aspiration; RCB, these cases. Similarly, the largest contiguous tumour focus in the
residual cancer burden. lymph nodes is used for ypN classification and adjacent treat-
ment-related fibrosis is not included in the measurements. While
ITCs after chemotherapy is still considered ypN0(i+), it cannot
cancer subtypes (table 2).45 52 62–67 However, consistent patterns
be regarded as a pCR. If a cancer is categorised clinical or patho-
have emerged to demonstrate highest pCR rates among patients
logical M1 prior to therapy, it remains classified as M1 regardless
with HER2-positive tumours, followed by those with triple-neg-
of the observed response to therapy.25 40 The post-neoadjuvant
ative tumours and finally, patients with hormone receptor–posi-
yAJCC 7th edition classification has been shown to have prog-
tive tumours.45 52 62–67 Even though hormone receptor–positive
tumours are relatively chemoresistant, they do have the best nostic value.62 71
prognosis in the absence of chemotherapy and pCR rates cannot The RCB system calculates a prognostic score based on (1) a
be used reliably to predict recurrence-free survival.62 two-dimensional measurement of the tumour bed demarcated
Pathological response (ie, reduction in pT and/or pN category) by residual carcinoma (figure 6), (2) the average residual tumour
that is less than a complete response is labelled a partial patho- cellularity (invasive and in situ disease), (3) the percentage of
logical response (pPR), while no apparent change in the T or N residual disease that represents in situ disease, (4) the number
categories compared with pre-treatment assignment, or increase of positive lymph nodes and (5) the diameter of the largest
in the T or N category indicates no response (NR) to treatment.25 metastatic focus, inclusive of intervening treatment-related
Alternatively, some systems use a reduction in tumour cellularity, fibrosis.38 A mathematical formula combines these variables into
as compared with pre-treatment tumour cellularity, to define a a continuous index to define four RCB categories; RCB-0 for
pPR to therapy.2 68–70 pCR and RCB-1–3 represent progressively greater extents of
Many studies have attempted to provide criteria for evalu- residual cancer. A web-based calculator and detailed instructions
ating pathological response after treatment. A limited list of such are publicly available to calculate the RCB scores (http://www.​
studies with their proposed classification categories is provided mdanderson.​org/​breastcancer_​RCB).
in table 3. There are some pitfalls to be aware of when calculating the
Merits of each classification system are beyond the scope of this RCB (summarised in Bossuyt, Surgical Pathology Clinics, March
review; however, they collectively demonstrate that the degree 2018).16 The residual tumour bed may differ in size from the
of response to treatment correlates with survival. Different clas- microscopic ‘fibrous’ tumour bed as well as from the grossly
sification systems also yield different estimates of future risk.13 60 identified tumour bed. The perimeter of the residual tumour
The two most widely used systems to measure residual disease bed is defined by residual cancer cells, even if these are widely
are the AJCC system, currently in its 8th edition, and the RCB scattered and separated by treatment-induced fibrosis. There-
calculator (table 4).25 38 fore, this measurement is determined following microscopic
Of note, when clearly separate tumours are identified grossly evaluation and it may be smaller or larger than the gross tumour
that are confirmed microscopically to have intervening areas bed measurement. Determining the dimensions of the residual
of normal breast tissue and when these presented as multifocal tumour bed can be aided by dotting the cancer perimeter in
disease pre-treatment, the dimension of the largest focus is used each slide and reconstructing the three-dimensional extent of
for staging using AJCC (with use of the (m) modifier, repre- the disease with the help of a block diagram. Based on this, the
senting multiple foci of residual disease) and for determining the two largest dimensions can be measured and recorded (figure 6).
tumour bed dimensions for the purpose of calculating the RCB Within the dotted perimeter of the largest cross-sectional area,
(see below). the percentage tumour cellularity should be assessed in each

Table 2  pCR rates across breast cancer subtypes


HR+/HER2− HR+/HER2+ HR−/HER2+ HR−/HER2−
pCR rates 4.4%–9% 17.3%–33% 36.4%–50.3% 31.1%–36.8%
Comments Best prognosis without Best survival, pCR less critical for Prognosis poor if pCR not achieved pCR associated with excellent
chemotherapy outcome prognosis
Lymph node conversion rates 13.1%–21% 36.6% 46.8%–64.7% 46.4%–49.4%
Ranges reflect different chemotherapy regimens, accrual methods and duration of therapy across trials. Increased pCR rates in HER2-amplified cancers were observed following
addition of trastuzumab or pertuzumab therapy, which are reflected in wide ranges of pCR rates.45 52 62–67
pCR, pathological complete response.

Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598 127


Review

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
Table 3  Systems for categorising neoadjuvant treatment response Table 3  Continued
System Categories Comments System Categories Comments
Feldman98 Group A: No macroscopic evidence of cancer in By macroscopic Clinical-Pathological Based on combined clinical stage (0–2 marks), HER2 status included
1986 mastectomy or axillary contents examination only Stage–Oestrogen/Grade pathological stage (0–2 marks), ER negative status (1 in new Neo-Bioscore
Group B: Macroscopic evidence of cancer in (CPS-OG) 84
mark) and high nuclear tumour grade (1 mark) system85
mastectomy specimen 2011
Bonadonna68 Grade 1: No overall reduction in tumour cells Requires comparison to Neoadjuvant Response Pre-treatment clinical stage by MRI minus post-
1990 compared with pre-treatment core biopsy pre-treatment biopsy Index (NRI)83 treatment ypT stage with additional points for near-
Grade 2: Mild loss of invasive cells specimen 2010 pCR (1 point) or pCR (2 points)
Grade 3: Considerable, up to 90%, reduction in 0=no response to therapy and 1=pCR
tumour cells
Grade 4: Marked reduction of tumour cells, only Residual Disease in Residual tumour size (cm)×0.2+index of positive Based on modified
clusters of dispersed cells detected Breast and Nodes lymph nodes (0–3: none, 1–4, 5–7 and >7)+tumour Nottingham Prognostic
Grade 5: No invasive carcinoma, DCIS only (RDBN)82 grade Index
2008 Level 1: pCR
Sinn99 0: No effect Requires comparison to Level 2: 0.1–2.9
1994 1: Resorption and tumour sclerosis pre-treatment biopsy Level 3: 3.0–4.3
2: Minimal residual invasive tumour (<0.5 cm) specimen Level 4: >4.4
3: Residual non-invasive tumour only
4: No tumour detectable yAJCC 8th edition25 pCR: no invasive carcinoma in breast and lymph node, Anatomical stage
2017 no LVI and no ITCs (ypT0 N0, ypTis pN0) groupings based on the
6
Chevallier Class 1: Disappearance of all tumour, microscopically Developed for pPR: reduction in T and/or N stage TNM staging system
1993 or macroscopically inflammatory carcinoma NR: no change or increase in the T or N stage
Class 2: DCIS only, no invasive tumour and negative
lymph nodes DCIS, ductal carcinoma in situ; ITC, isolated tumour cell; LVI, lymphatic vascular invasion; NR, no response; pCR,
Class 3: Presence of invasive carcinoma with stromal pathological complete response; pPR, partial pathological response.
alterations
Class 4: Few modifications in tumour appearance
Sataloff100 Tumour
1995 T-A: total or near total therapeutic effect; <5%
slide and the average cellularity calculated to include all fields
tumour surface that fall within the tumour bed perimeter, even those with very
T-B: >50% therapeutic effect but less than T-A
T-C: <50% therapeutic effect
low cellularity or no disease. Tools are freely available at http://
T-D: no therapeutic effect www.​mdanderson.​org/​breastcancer_​RCB that provide a visual
Nodes
N-A: no metastatic disease with evidence of
aid in estimating the percentage of residual tumour cellularity.
therapeutic effect RCB measurements are based on the largest residual tumour
N-B: no nodal metastases or therapeutic effect
N-C: metastases present with therapeutic effect bed. If smaller foci demonstrate higher cellularity, calculating
N-D: metastases present without therapeutic effect separate RCBs for these foci is worthwhile as these may yield
Honkoop101 pCR: no residual cancer in mastectomy or axillary pCR and MPR are higher RCB scores.
1998 lymph nodes regarded as one group
MPR: microscopic disease (scattered foci) with normal The RCB system has been shown to be reproducible.72 It
macroscopic examination provides prognostic value independent of the yAJCC stage for
MRD: gross residual disease with macroscopic tumour
or diffuse tumour infiltration microscopically patients with stage II and III disease and across all phenotypic
Kuerer102 1. No evidence of residual tumour subgroups of breast cancer.38 73 This has been shown to be the
1998 2. <1 cm3 of residual tumour macroscopically, case in multiple different institutional cohorts and with long-
residual microscopic tumour foci
3. >1 cm3 of residual tumour macroscopically term follow-up.45 60 62 74–76
Pinder2 Tumour Requires comparison A recent study compared the RCB and the AJCC staging
2007 1. pCR: (i) no residual carcinoma or (ii) DCIS only
2. Partial response: (i) <10% tumour cellularity,
with pre-treatment
biopsy specimen
systems using the I-SPY 1 trial dataset.62 This study used the
(ii) 10%–50% tumour cellularity, >50% tumour seventh edition of yAJCC staging system; however, this should
cellularity compared with pre-treatment biopsy
3. No response to therapy not have impacted the anatomical TNM tumour staging based on
Nodes the eighth edition. Both systems identify patients with interme-
1. No metastasis and no therapy changes
2. No metastasis but evidence of response diate (4-fold) risk of relapse (RCB-1–2 and yAJCC1–2) and high
3. Metastasis with evidence of response (11-fold) risk of relapse (RCB-3 and yAJCC3).62 Both systems
4. Metastasis without response
show the strongest association with recurrence-free survival in
NSABP B-1817 pCR: no invasive tumour cells present
2002 pPR: scattered individual or small clusters of tumour triple-negative breast cancers, while hormone receptor positive
cells in a desmoplastic/hyalinised stroma cancers have relatively low recurrence rates regardless of RCB
pNR: tumour lacking changes in pCR and pPR
or yAJCC class.62 Patients who are RCB-3 or yAJCC3 and have
Miller-Payne System69 Grade 1: no reduction in overall cellularity Requires comparison
2003 Grade 2: minor loss of tumour cells (<30%) with pre-treatment triple negative or HER2+ tumours have a higher risk of relapse
Grade 3: 30%–90% reduction in tumour cells
Grade 4: >90% loss of tumour cells
biopsy specimen
than all other patients.62 The study also found that in 34% of
Grade 5: no invasive cells present, DCIS may be cases, RCB and yAJCC staging systems are discrepant and this
present
is attributed to unequal weighting of positive lymph nodes
Smith70 Tumour Modification of the
2002 1. No reduction in overall cellularity Miller-Payne System to
and the weighting of tumour cellularity in RCB. In discrepant
2. Mild reduction in tumour cells include lymph nodes cases, however, if one classification system identifies a patient as
3. Up to 90% reduction in cellularity
4. Marked disappearance with only small clusters having a high risk of recurrence, then that patient tends to have
remaining an increased rate of recurrence regardless of the other system’s
5. No invasive tumour, DCIS remaining
Nodes ranking.62 Therefore, there is benefit to computing both scores
1. True negative: no metastasis and no changes for patients in order to identify those with the highest risk of
2. Metastasis with no changes
3. Metastasis with changes relapse.
4. No metastasis, but changes With rising interest in immunotherapeutic strategies, eval-
Residual Cancer Burden RCB-0: no carcinoma in breast and lymph node Based on an online uation of tumour-infiltrating lymphocytes (TILs) has garnered
(RCB)38 RCB-I: partial response (minimal residual disease) calculator
2007 RCB-II: partial response (moderate residual disease)
considerable research interest. Emerging evidence indicates
RCB-III: chemoresistant (extensive residual disease) that TIL density may be predictive for response to neoadju-
Continued vant therapy.77 A recent study indicates that high pre-therapy
TIL levels may be predictive of pCR for HER2-amplified breast
cancers.78 Furthermore, high TILs in post-neoadjuvant residual
128 Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598
Review

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
Table 4  Comparison of the AJCC 8th edition and RCB system
AJCC 8th edition RCB
Pathological complete response (pCR) No evidence of invasive carcinoma in the breast and axillary lymph No evidence of invasive carcinoma in the breast and axillary
nodes (ypT0 ypN0 or ypTis ypN0) lymph nodes
Tumour size Largest contiguous focus of residual invasive carcinoma for ypT stage Two dimensions of the largest cross-section of residual
carcinoma defines residual tumour bed
Tumour cellularity Not applicable Average cellularity of residual carcinoma
Tumour focality Multiple separate foci of residual invasive carcinoma designated as Calculate RCB for largest tumour focus±smaller foci if these
mypT stage show higher cellularity
LVI identified only Considered ypT0, but not pCR Area of the breast involved by LVI only determines the
residual tumour bed (in two dimensions)
Direct extension beyond breast Stage as for non-neoadjuvant setting Not applicable
Lymph nodes Size of largest contiguous metastatic focus for ypN staging Size of the largest extent of lymph node involvement by the
tumour cells including intervening fibrosis
No of lymph nodes involved Determines ypN stage, ITCs still considered ypN0(i+) All positive lymph nodes included
Location of lymph nodes involved Influences ypN2, ypN3 and ypN4 stages All positive lymph nodes included
Pathological partial response (pPR) Reduction in T and/or N stage Determined by the formula: RCB-I or RCB-II
No response (NR) No apparent change in the T or N categories or an increase in the T or Determined by the formula: RCB-III
N category
AJCC, American Joint Committee on Cancer; ITC, isolated tumour cell; LVI, lymphatic vascular invasion; RCB, residual cancer burden.

cancers may be an important predictor of survival in triple-neg- future classifications of neoadjuvant treatment response are in
ative breast cancers.79 80 The International Immuno-Oncology progress.77
Biomarker Working Group on Breast Cancer has recently More complex staging systems that combine clinical and patho-
published recommendations to assess TILs in residual tumours logical stage, tumour histological grade, proliferation index,
following neoadjuvant therapy.77 Initiatives to include TILs in biomarker status and/or molecular testing are continuously

Figure 6  Illustration contrasting AJCC and RCB measuring techniques. (A) In the breast tissue, measurement of the single largest tumour focus
(black oval) is used to determine AJCC ypT stage (blue arrow). Measurements of the residual tumour bed for RCB (red arrows) is determined by the
span of residual invasive carcinoma (black circles) within a fibrous scar (pink star) and measured in two dimensions as outlined by the red rectangle.
The residual tumour bed can be smaller or larger than the fibrous scar. Average tumour cellularity is estimated within the residual tumour bed taking
into account the invasive and in situ (pink circles) carcinoma. (B) A similar approach is taken to measure metastatic carcinoma in the lymph node.
Measurement of the single largest metastatic focus (black oval) is used to determine AJCC ypN stage (blue arrow). Measurement for the RCB (red
arrow) is determined by the span of metastatic carcinoma (black circles) inclusive of intervening fibrosis (pink oval). Distant metastatic foci or ITCs
(black circles outside pink oval) are not included in the measurement. (C) When more than one focus of invasive carcinoma within distinct tumour
beds are identified and separated by normal breast tissue, then each focus is measured separately. The dimension of the largest ‘dominant’ focus
(blue arrow) is used for ypT AJCC classification and the (m) modifier is applied. Tumour bed dimensions (red arrows and red rectangle) and cellularity
are calculated for the largest focus using RCB methods. If the additional focus demonstrates different residual tumour cellularity, then RCB can be
calculated for the second focus as it may yield different RCB classification. Note that when the tumour demonstrates concentric shrinkage, the yAJCC
and RCB maximum dimensions are equivalent. (D) When a single focus of metastatic carcinoma is identified in the tumour bed, the ypN AJCC and
RCB dimensions are also equivalent. AJCC, American Joint Committee on Cancer; ITC, isolated tumour cell; RCB, residual cancer burden.
Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598 129
Review
emerging. The Nottingham Prognostic Index was one of the first

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
systems to combine tumour size, histological grade and lymph
Take home messages
node status.81 While this system was not developed for use in the
►► A multidisciplinary approach when handling neoadjuvant
neoadjuvant setting, it forms the basis of the Residual Disease in
breast specimens is imperative with review of previous
Breast and Nodes system.82 The Neoadjuvant Response Index is
pathology and clinical documentation prior to specimen
based on pre-treatment clinical/radiological measurements and
examination.
post-treatment pathological measurements.83 Clinical-patho-
►► Correlation of macroscopic findings and diagnostic imaging
logical stage–oestrogen/grade (CPS-OG) combines pathological
is essential.
stage post-therapy with clinical stage pre-therapy, tumour nuclear
►► A standardised and systematic approach to tissue sampling
grade and ER status.84 CPS-EG was subsequently modified to
yields the most useful microscopic results.
incorporate the tumour HER2 amplification status in a system
►► Mindfulness of potential pitfalls and understanding the
called Neo-Bioscore.85 Post-treatment Ki-67 indices have been
limitations of applied classification system can help avoid
correlated with long-term outcome for neoadjuvant endocrine
misclassification of patients.
therapy and chemotherapy.86–88 Ki-67 index is increasingly being
►► Standardisation in reporting of neoadjuvant specimens
used as a component of newer multivariate prediction models in
will provide the most useful information to the clinicians
the post-neoadjuvant setting such as the Preoperative Endocrine
regarding patient outcomes.
Prognostic Index and Residual Proliferative Cancer Burden,
►► Recent evidence indicates possible benefits to reporting
the latter representing a modification to the RCB system that
additional classification system to the yAJCC 8th edition in
combines post-therapy Ki-67 index with the RCB.75 86 Finally, as
assessing treatment response to help identify patients at
the latest AJCC 8th edition of breast cancer staging includes new
highest risk of relapse.
parameters for prognostic staging that combine anatomic TNM
factors, tumour grade, biomarker status and multigene panel
testing,25 it seems probable that such a system will eventually
expand into the post-therapy yAJCC staging. Currently, there most updated yAJCC staging system and, if no other classifi-
are limited data on the use of multigene panel testing, such as cation systems are routinely reported, it is recommended that
Oncotype DX, in the neoadjuvant setting.89 90 sufficient information be provided so that other classification
systems, like the RCB, can be applied if needed.
Biomarkers status post-systemic therapy
Handling editor  Runjan Chetty.
There is no consensus on reassessment of hormone receptor and
HER2 status in residual cancer post-neoadjuvant therapy.18 In Contributors  All authors listed contributed to this manuscript.
general, tumour markers do not change before and after chemo- Funding  The authors have not declared a specific grant for this research from any
therapy in the majority of cases18 91–93; however, discordant funding agency in the public, commercial or not-for-profit sectors.
results have been reported for ER (13%–18%), PR (26%–32%) Competing interests  None declared.
and HER2 (6%–9%) before and after chemotherapy with various Patient consent for publication  Not required.
causes for the discordances being described.94 95 In some cases, Provenance and peer review  Commissioned; internally peer reviewed.
change in tumour marker expression may be related to the type
of neoadjuvant chemotherapy.1 96 Loss of HER2 amplification References
following neoadjuvant trastuzumab has been reported and also 1 Sahoo S, Lester SC. Pathology of breast carcinomas after neoadjuvant chemotherapy:
found to be associated with a worse outcome.97 Biomarkers must an overview with recommendations on specimen processing and reporting. Arch
be reassessed in cases where retesting for biomarkers is required Pathol Lab Med 2009;133:633–42.
2 Pinder SE, Provenzano E, Earl H, et al. Laboratory handling and histology reporting
by the clinical trial protocol and in cases where the biomarker
of breast specimens from patients who have received neoadjuvant chemotherapy.
status was previously unknown.18 Reassessment of biomarkers Histopathology 2007;50:409–17.
should be considered for negative or equivocal results on 3 Viale G. Characterization and clinical impact of residual disease after neoadjuvant
pre-treatment core biopsy, in cases where biopsies had insuffi- chemotherapy. Breast 2013;22(Suppl 2):S88–S91.
cient invasive carcinoma for accurate assessment, where biopsies 4 Hortobagyi GN, Ames FC, Buzdar AU, et al. Management of stage III primary
breast cancer with primary chemotherapy, surgery, and radiation therapy. Cancer
were performed and biomarkers assessed at other institutions, in 1988;62:2507–16.
post-treatment tumours demonstrating heterogeneous morphol- 5 Valagussa P, Zambetti M, Bonadonna G, et al. Prognostic factors in locally advanced
ogies or in cases where the tumour did not demonstrate any noninflammatory breast cancer. Long-term results following primary chemotherapy.
response to therapy.18 Routine biomarker reassessment is not Breast Cancer Res Treat 1990;15:137–47.
6 Chevallier B, Roche H, Olivier JP, et al. Inflammatory breast cancer. Pilot study of
recommended for triple-positive breast cancers.18
intensive induction chemotherapy (FEC-HD) results in a high histologic response rate.
Am J Clin Oncol 1993;16:223–8.
7 Schwartz GF, Hortobagyi GN, Masood S, et al. Proceedings of the consensus
Summary conference on neoadjuvant chemotherapy in carcinoma of the breast, April 26–28,
As neoadjuvant systemic therapies are becoming increasingly 2003, Philadelphia, PA. Hum Pathol 2004;35:781–4.
used in patients with breast cancer, a standardised approach to 8 Mauri D, Pavlidis N, Ioannidis JP. Neoadjuvant versus adjuvant systemic treatment in
the evaluation of post-neoadjuvant systemic therapy specimens is breast cancer: a meta-analysis. J Natl Cancer Inst 2005;97:188–94.
9 Kaufmann M, von Minckwitz G, Bear HD, et al. Recommendations from an
essential. Pathological evaluation of tumour response to neoad-
international expert panel on the use of neoadjuvant (primary) systemic treatment of
juvant chemotherapy remains the gold standard for judging operable breast cancer: new perspectives 2006. Ann Oncol 2007;18:1927–34.
the effectiveness of the therapy. As novel classification systems 10 Early Breast Cancer Trialists’ Collaborative Group (EBCTCG). Long-term outcomes for
for treatment responses are continuously developed, patholo- neoadjuvant versus adjuvant chemotherapy in early breast cancer: meta-analysis of
gists will remain at the forefront to drive the development of individual patient data from ten randomised trials. Lancet Oncol 2018;19:27–39.
11 Esserman LJ, Woodcock J. Accelerating identification and regulatory approval of
this field. Recent evidence demonstrates benefits of using more investigational cancer drugs. JAMA 2011;306:2608–9.
than one staging system to identify patients at the highest risk 12 US Food and Drug Administration. Guidance for industry: pathological complete
of relapse. Therefore, the pathology reports should provide the response in neoadjuvant treatment of high-risk early-stage breast cancer: use as

130 Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598


Review
an endpoint to support accelerated approval. 2018. Available: http://www.​fda.​ evaluation, pathology protocol for macroscopic and microscopic assessment of RCB].

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
gov/d​ ownloads/​Drugs/G ​ uid​ance​Comp​lian​ceRe​gulat​ ory​Infor​ mation/​Guidances/​ 2018. Available: http://w​ ww3.​mdanderson.​org/​app/m ​ edcalc/​index.​cfm?​pagename=​
UCM305501.​pdf [Accessed 7 Nov 2018]. jsconvert3 [Accessed 7 Nov 2018].
13 Bossuyt V, Provenzano E, Symmans WF, et al. Recommendations for standardized 40 Giuliano AE, Connolly JL, Edge SB, et al. Breast cancer—major changes in the
pathological characterization of residual disease for neoadjuvant clinical trials of American Joint Committee on Cancer eighth edition cancer staging manual. CA
breast cancer by the BIG-NABCG collaboration. Ann Oncol 2015;26:1280–91. Cancer J Clin 2017;67:290–303.
14 Kuerer HM, Newman LA, Smith TL, et al. Clinical course of breast cancer patients 41 Ellis IO, Al-Sam S, Anderson N. Pathology reporting of breast disease in surgical
with complete pathologic primary tumor and axillary lymph node response to excision specimens incorporating the dataset for histological reporting of breast
doxorubicin-based neoadjuvant chemotherapy. J Clin Oncol 1999;17:460–9. cancer. London: The Royal College of Pathologists, 2016.
15 Chollet P, Amat S, Cure H, et al. Prognostic significance of a complete pathological 42 Weaver DL. Pathology evaluation of sentinel lymph nodes in breast cancer: protocol
response after induction chemotherapy in operable breast cancer. Br J Cancer recommendations and rationale. Mod Pathol 2010;23(Suppl 2):S26–S32.
2002;86:1041–6. 43 Fan F. Evaluation and reporting of breast cancer after neoadjuvant chemotherapy.
16 Bossuyt V. Processing and reporting of breast specimens in the neoadjuvant setting. Open Pathol J 2009;3:58–63.
Surg Pathol Clin 2018;11:213–30. 44 Schnitt SJ, Collins LC, Effects T. Biopsy interpretation of the breast. 2nd edn.
17 Fisher ER, Wang J, Bryant J, et al. Pathobiology of preoperative chemotherapy: Philadelphia: Lippincott Williams and Wilkins, 2009: 435–9.
findings from the National Surgical Adjuvant Breast and Bowel (NSABP) protocol 45 Choi M, Park YH, Ahn JS, et al. Assessment of pathologic response and long-term
B-18. Cancer 2002;95:681–95. outcome in locally advanced breast cancers after neoadjuvant chemotherapy:
18 Provenzano E, Bossuyt V, Viale G, et al. Standardization of pathologic evaluation comparison of pathologic classification systems. Breast Cancer Res Treat
and reporting of postneoadjuvant specimens in clinical trials of breast 2016;160:475–89.
cancer: recommendations from an international working group. Mod Pathol 46 Sharkey FE, Addington SL, Fowler LJ, et al. Effects of preoperative chemotherapy on
2015;28:1185–201. the morphology of resectable breast carcinoma. Mod Pathol 1996;9:893–900.
19 Provenzano E, Vallier AL, Champ R, et al. A central review of histopathology reports 47 Chen YY, DeVries S, Anderson J, et al. Pathologic and biologic response to
after breast cancer neoadjuvant chemotherapy in the neo-tango trial. Br J Cancer preoperative endocrine therapy in patients with ER-positive ductal carcinoma in situ.
2013;108:866–72. BMC Cancer 2009;9:285.
20 Diéras V, Fumoleau P, Romieu G, et al. Randomized parallel study of doxorubicin 48 Donnelly J, Parham DM, Hickish T, et al. Axillary lymph node scarring and the
plus paclitaxel and doxorubicin plus cyclophosphamide as neoadjuvant treatment of association with tumour response following neoadjuvant chemoendocrine therapy
patients with breast cancer. J Clin Oncol 2004;22:4958–65. for breast cancer. Breast 2001;10:61–6.
21 Braeuning MP, Burke ET, Pisano ED. Embolization coils as tumor markers for 49 Newman LA, Pernick NL, Adsay V, et al. Histopathologic evidence of tumor regression
mammography in patients undergoing neoadjuvant chemotherapy for carcinoma of in the axillary lymph nodes of patients treated with preoperative chemotherapy
the breast. AJR Am J Roentgenol 2000;174:251–2. correlates with breast cancer outcome. Ann Surg Oncol 2003;10:734–9.
22 Park KU, Caudle A. Management of the axilla in the patient with breast cancer. Surg 50 Bossuyt V, Symmans WF. Standardizing of pathology in patients receiving
Clin North Am 2018;98:747–60. neoadjuvant chemotherapy. Ann Surg Oncol 2016;23:3153–61.
23 Carter CL, Allen C, Henson DE. Relation of tumor size, lymph node status, and 51 Feldman S, Krishnamurthy S, Gillanders W, et al. A novel automated assay for the
survival in 24,740 breast cancer cases. Cancer 1989;63:181–7. rapid identification of metastatic breast carcinoma in sentinel lymph nodes. Cancer
24 Beenken SW, Urist MM, Zhang Y, et al. Axillary lymph node status, but not tumor 2011;117:2599–607.
size, predicts locoregional recurrence and overall survival after mastectomy for breast 52 Cortazar P, Zhang L, Untch M, et al. Pathological complete response and long-
cancer. Ann Surg 2003;237:732–9. term clinical benefit in breast cancer: the CTNeoBC pooled analysis. The Lancet
25 AJCC. American Joint Committee on Cancer (AJCC) cancer staging manual. In: 8th 2014;384:164–72.
edn. New York: Springer International Publishing, 2017. 53 Mamounas EP, Anderson SJ, Dignam JJ, et al. Predictors of locoregional recurrence
26 National Comprehensive Cancer Network (NCCN). Clinical Practice Guidelines in after neoadjuvant chemotherapy: results from combined analysis of National Surgical
Oncology: Breast, Version 2, 2017, BINV-11. 2018. Available: http://www.​nccn.​org Adjuvant Breast and Bowel Project B-18 and B-27. J Clin Oncol 2012;30:3960–6.
[Accessed 7 Nov 2018]. 54 Rouzier R, Extra JM, Klijanienko J, et al. Incidence and prognostic significance of
27 Caudle AS, Yang WT, Krishnamurthy S, et al. Improved axillary evaluation following complete axillary downstaging after primary chemotherapy in breast cancer patients
neoadjuvant therapy for patients with node-positive breast cancer using selective with T1 to T3 tumors and cytologically proven axillary metastatic lymph nodes. J Clin
evaluation of clipped nodes: implementation of targeted axillary dissection. J Clin Oncol 2002;20:1304–10.
Oncol 2016;34:1072–8. 55 McCready DR, Hortobagyi GN, Kau SW, et al. The prognostic significance of lymph
28 Rajan R, Poniecka A, Smith TL, et al. Change in tumor cellularity of breast carcinoma node metastases after preoperative chemotherapy for locally advanced breast cancer.
after neoadjuvant chemotherapy as a variable in the pathologic assessment of Arch Surg 1989;124:21–5.
response. Cancer 2004;100:1365–73. 56 Rastogi P, Anderson SJ, Bear HD, et al. Preoperative chemotherapy: updates of
29 Peintinger F, Kuerer HM, McGuire SE, et al. Residual specimen cellularity after National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27. J Clin
neoadjuvant chemotherapy for breast cancer. Br J Surg 2008;95:433–7. Oncol 2008;26:778–85.
30 Chagpar AB, Middleton LP, Sahin AA, et al. Accuracy of physical examination, 57 Buchholz TA, Tucker SL, Masullo L, et al. Predictors of local-regional recurrence
ultrasonography, and mammography in predicting residual pathologic tumor size in after neoadjuvant chemotherapy and mastectomy without radiation. J Clin Oncol
patients treated with neoadjuvant chemotherapy. Ann Surg 2006;243:257–64. 2002;20:17–23.
31 Choi J, Laws A, Hu J, et al. Margins in breast-conserving surgery after neoadjuvant 58 Hennessy BT, Hortobagyi GN, Rouzier R, et al. Outcome after pathologic complete
therapy. Ann Surg Oncol 2018;25:3541–7. eradication of cytologically proven breast cancer axillary node metastases following
32 Boughey JC, Alvarado MD, Lancaster RB, et al. Surgical standards for management primary chemotherapy. J Clin Oncol 2005;23:9304–11.
of the axilla in breast cancer clinical trials with pathological complete response 59 Klauber-DeMore N, Ollila DW, Moore DT, et al. Size of residual lymph node
endpoint. NPJ Breast Cancer 2018;4:26. metastasis after neoadjuvant chemotherapy in locally advanced breast cancer
33 Pilewskie M, Morrow M. Axillary nodal management following neoadjuvant patients is prognostic. Ann Surg Oncol 2006;13:685–91.
chemotherapy: a review. JAMA Oncol 2017;3:549–55. 60 Corben AD, Abi-Raad R, Popa I, et al. Pathologic response and long-term follow-up
34 Nguyen TT, Hoskin TL, Day CN, et al. Decreasing use of axillary dissection in node- in breast cancer patients treated with neoadjuvant chemotherapy: a comparison
positive breast cancer patients treated with neoadjuvant chemotherapy. Ann Surg between classifications and their practical application. Arch Pathol Lab Med
Oncol 2018;25:2596–602. 2013;137:1074–82.
35 Boughey JC, Suman VJ, Mittendorf EA, et al. Sentinel lymph node surgery after 61 Mazouni C, Peintinger F, Wan-Kau S, et al. Residual ductal carcinoma in situ in
neoadjuvant chemotherapy in patients with node-positive breast cancer: the patients with complete eradication of invasive breast cancer after neoadjuvant
ACOSOG Z1071 (Alliance) clinical trial. JAMA 2013;310:1455–61. chemotherapy does not adversely affect patient outcome. J Clin Oncol
36 Boileau JF, Poirier B, Basik M, et al. Sentinel node biopsy after neoadjuvant 2007;25:2650–5.
chemotherapy in biopsy-proven node-positive breast cancer: the SN FNAC study. J 62 Campbell JI, Yau C, Krass P, et al. Comparison of residual cancer burden, American
Clin Oncol 2015;33:258–64. Joint Committee on Cancer staging and pathologic complete response in breast
37 Kuehn T, Bauerfeind I, Fehm T, et al. Sentinel-lymph-node biopsy in patients with cancer after neoadjuvant chemotherapy: results from the I-SPY 1 TRIAL (CALGB
breast cancer before and after neoadjuvant chemotherapy (SENTINA): a prospective, 150007/150012; ACRIN 6657). Breast Cancer Res Treat 2017;165:181–91.
multicentre cohort study. Lancet Oncol 2013;14:609–18. 63 von Minckwitz G, Untch M, Blohmer JU, et al. Definition and impact of pathologic
38 Symmans WF, Peintinger F, Hatzis C, et al. Measurement of residual breast complete response on prognosis after neoadjuvant chemotherapy in various intrinsic
cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol breast cancer subtypes. J Clin Oncol 2012;30:1796–804.
2007;25:4414–22. 64 von Minckwitz G, Untch M, Nüesch E, et al. Impact of treatment characteristics on
39 MD Anderson Cancer Center. [Residual Cancer Burden calculator and associated response of different breast cancer phenotypes: pooled analysis of the German neo-
documents [guide for measuring cancer cellularity, examples of gross & microscopic adjuvant chemotherapy trials. Breast Cancer Res Treat 2011;125:145–56.

Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598 131


Review
65 Esserman LJ, Berry DA, DeMichele A, et al. Pathologic complete response predicts 83 Rodenhuis S, Mandjes IA, Wesseling J, et al. A simple system for grading

J Clin Pathol: first published as 10.1136/jclinpath-2018-205598 on 22 January 2019. Downloaded from http://jcp.bmj.com/ on 23 January 2019 by guest. Protected by copyright.
recurrence-free survival more effectively by cancer subset: results from the I-SPY 1 the response of breast cancer to neoadjuvant chemotherapy. Ann Oncol
TRIAL—CALGB 150007/150012, ACRIN 6657. J Clin Oncol 2012;30:3242–9. 2010;21:481–7.
66 Houssami N, Macaskill P, von Minckwitz G, et al. Meta-analysis of the association 84 Mittendorf EA, Jeruss JS, Tucker SL, et al. Validation of a novel staging system for
of breast cancer subtype and pathologic complete response to neoadjuvant disease-specific survival in patients with breast cancer treated with neoadjuvant
chemotherapy. Eur J Cancer 2012;48:3342–54. chemotherapy. J Clin Oncol 2011;29:1956–62.
67 Fayanju OM, Ren Y, Thomas SM, et al. The clinical significance of breast-only and 85 Mittendorf EA, Vila J, Tucker SL, et al. The Neo-Bioscore update for staging breast
node-only pathologic complete response (pCR) after neoadjuvant chemotherapy cancer treated with neoadjuvant chemotherapy: incorporation of prognostic biologic
(NACT): a review of 20,000 breast cancer patients in the National Cancer Data Base factors into staging after treatment. JAMA Oncol 2016;2:929–36.
(NCDB). Ann Surg 2018;268:591–601. 86 Ellis MJ, Tao Y, Luo J, et al. Outcome prediction for estrogen receptor-positive breast
68 Bonadonna G, Veronesi U, Brambilla C, et al. Primary chemotherapy to avoid cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl
mastectomy in tumors with diameters of three centimeters or more. J Natl Cancer Cancer Inst 2008;100:1380–8.
Inst 1990;82:1539–45. 87 Jones RL, Salter J, A’Hern R, A’Hern R, et al. The prognostic significance of Ki67
69 Ogston KN, Miller ID, Payne S, et al. A new histological grading system to assess before and after neoadjuvant chemotherapy in breast cancer. Breast Cancer Res
response of breast cancers to primary chemotherapy: prognostic significance and Treat 2009;116:53–68.
survival. Breast 2003;12:320–7. 88 von Minckwitz G, Schmitt WD, Loibl S, et al. Ki67 measured after neoadjuvant
70 Smith IC, Heys SD, Hutcheon AW, et al. Neoadjuvant chemotherapy in breast cancer: chemotherapy for primary breast cancer. Clin Cancer Res 2013;19:4521–31.
significantly enhanced response with docetaxel. J Clin Oncol 2002;20:1456–66. 89 Yardley DA, Peacock NW, Shastry M, et al. A phase II trial of ixabepilone and
71 Sinn HP, Helmchen B, Wittekind CH. [TNM classification of breast cancer: changes cyclophosphamide as neoadjuvant therapy for patients with HER2-negative breast
and comments on the 7th edition]. Pathologe 2010;31:361–6. cancer: correlation of pathologic complete response with the 21-gene recurrence
72 Peintinger F, Sinn B, Hatzis C, et al. Reproducibility of residual cancer burden for score. Breast Cancer Res Treat 2015;154:299–308.
prognostic assessment of breast cancer after neoadjuvant chemotherapy. Mod 90 Siow ZR, De Boer RH, Lindeman GJ, et al. Spotlight on the utility of the Oncotype
Pathol 2015;28:913–20. DX® breast cancer assay. Int J Womens Health 2018;10:89–100.
73 Symmans WF, Wei C, Gould R, et al. Long-term prognostic risk after neoadjuvant 91 Chen S, Chen CM, Yu KD, et al. Prognostic value of a positive-to-negative change in
chemotherapy associated with residual cancer burden and breast cancer subtype. J hormone receptor status after neoadjuvant chemotherapy in patients with hormone
receptor-positive breast cancer. Ann Surg Oncol 2012;19:3002–11.
Clin Oncol 2017;35:1049–60.
92 Hirata T, Shimizu C, Yonemori K, et al. Change in the hormone receptor status
74 Romero A, García-Sáenz JA, Fuentes-Ferrer M, et al. Correlation between response
following administration of neoadjuvant chemotherapy and its impact on
to neoadjuvant chemotherapy and survival in locally advanced breast cancer
the long-term outcome in patients with primary breast cancer. Br J Cancer
patients. Ann Oncol 2013;24:655–61.
2009;101:1529–36.
75 Sheri A, Smith IE, Johnston SR, et al. Residual proliferative cancer burden to
93 Arens N, Bleyl U, Hildenbrand R. HER2/neu, p53, Ki67, and hormone receptors
predict long-term outcome following neoadjuvant chemotherapy. Ann Oncol
do not change during neoadjuvant chemotherapy in breast cancer. Virchows Arch
2015;26:75–80.
2005;446:489–96.
76 Lee HJ, Park IA, Song IH, et al. Comparison of pathologic response evaluation
94 Jabbour MN, Massad CY, Boulos FI. Variability in hormone and growth factor
systems after anthracycline with/without taxane-based neoadjuvant
receptor expression in primary versus recurrent, metastatic, and post-neoadjuvant
chemotherapy among different subtypes of breast cancers. PLoS One
breast carcinoma. Breast Cancer Res Treat 2012;135:29–37.
2015;10:e0137885.
95 Zhang N, Moran MS, Huo Q, et al. The hormonal receptor status in breast cancer
77 Dieci MV, Radosevic-Robin N, Fineberg S, et al. Update on tumor-infiltrating can be altered by neoadjuvant chemotherapy: a meta-analysis. Cancer Invest
lymphocytes (TILs) in breast cancer, including recommendations to assess TILs in 2011;29:594–8.
residual disease after neoadjuvant therapy and in carcinoma in situ: a report of the 96 Dowsett M, Ebbs SR, Dixon JM, et al. Biomarker changes during neoadjuvant
International Immuno-Oncology Biomarker Working Group on Breast Cancer. Semin anastrozole, tamoxifen, or the combination: influence of hormonal status and HER-2
Cancer Biol 2018;52(Pt 2):16–25. in breast cancer--a study from the IMPACT trialists. J Clin Oncol 2005;23:2477–92.
78 Hwang HW, Jung H, Hyeon J, et al. A nomogram to predict pathologic complete 97 Mittendorf EA, Wu Y, Scaltriti M, et al. Loss of HER2 amplification following
response (pCR) and the value of tumor-infiltrating lymphocytes (TILs) for prediction trastuzumab-based neoadjuvant systemic therapy and survival outcomes. Clin Cancer
of response to neoadjuvant chemotherapy (NAC) in breast cancer patients. Breast Res 2009;15:7381–8.
Cancer Res Treat 2018;27. 98 Feldman LD, Hortobagyi GN, Buzdar AU, et al. Pathological assessment of response
79 Dieci MV, Criscitiello C, Goubar A, et al. Prognostic value of tumor-infiltrating to induction chemotherapy in breast cancer. Cancer Res 1986;46:2578–81.
lymphocytes on residual disease after primary chemotherapy for triple-negative 99 Sinn HP, Schmid H, Junkermann H, et al. [Histologic regression of breast cancer after
breast cancer: a retrospective multicenter study. Ann Oncol 2014;25:611–8. primary (neoadjuvant) chemotherapy]. Geburtshilfe Frauenheilkd 1994;54:552–8.
80 Loi S, Dushyanthen S, Beavis PA, et al. RAS/MAPK activation is associated with 100 Sataloff DM, Mason BA, Prestipino AJ, et al. Pathologic response to induction
reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic chemotherapy in locally advanced carcinoma of the breast: a determinant of
cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin outcome. J Am Coll Surg 1995;180:297–306.
Cancer Res 2016;22:1499–509. 101 Honkoop AH, van Diest PJ, de Jong JS, et al. Prognostic role of clinical, pathological
81 Galea MH, Blamey RW, Elston CE, et al. The Nottingham Prognostic Index in primary and biological characteristics in patients with locally advanced breast cancer. Br J
breast cancer. Breast Cancer Res Treat 1992;22:207–19. Cancer 1998;77:621–6.
82 Chollet P, Abrial C, Durando X, et al. A new prognostic classification after primary 102 Kuerer HM, Newman LA, Buzdar AU, et al. Pathologic tumor response in the breast
chemotherapy for breast cancer: residual disease in breast and nodes (RDBN). following neoadjuvant chemotherapy predicts axillary lymph node status. Cancer J
Cancer J 2008;14:128–32. Sci Am 1998;4:230–6.

132 Mrkonjic M, et al. J Clin Pathol 2019;72:120–132. doi:10.1136/jclinpath-2018-205598

You might also like