Skip to main content

Deep learning-based risk stratification of preoperative breast biopsies using digital whole slide images

Abstract

Background

Nottingham histological grade (NHG) is a well established prognostic factor in breast cancer histopathology but has a high inter-assessor variability with many tumours being classified as intermediate grade, NHG2. Here, we evaluate if DeepGrade, a previously developed model for risk stratification of resected tumour specimens, could be applied to risk-stratify tumour biopsy specimens.

Methods

A total of 11,955,755 tiles from 1169 whole slide images of preoperative biopsies from 896 patients diagnosed with breast cancer in Stockholm, Sweden, were included. DeepGrade, a deep convolutional neural network model, was applied for the prediction of low- and high-risk tumours. It was evaluated against clinically assigned grades NHG1 and NHG3 on the biopsy specimen but also against the grades assigned to the corresponding resection specimen using area under the operating curve (AUC). The prognostic value of the DeepGrade model in the biopsy setting was evaluated using time-to-event analysis.

Results

Based on preoperative biopsy images, the DeepGrade model predicted resected tumour cases of clinical grades NHG1 and NHG3 with an AUC of 0.908 (95% CI: 0.88; 0.93). Furthermore, out of the 432 resected clinically-assigned NHG2 tumours, 281 (65%) were classified as DeepGrade-low and 151 (35%) as DeepGrade-high. Using a multivariable Cox proportional hazards model the hazard ratio between DeepGrade low- and high-risk groups was estimated as 2.01 (95% CI: 1.06; 3.79).

Conclusions

DeepGrade provided prediction of tumour grades NHG1 and NHG3 on the resection specimen using only the biopsy specimen. The results demonstrate that the DeepGrade model can provide decision support to identify high-risk tumours based on preoperative biopsies, thus improving early treatment decisions.

Background

Breast cancer is currently the most common cancer type globally [1]. In the majority of cases, suspicious breast lesions are initially identified by mammography screening, which is recommended in most developed countries for early detection of breast cancer [2, 3]. For women with a suspicious lesion, a preoperative core needle biopsy is performed to histologically assess the breast tissue [4]. Evaluation of the biopsy by pathologists is key to diagnose breast cancer, where morphological information and biomarker analysis are paramount to guide further surgical and oncological therapy decisions [5].

Tumour grading is a cornerstone in the histopathological assessment of breast cancer, not only in the resected tumour specimen but it is also of importance in the biopsy specimen [6]. Histological grade reflects the degree of differentiation of a tumour by comparing the similarity of malignant cells to that of normal breast terminal duct lobular units [7]. Currently, the most commonly used grading method is the Nottingham Histological Grade (NHG) adapted by Elston-Ellis following work from Bloom-Richardson [8, 9]. Histological grading relies on the performance and expertise of pathologists, and evaluates three morphological features: the degree of tubular formation (gland architecture), nuclear pleomorphism (nucleus size and shape) and the mitotic count [9]. Each of these three morphological features is given a score from 1 to 3 by the pathologist and are then combined to obtain the final NHG grade on a score from 1 to 3. Histological grade is an important prognostic feature of breast cancer, with NHG1 having a good prognosis and NHG3 tumours being associated with poor prognosis, independently of the morphological subtype and nodal status [10,11,12]. Therefore, tumour grade plays an important role in guiding treatment decisions [13]. However, about 50% of all resected breast cancer specimens are diagnosed as NHG2, which has limited clinical value for treatment decisions [14,15,16]. One of the major challenges with histological grading is that it relies on the experience, expertise and interpretation of the pathologist, and high inter-observer and inter-laboratory variabilities are well described [7, 14].

Histological grade assessment is even more complicated in biopsy specimens with very limited tumour material and frequent tissue artefacts [14]. This causes significant discrepancies between the biopsy grading and the histological grade assigned on the surgically resected specimen [17]. These uncertainties are accompanied by the fact that a greater number of biopsy samples are not even assigned a grade, and that up to 70% of the biopsy samples are assigned the intermediate grade, NHG2 [15, 18, 19].

The recent advances in computational pathology based on the availability of large amounts of digitised whole-slide histopathological images, as well as the development of novel artificial intelligence technologies, has enabled model-based grading of tumours in resected specimens [20,21,22]. Wang et al. have also shown by developing the DeepGrade model using resected NHG1 and NHG3 tumours that this technology enabled further risk stratification of intermediate-risk NHG2 patients into two risk subgroups with independent prognostic value [23]. Risk stratification is particularly relevant in patients with oestrogen receptor (ER)-positive/human epidermal growth factor receptor 2 (HER2)-negative tumours, since high-risk (NHG3) patients will typically be provided chemotherapy in addition to endocrine therapy, and low-risk (NHG1) patients would be spared chemotherapy in order to avoid overtreatment, whereas the intermediate NHG2 group is uninformative and has limited clinical value for treatment decisions.

In this study, we aim to assess if the DeepGrade model [23], developed using resected tumour specimens, could be applied to risk-stratify tumours using only the biopsy specimens. This would allow earlier identification of high-risk tumours from the initial biopsy specimens, and further improve information that can be used in the treatment planning at the preoperative stage.

Methods

Patients

This retrospective study included female patients who underwent a breast biopsy at the Stockholm South General Hospital in Stockholm, Sweden between June 2012 and May 2018. Patients diagnosed with invasive breast cancer as their primary diagnosis and who had undergone a surgical removal of their tumour within two months following their biopsy without receiving neoadjuvant therapy were included in the study. See Fig. 1 for detailed explanation of the selection criteria. A total of 1169 whole slide images (WSI) from 896 patients were included in the final analyses. The WSI from the resected tumour specimens of 801 of these patients were also available and used for comparison of the prediction of DeepGrade risk group on this material. Clinical data was retrieved retrospectively from the Swedish National Breast Cancer (NKBC) Registry as well as from the patient’s pathology reports when data on the NHG status was not available in the registry. The NKBC registry includes data from newly diagnosed patients with primary in-situ or invasive tumours in Sweden and covers both a full pathology report as well as survival based on follow-up routines [24]. The patients’ NHG were assessed as part of routine clinical care, and separately for the biopsy and resected tumour specimens. This study was reviewed and approved by the Swedish Ethical Review Authority.

Fig. 1
figure 1

CONSORT diagram. The data used contained whole slide images (WSI) of biopsies for 896 patients who had a resected tumour Nottingham Histological Grade (NHG) and who did not receive neoadjuvant chemotherapy. Out of these, a total of 666 patients had a biopsy NHG, 801 patients had a matching resected tumour slide, on which DeepGrade predictions could also be performed, and, survival data was available for 725 patients. A total of 525 patients (682 WSI) are in all three subanalyses

WSI and deep learning model

For each patient between one and seven haematoxylin and eosin (H&E) stained formalin-fixed paraffin-embedded (FFPE) histopathology slides of biopsy specimens were digitised in-house using either Hamamatsu Nanozoomer XR or Hamamatsu Nanozoomer S360 scanners (Hamamatsu Photonics K.K., Shizuoka, Japan) at 40X magnification (0.227 μm/pixel and 0.230 μm/pixel, respectively). Methodology for pre-processing of the WSI was performed according to the methodology previously described [23]. Initially, tissue segmentation was performed by transforming lower-level representations extracted from the WSI’s resolution pyramids obtained using OpenSlide [25]. These were then transformed from RGB to HSV colour space. Two masks were then generated for each slide, one for filtering out hue values lower than 0.75, the other adding a maximum value of 25 to the Otsu’s threshold [26] in order to remove non-tissue areas while reducing the removal of the tissue regions due to the high threshold value on the transformed saturation channel in some cases.

WSI regions included in the tissue mask were tiled into image tiles of 598 × 598 pixels with a down-sampled resolution equivalent to 20 × (271 μm x 271 μm). Due to the small tissue area in biopsy specimens, tiling was performed with 75% overlap on both the vertical and horizontal axes between two consecutive tiles. Next, in order to ensure quality of the data, remove unsharp tiles, any remaining tiles with background, those with adipose tissue, and blurred tiles were all excluded by measuring a variance of the Laplacian filter and excluding the tiles with a value lower than 500 [23]. Lastly, to address the stain variabilities in WSI, colour normalisation across each WSI was performed using the method described by Macenko et al [27], and as implemented by Wang et al [23]. Colour normalisation was applied with the same factor to all tiles within a WSI. Using reference stain vectors [28] and slide level stain vectors obtained using 100 randomly selected tiles per slide, colour normalisation could be applied to each tile towards the reference stain vectors. For the 801 patients with preoperative biopsies and a matching resected tumour WSI, a similar pre-processing method was performed for the WSI pre-processing with two significant changes. First, no overlap between two consecutive tiles was considered. Secondly, after the colour normalisation step, a tumour segmentation model previously developed [23] was applied to include only the tiles from the invasive cancer regions in the resected specimens for further downstream analysis. After pre-processing, a total of 11,955,755 tiles were used for predictions from biopsy specimens, and 1,157,871 were used from the surgical resection specimens.

Histological grade prediction

Prediction of low- (NHG1) and high- (NHG3) risk tumours on the biopsy WSI was performed using an ensemble of 20 convolutional neural network (CNN) models previously developed as the DeepGrade model [23]. The DeepGrade models were trained to classify NHG1 and NHG3 tumours in WSI from resected specimens. Each model uses Inception V3 model pre-trained with ImageNet [29] as the base model. The Inception V3 model consists of a stem block constituted of four 3 × 3 convolutional layers and one 1 × 1 convolutional layer as well as two max pooling layers. It then employs three inception blocks (A, B and C) each consisting of 1 × 1, 3 × 3 and 5 × 5 convolutional filters together with regularization. Inception blocks A and B also include an average pooling layer while block C includes a max pooling layer. Block A is followed by a reduction block that includes further convolutional layers and one max pooling layer while block B employs 3 × 3 convolutions with strides to downsample the feature maps. Finally, there is an auxiliary classifier block to prevent vanishing gradient. A fully connected layer of 1024 hidden units and Rectified Linear Unit (RELU) activation function were added before the final layer. Stochastic gradient descent was used to update parameters from all layers with an adaptive learning rate starting from 10− 3 but reduced by 50% each time the model performance stopped improving for 10 epochs. Cross-entropy loss was used for the binary outcome NHG1 versus NHG3. The initial 20 CNN models were trained on 844 WSI, of which 173 patients’ biopsy WSI were also included in this study. Each model in DeepGrade outputs the two class prediction probabilities for each tile (P(NHG3|tilei) and P(NHG1|tilei)). The P(NHG3|tilei) class probability from each of the 20 models in the ensemble were averaged to provide the tile-level prediction. In order to obtain the patient-level predictions, all the tile-level predictions of all the WSI from each patient and the upper-percentile (99%) of the tile level predictions were considered. Regarding resected tumour specimens, as tumour detection was previously performed, a lower threshold was used with the upper-quartile (75%) of the tile level predictions being considered. For NHG1 and NHG3, prediction performance of the DeepGrade model was evaluated against clinically assigned NHG by pathologists on both the biopsy specimen (biopsy NHG) and on the surgically resected specimen (resected tumour NHG). The prediction performances on the patient levels were measured using the receiver operating characteristic (ROC) curves and the linked area under the curve (AUC) using R package pROC [30]. The most optimal threshold for binary assignment into low- and high-risk groups was then determined using the Youden’s J statistic [31] compared to the resected tumour grade. A separate threshold was calculated for DeepGrade predictions on resected tumour specimens. Agreement between the assigned NHG (from the biopsy specimen or the resected tumour specimen) and the obtained DeepGrade risk group was measured using Cohen’s kappa and the following interpretations: 0-0.20: slight agreement, 0.21–0.40: fair agreement, 0.41–0.60: moderate agreement, 0.61–0.80: substantial agreement and 0.81-1.00: almost perfect agreement [32, 33]. Sensitivity or recall was measured as the probability of DeepGrade-high when the patient had NHG3, while specificity was measured as the probability of DeepGrade-low when the patient had NHG1. Furthermore, the DeepGrade model was applied on NHG2 tumours to sub-stratify the tumours into two groups: low- and high-risk groups. The classification performance of the DeepGrade on the biopsy WSI was also compared to the classification performed on the resection WSI.

Survival analyses

Finally, the rates of recurrence-free survival (RFS) as defined by the presence of a locoregional or distant recurrence or death were compared between patients who were assigned in the DeepGrade-high and DeepGrade-low groups. The time-to-event was defined as the number of days between the date of initial diagnosis and either date of recurrence or loss of follow-up. The R packages ‘survival’ and ‘survminer’ were used to visualise the survival outcomes between groups, and the ‘forestmodel’ package was used to estimate adjusted hazard ratios (HRs) using multivariate Cox proportional hazards regression models. The other risk factor used in the model was age, considered as the only factor available at time of biopsy. Further sub-analyses were performed on oestrogen receptor (ER)-positive and human epidermal growth factor receptor 2 (HER2)-negative cases as determined by immunohistochemical and/or in situ hybridisation staining and available in the pathology report.

Results

Agreement between clinical grades on biopsy and resected specimens

First, we assessed the discrepancies between the clinical assignment of NHG on the biopsy and subsequent resected specimens (Fig. 2). A quarter of the patients did not have a NHG assigned on the biopsy specimen in clinical routine, and 72% of the patients who had a biopsy NHG available were of NHG2. The overall agreement between the clinical grade assignments on the biopsy and on the resected tumour specimen was 65.5% when including patients for whom we had both diagnoses. When considering only cases that had a resected tumour NHG1 or NHG3, less than a third (148 out of 463 cases) also had a NHG1 or NHG3 in the biopsy specimen, the rest being assigned NHG2 or not having a grade at all. We observed a fair agreement with the Cohen’s kappa value of 0.40 (95% CIs: 0.34;0.46) between specimen types.

Fig. 2
figure 2

Comparison between clinical Notthingham Histological Grade (NHG) assigned by pathologists on the biopsy and surgically resected specimens. A. confusion matrix, B. Sankey plot, diagnoses were in agreement when the same NHG was assigned to the biopsy specimen and to the resected tumour specimen

Assessment of the DeepGrade classification performance on the biopsy specimen

We evaluated the risk classification performance of the DeepGrade model on the biopsy specimens. We observed an AUC score of 0.962 (95% CI: 0.934; 0.991) for DeepGrade predictions compared to biopsy NHG1 and NHG3 (Fig. 3A). For 168 out of 186 patients (90.3%) with biopsy NHG1 or NHG3 (Fig. 3B-C) the DeepGrade model and the pathologists were in agreement, representing an almost perfect agreement with a kappa value of 0.81. Sensitivity was of 91.7% while specificity was of 89.1%. Out of all 896 patients, only 0.8% (7) biopsy NHG3 tumours were assigned to the DeepGrade low-risk group (Fig. 3C-D). Out of the 230 patients without a biopsy NHG, 135 (58.7%) were classified in the DeepGrade low-risk group and 95 (41.3%) were classified in the high-risk group (Fig. 3C-D). In the ER-positive/HER2-negative subgroups, the observed AUC score was of 0.949 (95% CI: 0.901; 0.996) (Supplementary Fig. 1A-B).

Fig. 3
figure 3

DeepGrade prediction results obtained on biopsy specimens compared to the clinical biopsy Nottingham Histological Grade (NHG) assigned by pathologists. A. Receiver Operating Curve (ROC) of the patient-level predictions obtained by the DeepGrade model compared to biopsy NHG1 and NHG3. B. Sankey plot of the biopsy NHG compared to the obtained DeepGrade risk group for the 186 patients who had a biopsy NHG1 or NHG3. C. Confusion matrix for all 896 patients comparing biopsy NHG and predicted DeepGrade risk group. D. Sankey plot for all patients. Diagnoses were in agreement when patients were DeepGrade-low and NHG1 or DeepGrade-high and NHG3. Patients with biopsy NHG2 or with no biopsy NHG were stratified as either low-risk or high-risk

Assessment of the DeepGrade classification performance compared to the clinical grade on the resected tumour specimen

To test our hypothesis that not only can the DeepGrade model predict the NHG of the biopsy, but also predict the clinically assigned NHG1 and NHG3 grades assigned on the resected specimens, we compared the prediction results obtained (Fig. 4). An example of the DeepGrade prediction results on a biopsy WSI is illustrated in Fig. 4A. We observed an AUC score of 0.908 (95% CI: 0.882; 0.934) when comparing the DeepGrade model prediction obtained on the biopsy versus the clinically assigned NHG1 and NHG3 on the resected tumour (Fig. 4B). Agreement between the risk-group predictions obtained using the biopsy and that of the pathologist for resected specimens with NHG1 and NHG3 was observed for 382 out of 464 patients (82.3%) and the kappa value was 0.65 indicating substantial agreement (Fig. 4C). Sensitivity was of 78.8% and specificity was of 86.8%. When looking at the patients who were in the DeepGrade-low risk group, but who had a resected tumour of NHG3, the clinical biopsy grade was either NHG2 or not graded in 91% of the cases, and only five patients had NHG3 on both biopsy and resected tumour specimen (Fig. 4D). Out of the 432 patients who had a resected tumour with NHG2, 281 (65.0%) were assigned to the DeepGrade-low risk group while 151 (35.0%) were assigned to the DeepGrade-high risk group from biopsies (Fig. 4D). In the ER-positive/HER2-negative subgroup the obtained AUC was of 0.881 (95% CI: 0.846–0.917) (Supplementary Fig. 1C-D). The sensitivity in this subgroup was of 81.3% and the specificity was of 80.6%.

Fig. 4
figure 4

DeepGrade prediction results obtained on biopsy specimens compared to the clinical Nottingham Histological Grade (NHG) assigned by the pathologist on the resected specimen. A. Example of a whole slide image with prediction results. Red is more likely to be predicted as high risk, or in other words the predicted probability for each tile to be classified as high-risk. The patient is classified DeepGrade high as the upper percentile of the mean values across all tiles was over the obtained threshold of 0.83. B. Receiver Operating Curve (ROC) of the patient-level DeepGrade model prediction versus the resected tumour grades NHG1 and NHG3 assigned by a pathologist. C. Sankey plot of the proportion of patients predicted with DeepGrade-high and -low versus the resected tumour grade NHG1 and NHG3. D. Sankey plot with results of all biopsy specimen comparing the obtained DeepGrade risk group with both the biopsy NHG and the resected tumour NHG. Diagnoses were in agreement when patients were low-risk biopsy DeepGrade and resected tumour NHG1 or high-risk biopsy DeepGrade and resected tumour NHG3. Patients with resected tumour NHG2 were stratified as either low-risk or high-risk

Comparison between DeepGrade risk groups on biopsy and resected tumour specimens

To verify whether the results obtained on the biopsy specimens were in line with those obtained on the resected tumour specimen, we compared the DeepGrade risk group for 801 patients for which we had both specimens available (Fig. 5). Almost three quarters of the patients were assigned the same DeepGrade risk group on the biopsy and resected specimens. This proportion was even higher when considering only patients with a resected tumour of NHG1 or NHG3, with 80.8% of the patients assigned the same DeepGrade risk group.

Fig. 5
figure 5

Comparison of DeepGrade risk groups on biopsy and resected specimens. A. Confusion matrix for all Nottingham Histological Grade (NHG) grades combined. B. Confusion matrix for only resected grades NHG1 and NHG3

Prognostic performance of DeepGrade on tumour biopsies

The prognostic performance of the DeepGrade model on biopsy specimens was measured based on recurrence-free survival and was visualised using Kaplan-Meier curves. The independent prognostic value was measured using multivariable Cox proportional hazards model adjusting for age, resembling information available at the biopsy stage. When including all patients, the biopsy DeepGrade model was found to be a predictor of recurrence-free survival with an estimated hazard ratio of 2.01 (p = 0.033, 95% CI: 1.06; 3.79) for patients in the DeepGrade-high group compared to those in the DeepGrade-low group, independently of the patient’s age (Fig. 6A-B). Subgroup analyses on patients with ER+/HER2- tumours also showed that DeepGrade was a predictor of recurrence-free survival with an estimated hazard ratio of 2.12 (p = 0.044, 95% CI: 1.02; 4.43) for patients in the DeepGrade-high group compared to those in the DeepGrade-low group, independently of the patient’s age (Fig. 6C-D).

Fig. 6
figure 6

Recurrence-free survival outcomes for breast cancer patients by DeepGrade risk-group obtained on the biopsy specimen. A. Kaplan-Meier curves for patients stratified by biopsy DeepGrade-low and -high risk groups. The high-risk group had the worst prognosis. B. Forest plot from multivariable Cox proportional hazard regression including the biopsy DeepGrade risk groups and age at diagnosis. Three patients had missing data for age. C. Kaplan-Meier curves for ER-positive/HER2-negative patients stratified by biopsy DeepGrade-low and -high risk groups. D. Forest plot from multivariable Cox proportional hazard regression for ER-positive/HER2-negative patients including the biopsy DeepGrade risk groups and age at diagnosis

Discussion

The aim of this study was to investigate if the DeepGrade model, previously developed to risk-stratify patients based on resected tumour specimens, could also be used to risk-stratify biopsy specimens. We observed a high classification performance when comparing the DeepGrade predictions on the biopsy specimen to the pathologist-assigned biopsy NHG. Most interestingly, the DeepGrade model could predict the histological grade of the resected tumour specimen while analysing only biopsy material. Furthermore, classification of patients using the DeepGrade model was predictive of recurrence-free survival at point of biopsy.

Neoadjuvant therapy is currently recommended to most HER2-positive and triple negative breast cancers, of which the vast majority are high-grade tumours. The identification of patients with high-grade tumours at the time of biopsy is essential for the decision to treat a patient with neoadjuvant chemotherapy [7, 34], and especially within the larger ER-positive, HER2-negative subgroup [7]. However, conventional histological grading of biopsies by pathologists remains challenging and most biopsies are assigned the intermediate NHG2, or are not graded at all [14, 18]. This lack of precision in biopsy grading leads to a discrepancy between pathologists, and in one cohort up to 45% of women had a change in diagnosis between the biopsy and the resected tumour [18]. We found that 41% of patients who were not assigned a grade on the biopsy were assigned to the high-risk group by DeepGrade, of which 54% were actually assigned as NHG3 by pathologists on their resected tumour specimen. Earlier diagnosis could assist with earlier treatment decisions.

Several studies have developed models to predict grade using deep learning models on WSI from resected tumour specimens but not using core needle biopsies [20,21,22,23, 35, 36]. In particular, Wang et al. obtained an AUC of 0.907 for DeepGrade in their external data regarding resected tumours which is in line with the accuracy we obtained on the biopsy specimen when comparing to resected tumours of NHG1 versus NHG3 (0.908) [23]. Others who have predicted grade into two groups (low-grade and high-grade) on resected tumour specimens obtained agreements around 80%, and kappa values between 0.59 and 0.64 [35, 37]. Despite predicting the resected specimen grade using only biopsy material, we achieved high performance results among NHG1 and NHG3 tumours with an agreement of 82% and a kappa value of 0.65 between biopsy DeepGrade risk groups and pathologist-assigned NHG on resected tumours. As a comparison, only 32% of the 463 cases who were NHG1 or NHG3 on the resected specimen were also assigned NHG1 or NHG3 on the biopsy specimen by a pathologist. In the literature, agreement between biopsy and resected tumour NHG by pathologists for all three grades is usually around 75% and ranges from 59–91% [38]. The results presented were obtained without performing prior tumour predictions as the biopsy material is smaller and the presence of benign tissue should not influence significantly the presence of high risk morphological patterns that are identified by the DeepGrade model.

The use of biopsy specimens in computational pathology within breast cancer is relatively rare in the literature, as opposed to the work performed in prostate cancer [39,40,41]. A number of studies focused on the identification of tumour areas [42,43,44,45], while others aimed to predict the response to neoadjuvant therapy, in part using grade as their training material [46, 47]. The DeepGrade model extracts histological grade-related morphological information from images using deep CNN models. To date no risk stratification methods for survival prediction have been proposed using biopsy material, however different approaches have been suggested related to grade [20], grading sub-components [22, 48], and intra-tumour heterogeneity [49] using resected tumour specimens only. The proposed methodology in this study could be used as a decision support tool to complement pathologists and treating physicians, as it establishes a risk assessment of all tumours, including those that are hard to grade. It also has the benefit of providing a solution that is less costly and with shorter waiting times, both for the patient and for the healthcare providers than other methods used for risk stratification such as Oncotype DX (Exact Sciences Corp., Madison, WI, USA) or Prosigna (Veracyte Inc., South San Francisco, CA, USA) gene expression assays [50, 51]. Both have been developed for patient risk stratification and treatment decisions on the resected tumour specimens, but have also been applied outside of their intended use, for assessment of core needle biopsy specimens with conclusive results [52,53,54,55].

This is the first study demonstrating risk stratification of NHG2 tumours already at the time of biopsy using deep learning. Although several methods are available and implemented in clinical routine to risk stratify patients, most use gene expression profiling assays [52,53,54,55], which are time-consuming methods and remain costly [56]. The risk stratification method presented in this study has the advantages of providing a result to the pathologist in a short time-frame and at a very low cost given most pathology laboratories in high-income countries already use digitised WSI to some extent in routine diagnostics [23].

Limitations of this study include the fact that the study was based on retrospective material in order to obtain a large enough sample size when only including one hospital. Nonetheless, the small number of recurrence events leads to low-powered survival analyses. Even though most pathologists would not have direct access to the biopsy grade when assigning a grade to the resected specimen, it was possible for them to look into the patient’s electronic record and to make a decision based on the previously assigned grade. The discrepancies in diagnoses observed here as well as in previous work would however point into the direction that the two grades are given independently. Furthermore, a limitation of the present study is that a subset of this study, 173 patients (19.3%) were included as training data of the initial DeepGrade model [23]. However, the biopsy material itself was never used for the training of the original DeepGrade model representing in itself a fully independent set from the original data, and results presented in Supplementary Fig. 2 show that performance remains high. In the future, further analyses on patients from another hospital would be beneficial to confirm the results obtained.

Conclusions

In conclusion, we found that the resected tumour grade could be predicted by DeepGrade based on using only biopsy specimens. With relatively simple implementation, high-risk tumours could therefore be identified at the preoperative stage. Like in resected tumours, DeepGrade could also stratify NHG2 tumours on biopsy specimens into low- and high-risk groups. In the future, this could provide decision support to pathologists as well as treating physicians to improve the quality of relevant information for clinical decisions earlier on in the process, and thus potentially reduce both over- and under-treatment of patients in the neoadjuvant setting.

Data availability

The datasets analysed during the current study are not publicly available due to local privacy laws but are available from the corresponding author upon reasonable request.

References

  1. Sung H, Ferlay J, Siegel RL et al. Global Cancer statistics 2020: GLOBOCAN estimates of incidence and Mortality Worldwide for 36 cancers in 185 countries. https://doi.org/10.3322/caac.21660. Cancer J Clin. 2021/05/01 2021;71(3):209–49. doi:https://doi.org/10.3322/caac.21660.

  2. Oeffinger KC, Fontham ETH, Etzioni R, et al. Breast Cancer screening for women at average risk: 2015 Guideline Update from the American Cancer Society. JAMA. 2015;314(15):1599–614. https://doi.org/10.1001/jama.2015.12783.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Schünemann HJ, Lerda D, Quinn C et al. Breast Cancer Screening and Diagnosis: A Synopsis of the European Breast Guidelines. Annals of internal medicine. 2020/01/07 2019;172(1):46–56. https://doi.org/10.7326/M19-2125.

  4. Barba D, Leon-Sosa A, Caicedo A, et al. Breast cancer, screening and diagnostic tools: all you need to know. Crit Rev Oncol/Hematol. 2021;157:103174. https://doi.org/10.1016/j.critrevonc.2020.103174.

    Article  PubMed  Google Scholar 

  5. Buono G, Gerratana L, Bulfoni M, et al. Circulating tumor DNA analysis in breast cancer: is it ready for prime-time? Cancer Treat Rev Feb. 2019;73:73–83. https://doi.org/10.1016/j.ctrv.2019.01.004.

    Article  CAS  Google Scholar 

  6. Rakha EA, Tse GM, Quinn CM, Rakha EA, Tse GM, Quinn CM. An update on the pathological classification of breast cancer. Histopathology. 2023;82(1):5–16. https://doi.org/10.1111/his.14786.

    Article  PubMed  Google Scholar 

  7. Rakha EA, Reis-Filho JS, Baehner F, et al. Breast cancer prognostic classification in the molecular era: the role of histological grade. Breast Cancer Res. 2010;2010/07/30(4):207. https://doi.org/10.1186/bcr2607.

    Article  Google Scholar 

  8. Bloom HJ, Richardson WW. Histological grading and prognosis in breast cancer; a study of 1409 cases of which 359 have been followed for 15 years. Br J Cancer Sep. 1957;11(3):359–77. https://doi.org/10.1038/bjc.1957.43.

    Article  CAS  Google Scholar 

  9. Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology Nov. 1991;19(5):403–10. https://doi.org/10.1111/j.1365-2559.1991.tb00229.x.

    Article  CAS  Google Scholar 

  10. Schwartz AB, Siddiqui G, Barbieri JS, et al. The accuracy of mobile teleradiology in the evaluation of chest X-rays. J Telemed Telecare. 2014;20(8):460–3. https://doi.org/10.1177/1357633x14555639.

    Article  PubMed  Google Scholar 

  11. van Dooijeweert C, van Diest PJ, Ellis IO. Grading of invasive breast carcinoma: the way forward. Virchows Arch Jan. 2022;480(1):33–43. https://doi.org/10.1007/s00428-021-03141-2.

    Article  Google Scholar 

  12. Desmedt C, Haibe-Kains B, Wirapati P, et al. Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res Aug. 2008;15(16):5158–65. https://doi.org/10.1158/1078-0432.CCR-07-4756.

    Article  Google Scholar 

  13. Cardoso F, Kyriakides S, Ohno S, et al. Early breast cancer: ESMO Clinical Practice guidelines for diagnosis, treatment and follow-up. Ann Oncol Oct. 2019;01(10):1674. https://doi.org/10.1093/annonc/mdz189.

    Article  Google Scholar 

  14. Acs B, Fredriksson I, Rönnlund C, et al. Variability in breast Cancer Biomarker Assessment and the Effect on Oncological Treatment decisions: a Nationwide 5-Year Population-based study. Cancers. 2021;13(5). https://doi.org/10.3390/cancers13051166.

  15. Lorgis V, Algros MP, Villanueva C, et al. Discordance in early breast cancer for tumour grade, Estrogen Receptor, Progesteron receptors and human epidermal Receptor-2 status between core needle biopsy and surgical excisional primary tumour. Breast. 2011;20(3):284–7. https://doi.org/10.1016/j.breast.2010.12.007.

    Article  PubMed  Google Scholar 

  16. van Dooijeweert C, van Diest PJ, Willems SM et al. Significant inter- and intra-laboratory variation in grading of invasive breast cancer: A nationwide study of 33,043 patients in the Netherlands. https://doi.org/10.1002/ijc.32330. International Journal of Cancer. 2020/02/01 2020;146(3):769–780. doi:https://doi.org/10.1002/ijc.32330.

  17. Cahill RA, Walsh D, Landers RJ, Watson RG. Preoperative profiling of symptomatic breast cancer by diagnostic core biopsy. Ann Surg Oncol Jan. 2006;13(1):45–51. https://doi.org/10.1245/ASO.2006.03.047.

    Article  Google Scholar 

  18. Woeste MR, Jacob K, Duff MB, et al. Impact of routine expert breast pathology consultation and factors predicting discordant diagnosis. Surg Oncol. 2022;45:101860. https://doi.org/10.1016/j.suronc.2022.101860.

    Article  PubMed  Google Scholar 

  19. Newman EA, Guest AB, Helvie MA, et al. Cancer. 2006;107(10):2346–51. https://doi.org/10.1002/cncr.22266. Changes in surgical management resulting from case review at a breast cancer multidisciplinary tumor board/11/15 2006.

  20. Sharma A, Weitz P, Wang Y, et al. Development and prognostic validation of a three-level NHG-like deep learning-based model for histological grading of breast cancer. Breast Cancer Res Jan. 2024;29(1):17. https://doi.org/10.1186/s13058-024-01770-4.

    Article  CAS  Google Scholar 

  21. Couture HD, Williams LA, Geradts J, et al. Image analysis with deep learning to predict breast cancer grade, ER status, histologic subtype, and intrinsic subtype. npj Breast Cancer. 2018;2018/09/03(1):30. https://doi.org/10.1038/s41523-018-0079-1.

    Article  Google Scholar 

  22. Jaroensri R, Wulczyn E, Chen PHC, et al. Deep learning models for histologic grading of breast cancer and association with disease prognosis. NPJ Breast cancer. 2022;8(1). https://doi.org/10.1038/s41523-022-00478-y.

  23. Wang Y, Acs B, Robertson S, et al. Improved breast cancer histological grading using deep learning. Ann Oncol. 2022. https://doi.org/10.1016/j.annonc.2021.09.007.

    Article  PubMed  Google Scholar 

  24. Regionalt cancercentrum Stockholm Gotland. Nationellt Kvalitetsregister för Bröstcancer (NKBC) Sammanfattning och vägledning till den interaktiva årsrapporten för 2022. https://statistik.incanet.se/brostcancer/.

  25. Goode A, Gilbert B, Harkes J, Jukic D, Satyanarayanan M. OpenSlide: a vendor-neutral software foundation for digital pathology. J Pathol Inf. 2013;4:27. https://doi.org/10.4103/2153-3539.119005.

    Article  Google Scholar 

  26. Otsu N. A threshold selection method from Gray-Level Histograms. IEEE Trans Syst Man Cybernetics. 1979;9(1):62–6. https://doi.org/10.1109/TSMC.1979.4310076.

    Article  Google Scholar 

  27. Macenko M, Niethammer M, Marron JS et al. A method for normalizing histology slides for quantitative analysis. 2009:1107–10.

  28. Pech-Pacheco JL, Cristobal G, Chamorro-Martinez J, Fernandez-Valdivia J. Diatom autofocusing in brightfield microscopy: a comparative study. 2000:314–3173.

  29. Szegedy C, Vanhoucke V, Ioffe S, Shlens J, Wojna Z. Rethinking the Inception Architecture for Computer Vision. 2016:2818–2826.

  30. Robin X, Turck N, Hainard A et al. pROC: an open-source package for R and S + to analyze and compare ROC curves. BMC Bioinformatics. 2011/03/17 2011;12(1):77. https://doi.org/10.1186/1471-2105-12-77.

  31. Youden WJ. Index for rating diagnostic tests. Cancer Jan. 1950;3(1):32–5. https://doi.org/10.1002/1097-0142(1950)3:1<32::aid-cncr2820030106>3.0.co;2-3.

    Article  CAS  Google Scholar 

  32. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics Mar. 1977;33(1):159–74.

    Article  CAS  Google Scholar 

  33. Cohen J. A Coefficient of Agreement for Nominal Scales. Educational and Psychological Measurement. 1960/04/01 1960;20(1):37–46. https://doi.org/10.1177/001316446002000104.

  34. Jung YY, Hyun CL, Jin M-S, et al. Histomorphological factors Predicting the response to Neoadjuvant Chemotherapy in Triple-negative breast Cancer. J Breast Cancer 9/. 2016;19(3):261–7.

    Article  Google Scholar 

  35. Wetstein SC, de Jong VMT, Stathonikos N et al. Deep learning-based breast cancer grading and survival analysis on whole-slide histopathology images. Scientific reports. 2022/09/06 2022;12(1):15102. https://doi.org/10.1038/s41598-022-19112-9.

  36. Mantrala S, Ginter PS, Koka D, et al. Concordance in breast Cancer grading by Artificial Intelligence on whole slide images compares with a multi-institutional cohort of breast pathologists. Arch Pathol Lab Med. 2022;146(11):1369–77. https://doi.org/10.5858/arpa.2021-0299-OA.

    Article  CAS  PubMed  Google Scholar 

  37. Couture HD. Deep learning-based prediction of Molecular Tumor biomarkers from H&E: a practical review. J Personalized Med. 2022;12(12):2022. https://doi.org/10.3390/jpm12122022.

    Article  Google Scholar 

  38. Rakha EA, Ellis IO. An overview of assessment of prognostic and predictive factors in breast cancer needle core biopsy specimens. J Clin Pathol. 2007;60(12):1300. https://doi.org/10.1136/jcp.2006.045377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bulten W, Kartasalo K, Chen P-HC, et al. Artificial intelligence for diagnosis and Gleason grading of prostate cancer: the PANDA challenge. Nat Med. 2022;01(1):154–63. https://doi.org/10.1038/s41591-021-01620-2. /01 2022.

    Article  CAS  Google Scholar 

  40. Pantanowitz L, Quiroga-Garza GM, Bien L, et al. An artificial intelligence algorithm for prostate cancer diagnosis in whole slide images of core needle biopsies: a blinded clinical validation and deployment study. Lancet Digit Health Aug. 2020;2(8):e407–16. https://doi.org/10.1016/S2589-7500(20)30159-X.

    Article  Google Scholar 

  41. Ström P, Kartasalo K, Olsson H et al. Artificial intelligence for diagnosis and grading of prostate cancer in biopsies: a population-based, diagnostic study. Research Support, Non-U.S. Gov’t. Validation Study. Lancet Oncol. Feb 2020;21(2):222–232. doi: 10.1016/S1470-2045(19)30738-7. Epub 2020 Jan 8

  42. Fondón I, Sarmiento A, Garíca AI, et al. Automatic classification of tissue malignancy for breast carcinoma diagnosis. Article. Computers Biology Med May. 2018;96:41–51. https://doi.org/10.1016/j.compbiomed.2018.03.003.

    Article  Google Scholar 

  43. Chattopadhyay S, Dey A, Sarkar R, et al. MTRRE-Net: a deep learning model for detection of breast cancer from histopathological images. Comput Biol Med. 2022;150:106155. https://doi.org/10.1016/j.compbiomed.2022.106155.

    Article  PubMed  Google Scholar 

  44. Sandbank J, Bataillon G, Vincent-Salomon A, et al. Validation and real-world clinical application of an artificial intelligence algorithm for breast cancer detection in biopsies. NPJ Breast cancer. 2022;8(1). https://doi.org/10.1038/s41523-022-00496-w.

  45. Hameed Z, Zahia S, Garcia-Zapirain B, Javier Aguirre J, María Vanegas A. Breast Cancer histopathology image classification using an ensemble of Deep Learning models. Sens (Basel) Aug. 2020;05(16). https://doi.org/10.3390/s20164373.

  46. Ogier du Terrail J, Leopold A, Joly C et al. Federated learning for predicting histological response to neoadjuvant chemotherapy in triple-negative breast cancer. Nature medicine. 2023/01/01 2023;29(1):135–146. https://doi.org/10.1038/s41591-022-02155-w.

  47. Li B, Li FL, Tian J, et al. Deep learning with biopsy whole slide images for pretreatment prediction of pathological complete response to neoadjuvant chemotherapy in Breast cancer: a multicenter study. Breast. 2022;66:183–90. https://doi.org/10.1016/j.Breast.2022.10.004.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Chen Y, Li H, Janowczyk A, et al. Computational pathology improves risk stratification of a multi-gene assay for early stage ER + breast cancer. NPJ Breast Cancer May. 2023;17(1):40. https://doi.org/10.1038/s41523-023-00545-y.

    Article  CAS  Google Scholar 

  49. Wang Y, Ali MA, Vallon-Christersson J, Humphreys K, Hartman J, Rantalainen M. Transcriptional intra-tumour heterogeneity predicted by deep learning in routine breast histopathology slides provides independent prognostic information. Eur J Cancer Sep. 2023;191:112953. https://doi.org/10.1016/j.ejca.2023.112953.

    Article  CAS  Google Scholar 

  50. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med Dec. 2004;30(27):2817–26. https://doi.org/10.1056/NEJMoa041588.

    Article  Google Scholar 

  51. Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant recurrence after 5 years of endocrine treatment: a combined analysis of patients from the Austrian breast and colorectal cancer study group 8 and arimidex, tamoxifen alone or in combination randomized trials using the PAM50 risk of recurrence score. J Clin Oncol Mar. 2015;10(8):916–22. https://doi.org/10.1200/JCO.2014.55.6894.

    Article  CAS  Google Scholar 

  52. Stull TS, Goodwin MC, Frazier TG et al. P3-06-05: Comparison of Oncotype DX® Recurrence Scores between Surgical and Core Biopsy Specimens in Breast Cancer Patients. Cancer research. 2011;71(24_Supplement):P3-06-05-P3-06-05. https://doi.org/10.1158/0008-5472.SABCS11-P3-06-05.

  53. Bear HD, Wan W, Robidoux A, et al. Re: using the 21-gene assay from core needle biopsies to choose neoadjuvant therapy for breast cancer: a multicenter trial (115, pg 917, 2017). Correction. J Surg Oncol Sep. 2018;118(4):722–722. https://doi.org/10.1002/jso.24798.

    Article  Google Scholar 

  54. Picornell AC, Echavarria I, Alvarez E, et al. Breast cancer PAM50 signature: correlation and concordance between RNA-Seq and digital multiplexed gene expression technologies in a triple negative breast cancer series. Article. Bmc Genomics Jun. 2019;20:11. https://doi.org/10.1186/s12864-019-5849-0.

    Article  CAS  Google Scholar 

  55. Ohara AM, Naoi Y, Shimazu K, et al. PAM50 for prediction of response to neoadjuvant chemotherapy for ER-positive breast cancer. Article. Breast Cancer Res Treat Feb. 2019;173(3):533–43. https://doi.org/10.1007/s10549-018-5020-7.

    Article  CAS  Google Scholar 

  56. Kwa M, Makris A, Esteva FJ. Clinical utility of gene-expression signatures in early stage breast cancer. Nature Reviews Clinical Oncology. 2017/10/01 2017;14(10):595–610. https://doi.org/10.1038/nrclinonc.2017.74.

Download references

Funding

Open access funding provided by Karolinska Institute. This project was supported by funding from the Swedish Research Council under the frame of ERA PerMed (ERAPERMED2019-224 - ABCAP), Swedish Research Council, VINNOVA (SwAIPP project), Swedish Cancer Society, Karolinska Institutet (Cancer Research KI; StratCan), MedTechLabs, Swedish e-science Research Centre (SeRC), Stockholm Region, Stockholm Cancer Society and Swedish Breast Cancer Association.

Author information

Authors and Affiliations

Authors

Contributions

CB was responsible for data curation, analyses and drafting the manuscript, MR and JH were responsible for project conceptualisation and funding acquisition as well as manuscript drafting. YW, AS, PW, EK and SR assisted in data curation. All authors read and approved the final paper.

Corresponding author

Correspondence to Mattias Rantalainen.

Ethics declarations

Ethical approval

The study has approval from the Swedish Ethical Review Authority (2017/2106-31, with amendments 2018/1462-32 and 2019–02336).

Competing interests

The authors declare the following financial interests or personal relationships which may be considered as potential competing interests: JH has obtained speaker’s honoraria or advisory board remunerations from Roche, Novartis, AstraZeneca, Eli Lilly, MSD and Gilead. JH has received institutional research grants from Cepheid, Roche, Novartis and AstraZeneca. MR and JH are co-founders and shareholders of Stratipath AB. EK is employed by Stratipath AB. YW, PW and SR are employed by Stratipath AB and hold employee stock options. All remaining authors have declared no conflicts of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boissin, C., Wang, Y., Sharma, A. et al. Deep learning-based risk stratification of preoperative breast biopsies using digital whole slide images. Breast Cancer Res 26, 90 (2024). https://doi.org/10.1186/s13058-024-01840-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13058-024-01840-7

Keywords