1
|
Zwager MC, Yu S, Buikema HJ, de Bock GH, Ramsing TW, Thagaard J, Koopman T, van der Vegt B. Advancing Ki67 hotspot detection in breast cancer: a comparative analysis of automated digital image analysis algorithms. Histopathology 2025; 86:204-213. [PMID: 39104219 PMCID: PMC11649514 DOI: 10.1111/his.15294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 08/07/2024]
Abstract
AIM Manual detection and scoring of Ki67 hotspots is difficult and prone to variability, limiting its clinical utility. Automated hotspot detection and scoring by digital image analysis (DIA) could improve the assessment of the Ki67 hotspot proliferation index (PI). This study compared the clinical performance of Ki67 hotspot detection and scoring DIA algorithms based on virtual dual staining (VDS) and deep learning (DL) with manual Ki67 hotspot PI assessment. METHODS Tissue sections of 135 consecutive invasive breast carcinomas were immunohistochemically stained for Ki67. Two DIA algorithms, based on VDS and DL, automatically determined the Ki67 hotspot PI. For manual assessment; two independent observers detected hotspots and calculated scores using a validated scoring protocol. RESULTS Automated hotspot detection and assessment by VDS and DL could be performed in 73% and 100% of the cases, respectively. Automated hotspot detection by VDS and DL led to higher Ki67 hotspot PIs (mean 39.6% and 38.3%, respectively) compared to manual consensus Ki67 PIs (mean 28.8%). Comparing manual consensus Ki67 PIs with VDS Ki67 PIs revealed substantial correlation (r = 0.90), while manual consensus versus DL Ki67 PIs demonstrated high correlation (r = 0.95). CONCLUSION Automated Ki67 hotspot detection and analysis correlated strongly with manual Ki67 assessment and provided higher PIs compared to manual assessment. The DL-based algorithm outperformed the VDS-based algorithm in clinical applicability, because it did not depend on virtual alignment of slides and correlated stronger with manual scores. Use of a DL-based algorithm may allow clearer Ki67 PI cutoff values, thereby improving the clinical usability of Ki67.
Collapse
Affiliation(s)
- Mieke C Zwager
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Shibo Yu
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Henk J Buikema
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Geertruida H de Bock
- Department of EpidemiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | | | | | - Timco Koopman
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Pathologie FrieslandLeeuwardenThe Netherlands
| | - Bert van der Vegt
- Department of PathologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
2
|
Zilenaite-Petrulaitiene D, Rasmusson A, Valkiuniene RB, Laurinaviciene A, Petkevicius L, Laurinavicius A. Spatial distributions of CD8 and Ki67 cells in the tumor microenvironment independently predict breast cancer-specific survival in patients with ER+HER2- and triple-negative breast carcinoma. PLoS One 2024; 19:e0314364. [PMID: 39576843 PMCID: PMC11584100 DOI: 10.1371/journal.pone.0314364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024] Open
Abstract
INTRODUCTION Breast cancer (BC) presents diverse malignancies with varying biological and clinical behaviors, driven by an interplay between cancer cells and tumor microenvironment. Deciphering these interactions is crucial for personalized diagnostics and treatment. This study explores the prognostic impact of tumor proliferation and immune response patterns, assessed by computational pathology indicators, on breast cancer-specific survival (BCSS) models in estrogen receptor-positive HER2-negative (ER+HER2-) and triple-negative BC (TNBC) patients. MATERIALS AND METHODS Whole-slide images of tumor surgical excision samples from 252 ER+HER2- patients and 63 TNBC patients stained for estrogen and progesterone receptors, Ki67, HER2, and CD8 were analyzed. Digital image analysis (DIA) was performed for tumor tissue segmentation and quantification of immunohistochemistry (IHC) markers; the DIA outputs were subsampled by hexagonal grids to assess the spatial distributions of Ki67-positive tumor cells and CD8-positive (CD8+) cell infiltrates, expressed as Ki67-entropy and CD8-immunogradient indicators, respectively. Prognostic models for BCSS were generated using multivariable Cox regression analysis, integrating clinicopathological and computational IHC indicators. RESULTS In the ER+HER2- BC, multivariable Cox regression revealed that high CD8+ density within the tumor interface zone (IZ) (HR: 0.26, p = 0.0056), low immunodrop indicator of CD8+ density (HR: 2.93, p = 0.0051), and low Ki67-entropy (HR: 5.95, p = 0.0.0061) were independent predictors of better BCSS, while lymph node involvement predicted worse BCSS (HR: 3.30, p = 0.0013). In TNBC, increased CD8+ density in the IZ stroma (HR: 0.19, p = 0.0119) and Ki67-entropy (HR: 3.31, p = 0.0250) were independent predictors of worse BCSS. Combining these independent indicators enhanced prognostic stratification in both BC subtypes. CONCLUSIONS Computational biomarkers, representing spatial properties of the tumor proliferation and immune cell infiltrates, provided independent prognostic information beyond conventional IHC markers in BC. Integrating Ki67-entropy and CD8-immunogradient indicators into prognostic models can improve patient stratification with regard to BCSS.
Collapse
Affiliation(s)
- Dovile Zilenaite-Petrulaitiene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Institute of Informatics, Faculty of Mathematics and Informatics, Vilnius University, Vilnius, Lithuania
| | - Allan Rasmusson
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Ruta Barbora Valkiuniene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Aida Laurinaviciene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Linas Petkevicius
- Institute of Informatics, Faculty of Mathematics and Informatics, Vilnius University, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| |
Collapse
|
3
|
Lashen AG, Wahab N, Toss M, Miligy I, Ghanaam S, Makhlouf S, Atallah N, Ibrahim A, Jahanifar M, Lu W, Graham S, Bilal M, Bhalerao A, Mongan NP, Minhas F, Raza SEA, Provenzano E, Snead D, Rajpoot N, Rakha EA. Characterization of Breast Cancer Intra-Tumor Heterogeneity Using Artificial Intelligence. Cancers (Basel) 2024; 16:3849. [PMID: 39594804 PMCID: PMC11593220 DOI: 10.3390/cancers16223849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/24/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Intra-tumor heterogeneity (ITH) is a fundamental characteristic of breast cancer (BC), influencing tumor progression, prognosis, and therapeutic responses. However, the complexity of ITH in BC makes its accurate characterization challenging. This study leverages deep learning (DL) techniques to comprehensively evaluate ITH in early-stage luminal BC and provide a nuanced understanding of its impact on tumor behavior and patient outcomes. A large cohort (n = 2561) of early-stage luminal BC was evaluated using whole slide images (WSIs) of hematoxylin and eosin-stained slides of excision specimens. Morphological features of both the tumor and stromal components were meticulously annotated by a panel of pathologists in a subset of cases. A DL model was applied to develop an algorithm to assess the degree of heterogeneity of various morphological features per individual case utilizing defined patches. The results of extracted features were used to generate an overall heterogeneity score that was correlated with the clinicopathological features and outcome. Overall, 162 features were quantified and a significant positive correlation between these features was identified. Specifically, there was a significant association between a high degree of intra-tumor heterogeneity and larger tumor size, poorly differentiated tumors, highly proliferative tumors, tumors of no special type (NST), and those with low estrogen receptor (ER) expression. When all features are considered in combination, a high overall heterogeneity score was significantly associated with parameters characteristic of aggressive tumor behavior, and it was an independent predictor of poor patient outcome. In conclusion, DL models can be used to accurately decipher the complexity of ITH and provide extra information for outcome prediction.
Collapse
Affiliation(s)
- Ayat G. Lashen
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin El-Kom 6131567, Egypt
| | - Noorul Wahab
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Michael Toss
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
| | - Islam Miligy
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin El-Kom 6131567, Egypt
| | - Suzan Ghanaam
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
| | - Shorouk Makhlouf
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
| | - Nehal Atallah
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
- Department of Pathology, Faculty of Medicine, Menoufia University, Shibin El-Kom 6131567, Egypt
| | - Asmaa Ibrahim
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
| | - Mostafa Jahanifar
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Wenqi Lu
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Simon Graham
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Mohsin Bilal
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Abhir Bhalerao
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Nigel P. Mongan
- School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham LE12 5RD, UK;
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Fayyaz Minhas
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Shan E Ahmed Raza
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Elena Provenzano
- Department of Pathology, Cambridge Biomedical Research Centre, Cambridge University Hospitals, Cambridge CB2 0QQ, UK;
| | - David Snead
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
- Department of pathology, University Hospital Coventry and Warwickshire, Coventry CV2 2DX, UK
| | - Nasir Rajpoot
- Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK; (N.W.); (M.J.); (W.L.); (S.G.); (M.B.); (A.B.); (F.M.); (S.EA.R.); (D.S.); (N.R.)
| | - Emad A. Rakha
- Breast Cancer Research Unit, University of Nottingham, Nottingham NG7 2RD, UK; (A.G.L.); (M.T.); (I.M.); (S.G.); (S.M.); (N.A.); (A.I.)
- Pathology Department, Hamad General Hospital, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| |
Collapse
|
4
|
Zilenaite-Petrulaitiene D, Rasmusson A, Besusparis J, Valkiuniene RB, Augulis R, Laurinaviciene A, Plancoulaine B, Petkevicius L, Laurinavicius A. Intratumoral heterogeneity of Ki67 proliferation index outperforms conventional immunohistochemistry prognostic factors in estrogen receptor-positive HER2-negative breast cancer. Virchows Arch 2024:10.1007/s00428-024-03737-4. [PMID: 38217716 DOI: 10.1007/s00428-024-03737-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
In breast cancer (BC), pathologists visually score ER, PR, HER2, and Ki67 biomarkers to assess tumor properties and predict patient outcomes. This does not systematically account for intratumoral heterogeneity (ITH) which has been reported to provide prognostic value. This study utilized digital image analysis (DIA) and computational pathology methods to investigate the prognostic value of ITH indicators in ER-positive (ER+) HER2-negative (HER2-) BC patients. Whole slide images (WSIs) of surgically excised specimens stained for ER, PR, Ki67, and HER2 from 254 patients were used. DIA with tumor tissue segmentation and detection of biomarker-positive cells was performed. The DIA-generated data were subsampled by a hexagonal grid to compute Haralick's texture indicators for ER, PR, and Ki67. Cox regression analyses were performed to assess the prognostic significance of the immunohistochemistry (IHC) and ITH indicators in the context of clinicopathologic variables. In multivariable analysis, the ITH of Ki67-positive cells, measured by Haralick's texture entropy, emerged as an independent predictor of worse BC-specific survival (BCSS) (hazard ratio (HR) = 2.64, p-value = 0.0049), along with lymph node involvement (HR = 2.26, p-value = 0.0195). Remarkably, the entropy representing the spatial disarrangement of tumor proliferation outperformed the proliferation rate per se established either by pathology reports or DIA. We conclude that the Ki67 entropy indicator enables a more comprehensive risk assessment with regard to BCSS, especially in cases with borderline Ki67 proliferation rates. The study further demonstrates the benefits of high-capacity DIA-generated data for quantifying the essentially subvisual ITH properties.
Collapse
Affiliation(s)
- Dovile Zilenaite-Petrulaitiene
- Institute of Informatics, Faculty of Mathematics and Informatics, Vilnius University, Naugarduko Str. 24, 03225, Vilnius, Lithuania.
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania.
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania.
| | - Allan Rasmusson
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania
| | - Justinas Besusparis
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania
| | - Ruta Barbora Valkiuniene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania
| | - Renaldas Augulis
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania
| | - Aida Laurinaviciene
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania
| | - Benoit Plancoulaine
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- Path-Image/BioTiCla, University of Caen Normandy, François Baclesse Comprehensive Cancer Center, 3 Av. du Général Harris, 14000, Caen, France
| | - Linas Petkevicius
- Institute of Informatics, Faculty of Mathematics and Informatics, Vilnius University, Naugarduko Str. 24, 03225, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- Department of Pathology and Forensic Medicine, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, M. K. Ciurlionio Str. 21, 03101, Vilnius, Lithuania
- National Centre of Pathology, affiliate of Vilnius University Hospital Santaros Klinikos, P. Baublio Str. 5, 08406, Vilnius, Lithuania
| |
Collapse
|
5
|
Makhlouf S, Quinn C, Toss M, Alsaleem M, Atallah NM, Ibrahim A, Rutland CS, Mongan NP, Rakha EA. Quantitative expression of oestrogen receptor in breast cancer: Clinical and molecular significance. Eur J Cancer 2024; 197:113473. [PMID: 38103327 DOI: 10.1016/j.ejca.2023.113473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 12/04/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Oestrogen receptor (ER) positive breast cancer (BC) patients are eligible for endocrine therapy (ET), regardless of ER immunohistochemical expression level. There is a wide spectrum of ER expression and the response to ET is not uniform. This study aimed to assess the clinical and molecular consequences of ER heterogeneity with respect to ET-response. METHODS ER expression, categorised by percentage and staining intensity in a large BC cohort (n = 7559) was correlated with clinicopathological parameters and patient ET response. The Cancer Genome Atlas Data BC cohort (n = 1047) was stratified by ER expression and transcriptomic analysis completed to better understand the molecular basis of ER heterogeneity. RESULTS The quantitative proportional increase in ER expression was positively associated with favourable prognostic parameters. Tumours with 1-9% ER expression were characteristically similar to ER-negative (<1%) tumours. Maximum ET-response was observed in tumours with 100% ER expression, with responses significantly different to tumours exhibiting ER at < 100% and significantly decreased survival rates were observed in tumours with 50% and 10% of ER expression. The Histochemical-score (H-score), which considers both staining intensity and percentage, added significant prognostic value over ER percentage alone with significant outcome differences observed at H-scores of 30, 100 and 200. There was a positive correlation between ER expression and ESR1 mRNA expression and expression of ER-regulated genes. Pathway analysis identified differential expression in key cancer-related pathways in different ER-positive groups. CONCLUSION ET-response is statistically proportionally related to ER expression with significant differences observed at 10%, 50% and 100%. The H-score adds prognostic and predictive information.
Collapse
Affiliation(s)
- Shorouk Makhlouf
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Pathology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Cecily Quinn
- Irish National Breast Screening Programme and Department of Histopathology, St. Vincent's University Hospital, Dublin, Ireland
| | - Michael Toss
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Histopathology, Sheffield Teaching Hospitals NHS Trust, Sheffield, UK
| | - Mansour Alsaleem
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Unit of Scientific Research, Applied College, Qassim University, Saudi Arabia
| | - Nehal M Atallah
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Pathology, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Asmaa Ibrahim
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Catrin S Rutland
- School of Veterinary Medicine and Sciences, University of Nottingham, Sutton Bonington, UK
| | - Nigel P Mongan
- Biodiscovery Institute, School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, UK; Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Emad A Rakha
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK; Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham, UK; Department of Pathology, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
6
|
Digital Image Analysis of Ki67 Heterogeneity Improves the Diagnosis and Prognosis of Gastroenteropancreatic Neuroendocrine Neoplasms. Mod Pathol 2023; 36:100017. [PMID: 36788066 DOI: 10.1016/j.modpat.2022.100017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/29/2022] [Accepted: 09/20/2022] [Indexed: 01/19/2023]
Abstract
Ki67 is a reliable grading and prognostic biomarker of gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs). The intratumor heterogeneity of Ki67, correlated with tumor progression, is a valuable factor that requires image analysis. The application of digital image analysis (DIA) enables new approaches for the assessment of Ki67 heterogeneity distribution. We investigated the diagnostic utility of Ki67 heterogeneity parameters in the classification and grading of GEP-NENs and explored their clinical values with regard to their prognostic relevance. The DIA algorithm was performed on whole-slide images of 102 resection samples with Ki67 staining. Good agreement was observed between the manual and DIA methods in the hotspot evaluation (R2 = 0.94, P < .01). Using the grid-based region of interest approach, score-based heat maps provided a distinctive overview of the intratumoral distribution of Ki67 between neuroendocrine carcinomas and neuroendocrine tumors. The computation of heterogeneity parameters related to DIA-determined Ki67 showed that the coefficient of variation and Morisita-Horn index were directly related to the classification and grading of GEP-NENs and provided insights into distinguishing high-grade neuroendocrine neoplasms (grade 3 neuroendocrine tumor vs neuroendocrine carcinoma, P < .01). Our study showed that a high Morisita-Horn index correlated with poor disease-free survival (multivariate analysis: hazard ratio, 56.69), which was found to be the only independent predictor of disease-free survival in patients with GEP-NEN. These spatial biomarkers have an impact on the classification and grading of tumors and highlight the prognostic associations of tumor heterogeneity. Digitization of Ki67 variations provides a direct and objective measurement of tumor heterogeneity and better predicts the biological behavior of GEP-NENs.
Collapse
|
7
|
Reinert T, Cascelli F, de Resende CAA, Gonçalves AC, Godo VSP, Barrios CH. Clinical implication of low estrogen receptor (ER-low) expression in breast cancer. Front Endocrinol (Lausanne) 2022; 13:1015388. [PMID: 36506043 PMCID: PMC9729538 DOI: 10.3389/fendo.2022.1015388] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Breast cancer is a heterogeneous disease, and the estrogen receptor (ER) remains the most important biomarker in breast oncology. Most guidelines set a positive expression threshold of 1% staining in immunohistochemistry (IHC) to define ER positivity. However, different expression levels may be associated with diverse degrees of sensitivity to endocrine therapy as ER expression may impact breast cancer molecular biology as a continuous variable. ER-lo tumors, defined as those with 1-10% ER expression, represent a relatively small subgroup of breast cancer patients, with an estimated prevalence of 2-7%. These tumors are similar to ERneg disease in their molecular landscape, clinicopathological characteristics, prognosis, and response to therapy. Nevertheless, a proportion may retain some degree of ER signaling dependency, and the possibility of responding to some degree to endocrine therapy cannot be completely ruled out. This review article discusses the most important considerations regarding the definition of ER positivity, pathology assessment, prognosis, and therapeutic implication of ERlo breast cancer from the medical oncology perspective.
Collapse
Affiliation(s)
- Tomás Reinert
- Breast Medical Oncology, Oncoclínicas, Porto Alegre, Brazil
- Breast Cancer Group, Latin American Cooperative Oncology Group, Porto Alegre, Brazil
| | - Fanny Cascelli
- Breast Medical Oncology, Oncoclínicas, São Paulo, Brazil
| | | | | | | | - Carlos Henrique Barrios
- Breast Medical Oncology, Oncoclínicas, Porto Alegre, Brazil
- Breast Cancer Group, Latin American Cooperative Oncology Group, Porto Alegre, Brazil
| |
Collapse
|
8
|
Kreipe H, Harbeck N, Christgen M. Clinical validity and clinical utility of Ki67 in early breast cancer. Ther Adv Med Oncol 2022; 14:17588359221122725. [PMID: 36105888 PMCID: PMC9465566 DOI: 10.1177/17588359221122725] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022] Open
Abstract
Ki67 represents an immunohistochemical nuclear localized marker that is widely
used in surgical pathology. Nuclear immunoreactivity for Ki67 indicates that
cells are cycling and are in G1- to S-phase. The percentage of Ki67-positive
tumor cells (Ki67 index) therefore provides an estimate of the growth fraction
in tumor specimens. In breast cancer (BC), tumor cell proliferation rate is one
of the most relevant prognostic markers and Ki67 is consequently helpful in
prognostication similar to histological grading and mRNA profiling-based BC risk
stratification. In BCs treated with short-term preoperative endocrine therapy,
Ki67 dynamics enable distinguishing between endocrine sensitive and resistant
tumors. Despite its nearly universal use in pathology laboratories worldwide, no
internationally accepted consensus has yet been achieved for some methodological
details related to Ki67 immunohistochemistry (IHC). Controversial issues refer
to choice of IHC antibody clones, scoring methods, inter-laboratory
reproducibility, and the potential value of computer-assisted imaging analysis
and/or artificial intelligence for Ki67 assessment. Prospective clinical trials
focusing on BC treatment have proven that Ki67, as determined by standardized
central pathology assessment, is of clinical validity. Clinical utility has been
demonstrated in huge observational studies.
Collapse
Affiliation(s)
- Hans Kreipe
- Institute of Pathology, Hannover Medical School, Carl-Neubergstraße 1, Hannover 30625, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München (LMU) Frauenklinik Maistrasse-Innenstadt und Klinikum Großhadern, Germany
| | | |
Collapse
|
9
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
10
|
Radziuviene G, Rasmusson A, Augulis R, Grineviciute RB, Zilenaite D, Laurinaviciene A, Ostapenko V, Laurinavicius A. Intratumoral Heterogeneity and Immune Response Indicators to Predict Overall Survival in a Retrospective Study of HER2-Borderline (IHC 2+) Breast Cancer Patients. Front Oncol 2021; 11:774088. [PMID: 34858854 PMCID: PMC8631965 DOI: 10.3389/fonc.2021.774088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Breast cancer (BC) categorized as human epidermal growth factor receptor 2 (HER2) borderline [2+ by immunohistochemistry (IHC 2+)] presents challenges for the testing, frequently obscured by intratumoral heterogeneity (ITH). This leads to difficulties in therapy decisions. We aimed to establish prognostic models of overall survival (OS) of these patients, which take into account spatial aspects of ITH and tumor microenvironment by using hexagonal tiling analytics of digital image analysis (DIA). In particular, we assessed the prognostic value of Immunogradient indicators at the tumor–stroma interface zone (IZ) as a feature of antitumor immune response. Surgical excision samples stained for estrogen receptor (ER), progesterone receptor (PR), Ki67, HER2, and CD8 from 275 patients with HER2 IHC 2+ invasive ductal BC were used in the study. DIA outputs were subsampled by HexT for ITH quantification and tumor microenvironment extraction for Immunogradient indicators. Multiple Cox regression revealed HER2 membrane completeness (HER2 MC) (HR: 0.18, p = 0.0007), its spatial entropy (HR: 0.37, p = 0.0341), and ER contrast (HR: 0.21, p = 0.0449) as independent predictors of better OS, with worse OS predicted by pT status (HR: 6.04, p = 0.0014) in the HER2 non-amplified patients. In the HER2-amplified patients, HER2 MC contrast (HR: 0.35, p = 0.0367) and CEP17 copy number (HR: 0.19, p = 0.0035) were independent predictors of better OS along with worse OS predicted by pN status (HR: 4.75, p = 0.0018). In the non-amplified tumors, three Immunogradient indicators provided the independent prognostic value: CD8 density in the tumor aspect of the IZ and CD8 center of mass were associated with better OS (HR: 0.23, p = 0.0079 and 0.14, p = 0.0014, respectively), and CD8 density variance along the tumor edge predicted worse OS (HR: 9.45, p = 0.0002). Combining these three computational indicators of the CD8 cell spatial distribution within the tumor microenvironment augmented prognostic stratification of the patients. In the HER2-amplified group, CD8 cell density in the tumor aspect of the IZ was the only independent immune response feature to predict better OS (HR: 0.22, p = 0.0047). In conclusion, we present novel prognostic models, based on computational ITH and Immunogradient indicators of the IHC biomarkers, in HER2 IHC 2+ BC patients.
Collapse
Affiliation(s)
- Gedmante Radziuviene
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania.,Institute of Biosciences, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Allan Rasmusson
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania.,Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| | - Renaldas Augulis
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania.,Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| | - Ruta Barbora Grineviciute
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Dovile Zilenaite
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania.,Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| | - Aida Laurinaviciene
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania.,Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| | - Valerijus Ostapenko
- Department of Breast Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania.,Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
11
|
Yousif M, van Diest PJ, Laurinavicius A, Rimm D, van der Laak J, Madabhushi A, Schnitt S, Pantanowitz L. Artificial intelligence applied to breast pathology. Virchows Arch 2021; 480:191-209. [PMID: 34791536 DOI: 10.1007/s00428-021-03213-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/12/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022]
Abstract
The convergence of digital pathology and computer vision is increasingly enabling computers to perform tasks performed by humans. As a result, artificial intelligence (AI) is having an astoundingly positive effect on the field of pathology, including breast pathology. Research using machine learning and the development of algorithms that learn patterns from labeled digital data based on "deep learning" neural networks and feature-engineered approaches to analyze histology images have recently provided promising results. Thus far, image analysis and more complex AI-based tools have demonstrated excellent success performing tasks such as the quantification of breast biomarkers and Ki67, mitosis detection, lymph node metastasis recognition, tissue segmentation for diagnosing breast carcinoma, prognostication, computational assessment of tumor-infiltrating lymphocytes, and prediction of molecular expression as well as treatment response and benefit of therapy from routine H&E images. This review critically examines the literature regarding these applications of AI in the area of breast pathology.
Collapse
Affiliation(s)
- Mustafa Yousif
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Arvydas Laurinavicius
- Department of Pathology, Pharmacology and Forensic Medicine, Faculty of Medicine, Vilnius University, and National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - David Rimm
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jeroen van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, and Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA
| | - Stuart Schnitt
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Breast Oncology Program, Dana-Farber/Brigham and Women's Cancer Center, Boston, MA, USA
| | | |
Collapse
|
12
|
Laurinavicius A, Rasmusson A, Plancoulaine B, Shribak M, Levenson R. Machine-Learning-Based Evaluation of Intratumoral Heterogeneity and Tumor-Stroma Interface for Clinical Guidance. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1724-1731. [PMID: 33895120 PMCID: PMC11727842 DOI: 10.1016/j.ajpath.2021.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022]
Abstract
Assessment of intratumoral heterogeneity and tumor-host interaction within the tumor microenvironment is becoming increasingly important for innovative cancer therapy decisions because of the unique information it can generate about the state of the disease. However, its assessment and quantification are limited by ambiguous definitions of the tumor-host interface and by human cognitive capacity in current pathology practice. Advances in machine learning and artificial intelligence have opened the field of digital pathology to novel tissue image analytics and feature extraction for generation of high-capacity computational disease management models. A particular benefit is expected from machine-learning applications that can perform extraction and quantification of subvisual features of both intratumoral heterogeneity and tumor microenvironment aspects. These methods generate information about cancer cell subpopulation heterogeneity, potential tumor-host interactions, and tissue microarchitecture, derived from morphologically resolved content using both explicit and implicit features. Several studies have achieved promising diagnostic, prognostic, and predictive artificial intelligence models that often outperform current clinical and pathology criteria. However, further effort is needed for clinical adoption of such methods through development of standardizable high-capacity workflows and proper validation studies.
Collapse
Affiliation(s)
- Arvydas Laurinavicius
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences of the Faculty of Medicine of Vilnius University, Vilnius, Lithuania; National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania.
| | - Allan Rasmusson
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences of the Faculty of Medicine of Vilnius University, Vilnius, Lithuania; National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
| | - Benoit Plancoulaine
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences of the Faculty of Medicine of Vilnius University, Vilnius, Lithuania; ANTICIPE, Inserm (UMR 1086), Cancer Center F. Baclesse, Normandy University, Caen, France
| | - Michael Shribak
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences of the Faculty of Medicine of Vilnius University, Vilnius, Lithuania; Marine Biological Laboratory of University of Chicago, Woods Hole, Massachusetts
| | - Richard Levenson
- Department of Pathology, Forensic Medicine and Pharmacology, Institute of Biomedical Sciences of the Faculty of Medicine of Vilnius University, Vilnius, Lithuania; Department of Pathology and Laboratory Medicine, University of California Davis Health, Sacramento, California
| |
Collapse
|
13
|
Lashen AG, Toss MS, Katayama A, Gogna R, Mongan NP, Rakha EA. Assessment of proliferation in breast cancer: cell cycle or mitosis? An observational study. Histopathology 2021; 79:1087-1098. [PMID: 34455622 DOI: 10.1111/his.14542] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/25/2021] [Accepted: 08/15/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Proliferation is an important indicator of breast cancer (BC) prognosis, but is assessed using different approaches. Not all cells in the cell cycle are committed to division. This study aimed to characterise quantitative differences between BC cells in the cell cycle and those in mitosis and assess their relationship with other pathological parameters. METHODS AND RESULTS A cohort of BC sections (n = 621) was stained with haematoxylin and eosin and immunohistochemistry for Ki-67. The proportion of mitotic cells and Ki-67-positive cells was assessed in the same areas. The Cancer Genome Atlas (TCGA) BC cohort was used to assess MKI-67 transcriptome level and its association with the mitotic counts. The mean proportion of BC cells in the cell cycle was 24% (range = 1-90%), while the mean proportion of BC cells in mitosis was 5% (range = 0-73%). A low proportion of mitoses to whole cycling cells was associated with low histological grade tumours and the luminal A molecular subtype, while tumours with a high proportion of mitoses to the overall cycling cells were associated with triple-negative subtype, larger tumour size, grade 3 tumours and lymph node metastasis. The high mitosis/low Ki-67-positive cells tumours showed a significant association with variables of poor prognosis, including high-grade and triple-negative subtypes. CONCLUSION The proportion of BC cells in the cell cycle and mitosis is variable. We show that not only the number of cells in the cell cycle or mitosis, but also the difference between them, provides valuable information on tumour aggressiveness.
Collapse
Affiliation(s)
- Ayat G Lashen
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK.,Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Michael S Toss
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ayaka Katayama
- Diagnostic Pathology, Gunma University Graduate School of Medicine, Maebaashi, Japan
| | - Rajan Gogna
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| | - Nigel P Mongan
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK.,School of Veterinary Medicine and Sciences, University of Nottingham, Nottingham, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
14
|
How the variability between computer-assisted analysis procedures evaluating immune markers can influence patients' outcome prediction. Histochem Cell Biol 2021; 156:461-478. [PMID: 34383240 DOI: 10.1007/s00418-021-02022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Differences between computer-assisted image analysis (CAI) algorithms may cause discrepancies in the identification of immunohistochemically stained immune biomarkers in biopsies of breast cancer patients. These discrepancies have implications for their association with disease outcome. This study aims to compare three CAI procedures (A, B and C) to measure positive marker areas in post-neoadjuvant chemotherapy biopsies of patients with triple-negative breast cancer (TNBC) and to explore the differences in their performance in determining the potential association with relapse in these patients. A total of 3304 digital images of biopsy tissue obtained from 118 TNBC patients were stained for seven immune markers using immunohistochemistry (CD4, CD8, FOXP3, CD21, CD1a, CD83, HLA-DR) and were analyzed with procedures A, B and C. The three methods measure the positive pixel markers in the total tissue areas. The extent of agreement between paired CAI procedures, a principal component analysis (PCA) and Cox multivariate analysis was assessed. Comparisons of paired procedures showed close agreement for most of the immune markers at low concentration. The probability of differences between the paired procedures B/C and B/A was generally higher than those observed in C/A. The principal component analysis, largely based on data from CD8, CD1a and HLA-DR, identified two groups of patients with a significantly lower probability of relapse than the others. The multivariate regression models showed similarities in the factors associated with relapse for procedures A and C, as opposed to those obtained with procedure B. General agreement among the results of CAI procedures would not guarantee that the same predictive breast cancer markers were consistently identified. These results highlight the importance of developing additional strategies to improve the sensitivity of CAI procedures.
Collapse
|
15
|
Detection of Ki67 Hot-Spots of Invasive Breast Cancer Based on Convolutional Neural Networks Applied to Mutual Information of H&E and Ki67 Whole Slide Images. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10217761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ki67 hot-spot detection and its evaluation in invasive breast cancer regions play a significant role in routine medical practice. The quantification of cellular proliferation assessed by Ki67 immunohistochemistry is an established prognostic and predictive biomarker that determines the choice of therapeutic protocols. In this paper, we present three deep learning-based approaches to automatically detect and quantify Ki67 hot-spot areas by means of the Ki67 labeling index. To this end, a dataset composed of 100 whole slide images (WSIs) belonging to 50 breast cancer cases (Ki67 and H&E WSI pairs) was used. Three methods based on CNN classification were proposed and compared to create the tumor proliferation map. The best results were obtained by applying the CNN to the mutual information acquired from the color deconvolution of both the Ki67 marker and the H&E WSIs. The overall accuracy of this approach was 95%. The agreement between the automatic Ki67 scoring and the manual analysis is promising with a Spearman’s ρ correlation of 0.92. The results illustrate the suitability of this CNN-based approach for detecting hot-spots areas of invasive breast cancer in WSI.
Collapse
|
16
|
Levy-Jurgenson A, Tekpli X, Kristensen VN, Yakhini Z. Spatial transcriptomics inferred from pathology whole-slide images links tumor heterogeneity to survival in breast and lung cancer. Sci Rep 2020; 10:18802. [PMID: 33139755 PMCID: PMC7606448 DOI: 10.1038/s41598-020-75708-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Digital analysis of pathology whole-slide images is fast becoming a game changer in cancer diagnosis and treatment. Specifically, deep learning methods have shown great potential to support pathology analysis, with recent studies identifying molecular traits that were not previously recognized in pathology H&E whole-slide images. Simultaneous to these developments, it is becoming increasingly evident that tumor heterogeneity is an important determinant of cancer prognosis and susceptibility to treatment, and should therefore play a role in the evolving practices of matching treatment protocols to patients. State of the art diagnostic procedures, however, do not provide automated methods for characterizing and/or quantifying tumor heterogeneity, certainly not in a spatial context. Further, existing methods for analyzing pathology whole-slide images from bulk measurements require many training samples and complex pipelines. Our work addresses these two challenges. First, we train deep learning models to spatially resolve bulk mRNA and miRNA expression levels on pathology whole-slide images (WSIs). Our models reach up to 0.95 AUC on held-out test sets from two cancer cohorts using a simple training pipeline and a small number of training samples. Using the inferred gene expression levels, we further develop a method to spatially characterize tumor heterogeneity. Specifically, we produce tumor molecular cartographies and heterogeneity maps of WSIs and formulate a heterogeneity index (HTI) that quantifies the level of heterogeneity within these maps. Applying our methods to breast and lung cancer slides, we show a significant statistical link between heterogeneity and survival. Our methods potentially open a new and accessible approach to investigating tumor heterogeneity and other spatial molecular properties and their link to clinical characteristics, including treatment susceptibility and survival.
Collapse
Affiliation(s)
- Alona Levy-Jurgenson
- Department of Computer Science, Technion - Israel Institute of Technology, Haifa, 32000, Israel.
| | - Xavier Tekpli
- Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310, Oslo, Norway
| | - Vessela N Kristensen
- Department of Medical Genetics, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 0310, Oslo, Norway
- Division of Medicine, Department of Clinical Molecular Biology and Laboratory Science (EpiGen), Akershus University Hospital, Lørenskog, Norway
| | - Zohar Yakhini
- Department of Computer Science, Technion - Israel Institute of Technology, Haifa, 32000, Israel.
- Interdisciplinary Center, Arazi School of Computer Science, Herzliya, 4610101, Israel.
| |
Collapse
|
17
|
Report From the International Society of Urological Pathology (ISUP) Consultation Conference on Molecular Pathology of Urogenital Cancers. I. Molecular Biomarkers in Prostate Cancer. Am J Surg Pathol 2020; 44:e15-e29. [PMID: 32044806 DOI: 10.1097/pas.0000000000001450] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The combined clinical and molecular heterogeneity of prostate cancer necessitates the use of prognostic, predictive, and diagnostic biomarkers to assist the clinician with treatment selection. The pathologist plays a critical role in guiding molecular biomarker testing in prostate cancer and requires a thorough knowledge of the current testing options. In the setting of clinically localized prostate cancer, prognostic biomarkers such as Ki-67 labeling, PTEN loss or mRNA-based genomic signatures can be useful to help determine whether definitive therapy is required. In the setting of advanced disease, predictive biomarkers, such as the presence of DNA repair deficiency mediated by BRCA2 loss or mismatch repair gene defects, may suggest the utility of poly-ADP ribosylase inhibition or immune checkpoint blockade. Finally, androgen receptor-related biomarkers or diagnostic biomarkers indicating the presence of small cell neuroendocrine prostate cancer may help guide the use of androgen receptor signaling inhibitors and chemotherapy. In this review, we examine the current evidence for several prognostic, predictive and diagnostic tissue-based molecular biomarkers in prostate cancer management. For each assay, we summarize a recent survey of the International Society of Urology Pathology (ISUP) members on current testing practices and include recommendations for testing that emerged from the ISUP Working Group on Molecular Pathology of Prostate Cancer and the 2019 Consultation Conference on Molecular Pathology of Urogenital Cancers.
Collapse
|
18
|
Tendl-Schulz KA, Rössler F, Wimmer P, Heber UM, Mittlböck M, Kozakowski N, Pinker K, Bartsch R, Dubsky P, Fitzal F, Filipits M, Eckel FC, Langthaler EM, Steger G, Gnant M, Singer CF, Helbich TH, Bago-Horvath Z. Factors influencing agreement of breast cancer luminal molecular subtype by Ki67 labeling index between core needle biopsy and surgical resection specimens. Virchows Arch 2020; 477:545-555. [PMID: 32383007 PMCID: PMC7508960 DOI: 10.1007/s00428-020-02818-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/20/2020] [Accepted: 04/16/2020] [Indexed: 11/09/2022]
Abstract
Reliable determination of Ki67 labeling index (Ki67-LI) on core needle biopsy (CNB) is essential for determining breast cancer molecular subtype for therapy planning. However, studies on agreement between molecular subtype and Ki67-LI between CNB and surgical resection (SR) specimens are conflicting. The present study analyzed the influence of clinicopathological and sampling-associated factors on agreement. Molecular subtype was determined visually by Ki67-LI in 484 pairs of CNB and SR specimens of invasive estrogen receptor (ER)-positive, human epidermal growth factor (HER2)-negative breast cancer. Luminal B disease was defined by Ki67-LI > 20% in SR. Correlation of molecular subtype agreement with age, menopausal status, CNB method, Breast Imaging Reporting and Data System imaging category, time between biopsies, type of surgery, and pathological tumor parameters was analyzed. Recurrence-free survival (RFS) and overall survival (OS) were analyzed using the Kaplan-Meier method. CNB had a sensitivity of 77.95% and a specificity of 80.97% for identifying luminal B tumors in CNB, compared with the final molecular subtype determination after surgery. The correlation of Ki67-LI between CNB and SR was moderate (ROC-AUC 0.8333). Specificity and sensitivity for CNB to correctly define molecular subtype of tumors according to SR were significantly associated with tumor grade, immunohistochemical progesterone receptor (PR) and p53 expression (p < 0.05). Agreement of molecular subtype did not significantly impact RFS and OS (p = 0.22 for both). The identified factors likely mirror intratumoral heterogeneity that might compromise obtaining a representative CNB. Our results challenge the robustness of a single CNB-driven measurement of Ki67-LI to identify luminal B breast cancer of low (G1) or intermediate (G2) grade.
Collapse
Affiliation(s)
- Kristina A Tendl-Schulz
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Fabian Rössler
- Department of Surgery and Transplantation, University Hospital and University of Zurich, Zurich, Switzerland
| | - Philipp Wimmer
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Ulrike M Heber
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Martina Mittlböck
- Center for Medical Statistics, Informatics, and Intelligent Systems, Section for Clinical Biometrics, Medical University of Vienna, Vienna, Austria
| | - Nicolas Kozakowski
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Katja Pinker
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Vienna, Austria
- Department of Radiology, Breast Imaging Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rupert Bartsch
- Department for Medicine I/Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Peter Dubsky
- Department of Surgery and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Hirslanden Klinik St. Anna Brustzentrum, Lucerne, Switzerland
| | - Florian Fitzal
- Department of Surgery and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Martin Filipits
- Institute of Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Fanny Carolina Eckel
- Department of Surgery and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Eva-Maria Langthaler
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria
| | - Günther Steger
- Department for Medicine I/Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christian F Singer
- Department of Obstetrics and Gynaecology and Breast Health Center, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-guided Therapy, Division of Molecular and Gender Imaging, Medical University of Vienna, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Department of Pathology and Comprehensive Cancer Center, Medical University of Vienna, 18-20 Waehringer Guertel, A-1090, Vienna, Austria.
| |
Collapse
|
19
|
Zilenaite D, Rasmusson A, Augulis R, Besusparis J, Laurinaviciene A, Plancoulaine B, Ostapenko V, Laurinavicius A. Independent Prognostic Value of Intratumoral Heterogeneity and Immune Response Features by Automated Digital Immunohistochemistry Analysis in Early Hormone Receptor-Positive Breast Carcinoma. Front Oncol 2020; 10:950. [PMID: 32612954 PMCID: PMC7308549 DOI: 10.3389/fonc.2020.00950] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 05/14/2020] [Indexed: 12/11/2022] Open
Abstract
Immunohistochemistry (IHC) for ER, PR, HER2, and Ki67 is used to predict outcome and therapy response in breast cancer patients. The current IHC assessment, visual or digital, is based mostly on global biomarker expression levels in the tissue sample. In our study, we explored the prognostic value of digital image analysis of conventional breast cancer IHC biomarkers supplemented with their intratumoral heterogeneity and tissue immune response indicators. Surgically excised tumor samples from 101 female patients with hormone receptor-positive breast cancer (HRBC) were stained for ER, PR, HER2, Ki67, SATB1, CD8, and scanned at 20x. Digital image analysis was performed using the HALO™ platform. Subsequently, hexagonal tiling was used to compute intratumoral heterogeneity indicators for ER, PR and Ki67 expression. Multiple Cox regression analysis revealed three independent predictors of the patient's overall survival: Haralick's texture entropy of PR (HR = 0.19, p = 0.0005), Ki67 Ashman's D bimodality (HR = 3.0, p = 0.01), and CD8+SATB1+ cell density in tumor tissue (HR = 0.32, p = 0.02). Remarkably, the PR and Ki67 intratumoral heterogeneity indicators were prognostically more informative than the rates of their expression. In particular, a distinct non-linear relationship between the rate of PR expression and its intratumoral heterogeneity was observed and revealed a non-linear prognostic effect of PR expression. The independent prognostic significance of CD8+SATB1+ cells infiltrating the tumor could indicate their role in anti-tumor immunity. In conclusion, we suggest that prognostic modeling, based entirely on the computational image-based IHC biomarkers, is possible in HRBC patients. The intratumoral heterogeneity and immune response indicators outperformed both conventional breast cancer IHC and clinicopathological variables while markedly increasing the power of the model.
Collapse
Affiliation(s)
- Dovile Zilenaite
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,National Centre of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Allan Rasmusson
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,National Centre of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Renaldas Augulis
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,National Centre of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Justinas Besusparis
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,National Centre of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Aida Laurinaviciene
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,National Centre of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| | - Benoit Plancoulaine
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,ANTICIPE, Inserm (UMR 1086), Cancer Center F. Baclesse, Normandy University, Caen, France
| | - Valerijus Ostapenko
- Department of Breast Surgery and Oncology, National Cancer Institute, Vilnius, Lithuania
| | - Arvydas Laurinavicius
- Department of Pathology, Forensic Medicine and Pharmacology, Faculty of Medicine, Institute of Biomedical Sciences, Vilnius University, Vilnius, Lithuania.,National Centre of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, Vilnius, Lithuania
| |
Collapse
|
20
|
Practical approaches to automated digital image analysis of Ki-67 labeling index in 997 breast carcinomas and causes of discordance with visual assessment. PLoS One 2019; 14:e0212309. [PMID: 30785924 PMCID: PMC6382355 DOI: 10.1371/journal.pone.0212309] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/31/2019] [Indexed: 12/20/2022] Open
Abstract
The Ki-67 labeling index (LI) is an important prognostic factor in breast carcinoma. The Ki-67 LI is traditionally calculated via unaided microscopic estimation; however, inter-observer and intra-observer variability and low reproducibility are problems with this visual assessment (VA) method. For more accurate assessment and better reproducibility with Ki-67 LI, digital image analysis was introduced recently. We used both VA and automated digital image analysis (ADIA) (Ventana Virtuoso image management software) to estimate Ki-67 LI for 997 cases of breast carcinoma, and compared VA and ADIA results. VA and ADIA were highly correlated (intraclass correlation coefficient 0.982, and Spearman’s correlation coefficient 0.966, p<0.05). We retrospectively analyzed cases with a greater than 5% difference between VA and ADIA results. The cause of these differences was: (1) tumor heterogeneity (98 cases, 56.0%), (2) VA interpretation error (32 cases, 18.3%), (3) misidentification of tumor cells (26 cases, 14.9%), (4) poor immunostaining or slide quality (16 cases, 9.1%), and (5) Estimation of non-tumor cells (3 cases, 1.7%). There were more discrepancies between VA and ADIA results in the group with a VA value of 10–20% compared to groups with <10% and ≥20%. Although ADIA is more accurate than VA, there are some limitations. Therefore, ADIA findings require confirmation by a pathologist.
Collapse
|
21
|
Allison KH. Ancillary Prognostic and Predictive Testing in Breast Cancer: Focus on Discordant, Unusual, and Borderline Results. Surg Pathol Clin 2018; 11:147-176. [PMID: 29413654 DOI: 10.1016/j.path.2017.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Ancillary testing in breast cancer has become standard of care to determine what therapies may be most effective for individual patients with breast cancer. Single-marker tests are required on all newly diagnosed and newly metastatic breast cancers. Markers of proliferation are also used, and include both single-marker tests like Ki67 as well as panel-based gene expression tests, which have made more recent contributions to prognostic and predictive testing in breast cancers. This review focuses on pathologist interpretation of these ancillary test results, with a focus on expected versus unexpected results and troubleshooting borderline, unusual, or discordant results.
Collapse
Affiliation(s)
- Kimberly H Allison
- Department of Pathology, Stanford University School of Medicine, 300 Pasteur Drive, Lane 235, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Hsieh AMY, Polyakova O, Fu G, Chazen RS, MacMillan C, Witterick IJ, Ralhan R, Walfish PG. Programmed death-ligand 1 expression by digital image analysis advances thyroid cancer diagnosis among encapsulated follicular lesions. Oncotarget 2018; 9:19767-19782. [PMID: 29731981 PMCID: PMC5929424 DOI: 10.18632/oncotarget.24833] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 02/24/2018] [Indexed: 01/09/2023] Open
Abstract
Recognition of noninvasive follicular thyroid neoplasms with papillary-like nuclear features (NIFTP) that distinguishes them from invasive malignant encapsulated follicular variant of papillary thyroid carcinoma (EFVPTC) can prevent overtreatment of NIFTP patients. We and others have previously reported that programmed death-ligand 1 (PD-L1) is a useful biomarker in thyroid tumors; however, all reports to date have relied on manual scoring that is time consuming as well as subject to individual bias. Consequently, we developed a digital image analysis (DIA) protocol for cytoplasmic and membranous stain quantitation (ThyApp) and evaluated three tumor sampling methods [Systemic Uniform Random Sampling, hotspot nucleus, and hotspot nucleus/3,3'-Diaminobenzidine (DAB)]. A patient cohort of 153 cases consisting of 48 NIFTP, 44 EFVPTC, 26 benign nodules and 35 encapsulated follicular lesions/neoplasms with lymphocytic thyroiditis (LT) was studied. ThyApp quantitation of PD-L1 expression revealed a significant difference between invasive EFVPTC and NIFTP; but none between NIFTP and benign nodules. ThyApp integrated with hotspot nucleus tumor sampling method demonstrated to be most clinically relevant, consumed least processing time, and eliminated interobserver variance. In conclusion, the fully automatic DIA algorithm developed using a histomorphological approach objectively quantitated PD-L1 expression in encapsulated thyroid neoplasms and outperformed manual scoring in reproducibility and higher efficiency.
Collapse
Affiliation(s)
- Anne M-Y Hsieh
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada
| | - Olena Polyakova
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada
| | - Guodong Fu
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada
| | - Ronald S Chazen
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada
| | - Christina MacMillan
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ian J Witterick
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada.,Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Sinai Health System, Toronto, Ontario, Canada.,Department of Otolaryngology-Head and Neck Surgery, Sinai Health System, Toronto, Ontario, Canada
| | - Ranju Ralhan
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada.,Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Otolaryngology-Head and Neck Surgery, Sinai Health System, Toronto, Ontario, Canada
| | - Paul G Walfish
- Alex and Simona Shnaider Research Laboratory in Molecular Oncology, Sinai Health System, Toronto, ON, Canada.,Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Sinai Health System, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Otolaryngology-Head and Neck Surgery, Sinai Health System, Toronto, Ontario, Canada.,Department of Medicine, Endocrine Division, Sinai Health System and University of Toronto Medical School, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Koopman T, Buikema HJ, Hollema H, de Bock GH, van der Vegt B. Digital image analysis of Ki67 proliferation index in breast cancer using virtual dual staining on whole tissue sections: clinical validation and inter-platform agreement. Breast Cancer Res Treat 2018; 169:33-42. [PMID: 29349710 PMCID: PMC5882622 DOI: 10.1007/s10549-018-4669-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/13/2018] [Indexed: 12/14/2022]
Abstract
Purpose The Ki67 proliferation index is a prognostic and predictive marker in breast cancer. Manual scoring is prone to inter- and intra-observer variability. The aims of this study were to clinically validate digital image analysis (DIA) of Ki67 using virtual dual staining (VDS) on whole tissue sections and to assess inter-platform agreement between two independent DIA platforms. Methods Serial whole tissue sections of 154 consecutive invasive breast carcinomas were stained for Ki67 and cytokeratin 8/18 with immunohistochemistry in a clinical setting. Ki67 proliferation index was determined using two independent DIA platforms, implementing VDS to identify tumor tissue. Manual Ki67 score was determined using a standardized manual counting protocol. Inter-observer agreement between manual and DIA scores and inter-platform agreement between both DIA platforms were determined and calculated using Spearman’s correlation coefficients. Correlations and agreement were assessed with scatterplots and Bland–Altman plots. Results Spearman’s correlation coefficients were 0.94 (p < 0.001) for inter-observer agreement between manual counting and platform A, 0.93 (p < 0.001) between manual counting and platform B, and 0.96 (p < 0.001) for inter-platform agreement. Scatterplots and Bland–Altman plots revealed no skewness within specific data ranges. In the few cases with ≥ 10% difference between manual counting and DIA, results by both platforms were similar. Conclusions DIA using VDS is an accurate method to determine the Ki67 proliferation index in breast cancer, as an alternative to manual scoring of whole sections in clinical practice. Inter-platform agreement between two different DIA platforms was excellent, suggesting vendor-independent clinical implementability. Electronic supplementary material The online version of this article (10.1007/s10549-018-4669-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Timco Koopman
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Henk J Buikema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Harry Hollema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, PO Box 30001, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
24
|
Large-scale DNA organization is a prognostic marker of breast cancer survival. Med Oncol 2017; 35:9. [PMID: 29214466 DOI: 10.1007/s12032-017-1068-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 11/30/2017] [Indexed: 01/22/2023]
Abstract
Breast cancer is the leading cause of cancer-related deaths among women worldwide. We investigated whether changes in large-scale DNA organization (LDO) of tumor epithelial nuclei are an indicator of the aggressiveness of the tumor. We tested our algorithm on a set of 172 duplicates TMA cores samples coming from 95 breast cancer patients. Thirty-five patients died of breast cancer, and 60 were still alive 10 years after surgery. Duplicates cores were used to create training and test set. The TMA slides were stained with Feulgen-thionin and imaged using our in-house high-resolution Imaging system. Automated segmentation of cell nuclei followed by manual selection of intact, in-focus nuclei resulted in an average of 50 cell nuclei per sample available for analysis. Using forward stepwise linear discriminant analysis, a combination of six features that combined linearly gave the best discrimination between the two groups of cells: cells collected from 'deceased' patients TMA specimens and cells collected from "survivors" patients TMA specimens. Five of these features measure the spatial organization of DNA chromatin. The resulting canonical score is named cell LDO score. A patient LDO score, percentage of cell nuclei with a cell LDO score higher than a predefined cutoff value, was processed for the specimens in the test set, and a cutoff value was defined to classify patients with a low or a high LDO score. Using this binary test, 82.1% of patients were correctly classified are "deceased" or "survivors," with a specificity of 79% and a sensitivity of 88%. The relative risk of death of an individual with a high LDO score was nine times higher than for a patient with a low LDO score. When testing the combination of LDO score, node status, histological grade, and tumor grade to predict breast cancer survival, LDO was the most significant predictor. LDO classification was also highly associated with survival for only grade 1 and 2 patients as well as for only grade 3 patients. Our result confirms the potential of LDO to measure phenotypic changes associated with more aggressive disease and could be evaluated to identify patients more likely to benefit from adjuvant therapies.
Collapse
|
25
|
Graf JF, Zavodszky MI. Characterizing the heterogeneity of tumor tissues from spatially resolved molecular measures. PLoS One 2017; 12:e0188878. [PMID: 29190747 PMCID: PMC5708750 DOI: 10.1371/journal.pone.0188878] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 11/14/2017] [Indexed: 11/18/2022] Open
Abstract
Background Tumor heterogeneity can manifest itself by sub-populations of cells having distinct phenotypic profiles expressed as diverse molecular, morphological and spatial distributions. This inherent heterogeneity poses challenges in terms of diagnosis, prognosis and efficient treatment. Consequently, tools and techniques are being developed to properly characterize and quantify tumor heterogeneity. Multiplexed immunofluorescence (MxIF) is one such technology that offers molecular insight into both inter-individual and intratumor heterogeneity. It enables the quantification of both the concentration and spatial distribution of 60+ proteins across a tissue section. Upon bioimage processing, protein expression data can be generated for each cell from a tissue field of view. Results The Multi-Omics Heterogeneity Analysis (MOHA) tool was developed to compute tissue heterogeneity metrics from MxIF spatially resolved tissue imaging data. This technique computes the molecular state of each cell in a sample based on a pathway or gene set. Spatial states are then computed based on the spatial arrangements of the cells as distinguished by their respective molecular states. MOHA computes tissue heterogeneity metrics from the distributions of these molecular and spatially defined states. A colorectal cancer cohort of approximately 700 subjects with MxIF data is presented to demonstrate the MOHA methodology. Within this dataset, statistically significant correlations were found between the intratumor AKT pathway state diversity and cancer stage and histological tumor grade. Furthermore, intratumor spatial diversity metrics were found to correlate with cancer recurrence. Conclusions MOHA provides a simple and robust approach to characterize molecular and spatial heterogeneity of tissues. Research projects that generate spatially resolved tissue imaging data can take full advantage of this useful technique. The MOHA algorithm is implemented as a freely available R script (see supplementary information).
Collapse
Affiliation(s)
- John F. Graf
- GE Global Research, Niskayuna, New York, United States of America
- * E-mail: (JFG); (MIZ)
| | - Maria I. Zavodszky
- GE Global Research, Niskayuna, New York, United States of America
- * E-mail: (JFG); (MIZ)
| |
Collapse
|
26
|
Calin VL, Mihailescu M, Scarlat EI, Baluta AV, Calin D, Kovacs E, Savopol T, Moisescu MG. Evaluation of the metastatic potential of malignant cells by image processing of digital holographic microscopy data. FEBS Open Bio 2017; 7:1527-1538. [PMID: 28979841 PMCID: PMC5623698 DOI: 10.1002/2211-5463.12282] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/13/2017] [Accepted: 08/02/2017] [Indexed: 12/20/2022] Open
Abstract
The cell refractive index has been proposed as a putative cancer biomarker of great potential, being correlated with cell content and morphology, cell division rate and membrane permeability. We used digital holographic microscopy to compare the refractive index and dry mass density of two B16 murine melanoma sublines of different metastatic potential. Using statistical methods, the distribution of phase shifts within the reconstructed quantitative phase images was analyzed by the method of bimodality coefficients. The observed correlation of refractive index, dry mass density and bimodality profile with the metastatic potential of the cells was validated by real time impedance-based assay and clonogenic tests. We suggest that the refractive index and bimodality analysis of quantitative phase image histograms could be developed as optical biomarkers useful in label-free detection and quantitative evaluation of cell metastatic potential.
Collapse
Affiliation(s)
- Violeta L. Calin
- Biophysics and Cellular Biotechnology DepartmentFaculty of MedicineCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Mona Mihailescu
- Physics DepartmentFaculty of Applied SciencesPolitehnica University of BucharestRomania
| | - Eugen I. Scarlat
- Physics DepartmentFaculty of Applied SciencesPolitehnica University of BucharestRomania
| | - Alexandra V. Baluta
- Applied Electronics and Informatics Engineering DepartmentFaculty of ElectronicsTelecommunications and Information TechnologyPolitehnica University of BucharestRomania
| | - Daniel Calin
- Biophysics and Cellular Biotechnology DepartmentFaculty of MedicineCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Eugenia Kovacs
- Biophysics and Cellular Biotechnology DepartmentFaculty of MedicineCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Tudor Savopol
- Biophysics and Cellular Biotechnology DepartmentFaculty of MedicineCarol Davila University of Medicine and PharmacyBucharestRomania
| | - Mihaela G. Moisescu
- Biophysics and Cellular Biotechnology DepartmentFaculty of MedicineCarol Davila University of Medicine and PharmacyBucharestRomania
| |
Collapse
|
27
|
Automated Image Analysis of HER2 Fluorescence In Situ Hybridization to Refine Definitions of Genetic Heterogeneity in Breast Cancer Tissue. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2321916. [PMID: 28752092 PMCID: PMC5511668 DOI: 10.1155/2017/2321916] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/20/2017] [Accepted: 04/26/2017] [Indexed: 12/15/2022]
Abstract
Human epidermal growth factor receptor 2 gene- (HER2-) targeted therapy for breast cancer relies primarily on HER2 overexpression established by immunohistochemistry (IHC) with borderline cases being further tested for amplification by fluorescence in situ hybridization (FISH). Manual interpretation of HER2 FISH is based on a limited number of cells and rather complex definitions of equivocal, polysomic, and genetically heterogeneous (GH) cases. Image analysis (IA) can extract high-capacity data and potentially improve HER2 testing in borderline cases. We investigated statistically derived indicators of HER2 heterogeneity in HER2 FISH data obtained by automated IA of 50 IHC borderline (2+) cases of invasive ductal breast carcinoma. Overall, IA significantly underestimated the conventional HER2, CEP17 counts, and HER2/CEP17 ratio; however, it collected more amplified cells in some cases below the lower limit of GH definition by manual procedure. Indicators for amplification, polysomy, and bimodality were extracted by factor analysis and allowed clustering of the tumors into amplified, nonamplified, and equivocal/polysomy categories. The bimodality indicator provided independent cell diversity characteristics for all clusters. Tumors classified as bimodal only partially coincided with the conventional GH heterogeneity category. We conclude that automated high-capacity nonselective tumor cell assay can generate evidence-based HER2 intratumor heterogeneity indicators to refine GH definitions.
Collapse
|
28
|
Pilutti D, Della Mea V, Pegolo E, La Marra F, Antoniazzi F, Di Loreto C. An adaptive positivity thresholding method for automated Ki67 hotspot detection (AKHoD) in breast cancer biopsies. Comput Med Imaging Graph 2017; 61:28-34. [PMID: 28499621 DOI: 10.1016/j.compmedimag.2017.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 04/18/2017] [Accepted: 04/23/2017] [Indexed: 12/17/2022]
Abstract
The proliferative activity of breast cancer tissue can be estimated using the Ki67 biomarker. The percentage of positivity of such biomarker is correlated with proliferation and consequently with the prognosis of a breast tumor. Ki67 marked tissue samples are analyzed by an experienced pathologist who identifies the most active areas of tumor cell proliferation called hotspots, and estimates the positivity of each case. A method for the Automated Ki67 Hotspot Detection (AKHoD) is presented in this work. The main objective of the AKHoD method is to automatically and efficiently provide the pathologist with suggestions about Ki67 hotspot areas as a decision support. The input of AKHoD is a digital slide that is divided in tiles. For each tile, AKHoD provides a rough estimate of positivity and cellularity, summarized in very low resolution positivity and cellularity images. In a second step, an adaptive thresholding is applied to such positivity image to identify the most positive connected and convex areas, within cellularity limits set by current guidelines (that is, 500-2000). The method has been preliminarily validated on 50 digital slides for which three expert pathologists provided gold standard hotspots. 82% of the gold standard hotspots have been successfully recognized by the system, spending an average of 54s per slide. While further validation is needed taking into account also patients follow-up, this first experimentation suggests that the proposed method could be adequate for supporting the pathologist in hotspot detection.
Collapse
Affiliation(s)
- David Pilutti
- Department of Mathematics, Computer Science, and Physics, University of Udine, Udine, Italy.
| | - Vincenzo Della Mea
- Department of Mathematics, Computer Science, and Physics, University of Udine, Udine, Italy
| | - Enrico Pegolo
- Institute of Pathology, University Hospital "Santa Maria della Misericordia", Udine, Italy
| | - Francesco La Marra
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Fulvio Antoniazzi
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| | - Carla Di Loreto
- Department of Medical and Biological Science, University of Udine, Udine, Italy
| |
Collapse
|
29
|
Quality assurance trials for Ki67 assessment in pathology. Virchows Arch 2017; 471:501-508. [DOI: 10.1007/s00428-017-2142-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/21/2017] [Accepted: 05/01/2017] [Indexed: 12/20/2022]
|