1
|
Barroso VM, Weng Z, Glamann L, Bauer M, Wickenhauser C, Zander T, Büttner R, Quaas A, Tolkach Y. Artificial Intelligence-Based Single-Cell Analysis as a Next-Generation Histologic Grading Approach in Colorectal Cancer: Prognostic Role and Tumor Biology Assessment. Mod Pathol 2025; 38:100771. [PMID: 40222652 DOI: 10.1016/j.modpat.2025.100771] [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: 11/06/2024] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
The management of colorectal carcinoma (CRC) relies on pathological interpretation. Digital pathology approaches allow for development of new potent artificial intelligence-based prognostic parameters. The study aimed to develop an artificial intelligence-based image analysis platform allowing fully automatized, quantitative, and explainable tumor microenvironment analysis and extraction of prognostic information from hematoxylin and eosin-stained whole-slide images of CRC patients. Three well--characterized, multi-institutional patient cohorts were included (patient n = 1438, whole-slide image n > 2400). The developed image analysis platform implements quality control and established algorithms to segment tissue and detect cell types. It enabled systematic analysis of immune infiltrate, assessing its prognostic relevance, intratumoral heterogeneity, and biological concepts across multiple survival end points. Analyzing single-cell types and their combinations reveals independent, prognostic parameters, highlighting significant intratumoral heterogeneity, especially in the biopsy setting, which must be accounted for. A key morphologic concept related to tumor control by the immune system is described, resulting in a capable, independent prognostic parameter (tumor "out of control"). Our findings have direct clinical implications and can be used as a foundation for updating the existing CRC grading systems.
Collapse
Affiliation(s)
- Vincenzo Mitchell Barroso
- Medical Faculty, University of Cologne, Cologne, Germany; Insitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Zhilong Weng
- Insitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Lennert Glamann
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Salle), Germany
| | - Marcus Bauer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Salle), Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Salle), Germany
| | - Thomas Zander
- Clinic of Internal Medicine, Oncology, University Hospital Cologne, Cologne, Germany
| | - Reinhard Büttner
- Insitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Alexander Quaas
- Insitute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Yuri Tolkach
- Insitute of Pathology, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
2
|
Zhang Y, Abousamra S, Hasan M, Torre-Healy L, Krichevsky S, Shrestha S, Bremer E, Oldridge DA, Rech AJ, Furth EE, Bocklage TJ, Levens JS, Hands I, Durbin EB, Samaras D, Kurc T, Saltz JH, Gupta R. Pathomics Image Analysis of Tumor Infiltrating Lymphocytes (TILs) in Colon Cancer. RESEARCH SQUARE 2025:rs.3.rs-6173056. [PMID: 40235501 PMCID: PMC11998795 DOI: 10.21203/rs.3.rs-6173056/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
We developed a deep learning Pathomics image analysis workflow to generate spatial Tumor-TIL maps to visualize and quantify the abundance and spatial distribution of tumor infiltrating lymphocytes (TILs) in colon cancer. Colon cancer and lymphocyte detection in hematoxylin and eosin (H&E) stained whole slide images (WSIs) has revealed complex immuno-oncologic interactions that form TIL-rich and TIL-poor tumor habitats, which are unique in each patient sample. We compute Tumor%, total lymphocyte%, and TILs% as the proportion of the colon cancer microenvironment occupied by intratumoral lymphocytes for each WSI. Kaplan-Meier survival analyses and multivariate Cox regression were utilized to evaluate the prognostic significance of TILs% as a Pathomics biomarker. High TILs% was associated with improved overall survival (OS) and progression-free interval (PFI) in localized and metastatic colon cancer and other clinicopathologic variables, supporting the routine use of Pathomics Tumor-TIL mapping in biomedical research, clinical trials, laboratory medicine, and precision oncology.
Collapse
|
3
|
Zhang H, Chen L, Li L, Liu Y, Das B, Zhai S, Tan J, Jiang Y, Turco S, Yao Y, Frishman D. Prediction and analysis of tumor infiltrating lymphocytes across 28 cancers by TILScout using deep learning. NPJ Precis Oncol 2025; 9:76. [PMID: 40108446 PMCID: PMC11923303 DOI: 10.1038/s41698-025-00866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
The density of tumor-infiltrating lymphocytes (TILs) serves as a valuable indicator for predicting anti-tumor responses, but its broad impact across various types of cancers remains underexplored. We introduce TILScout, a pan-cancer deep-learning approach to compute patch-level TIL scores from whole slide images (WSIs). TILScout achieved accuracies of 0.9787 and 0.9628, and AUCs of 0.9988 and 0.9934 in classifying WSI patches into three categories-TIL-positive, TIL-negative, and other/necrotic-on validation and independent test sets, respectively, surpassing previous studies. The biological significance of TILScout-derived TIL scores across 28 cancers was validated through comprehensive functional and correlational analyses. A consistent decrease in TIL scores with an increase in cancer stage provides direct evidence that the lower TIL content may stimulate cancer progression. Additionally, TIL scores correlated with immune checkpoint gene expression and genomic variation in common cancer driver genes. Our comprehensive pan-cancer survey highlights the critical prognostic significance of TILs within the tumor microenvironment.
Collapse
Affiliation(s)
- Huibo Zhang
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Barnali Das
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Shuang Zhai
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Jiang
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Simona Turco
- Electrical Engineering, Eindhoven University of Technology, Den Dolech 12, Eindhoven, 5612AZ, the Netherlands
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Dmitrij Frishman
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
4
|
Millward J, He Z, Nibali A, Mouradov D, Mielke LA, Tran K, Chou A, Hawkins NJ, Ward RL, Gill AJ, Sieber OM, Williams DS. Automated deep learning-based assessment of tumour-infiltrating lymphocyte density determines prognosis in colorectal cancer. J Transl Med 2025; 23:298. [PMID: 40065354 PMCID: PMC11892243 DOI: 10.1186/s12967-025-06254-3] [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: 04/19/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The presence of tumour-infiltrating lymphocytes (TILs) is a well-established prognostic biomarker across multiple cancer types, with higher TIL counts being associated with lower recurrence rates and improved patient survival. We aimed to examine whether an automated intraepithelial TIL (iTIL) assessment could stratify patients by risk, with the ability to generalise across independent patient cohorts, using routine H&E slides of colorectal cancer (CRC). To our knowledge, no other existing fully automated iTIL system has demonstrated this capability. METHODS An automated method employing deep neural networks was developed to enumerate iTILs in H&E slides of CRC. The method was applied to a Stage III discovery cohort (n = 353) to identify an optimal threshold of 17 iTILs per-mm2 tumour for stratifying relapse-free survival. Using this threshold, patients from two independent Stage II-III validation cohorts (n = 1070, n = 885) were classified as "TIL-High" or "TIL-Low". RESULTS Significant stratification was observed in terms of overall survival for a combined validation cohort univariate (HR 1.67, 95%CI 1.39-2.00; p < 0.001) and multivariate (HR 1.37, 95%CI 1.13-1.66; p = 0.001) analysis. Our iTIL classifier was an independent prognostic factor within proficient DNA mismatch repair (pMMR) Stage II CRC cases with clinical high-risk features. Of these, those classified as TIL-High had outcomes similar to pMMR clinical low risk cases, and those classified TIL-Low had significantly poorer outcomes (univariate HR 2.38, 95%CI 1.57-3.61; p < 0.001, multivariate HR 2.17, 95%CI 1.42-3.33; p < 0.001). CONCLUSIONS Our deep learning method is the first fully automated system to stratify patient outcome by analysing TILs in H&E slides of CRC, that has shown generalisation capabilities across multiple independent cohorts.
Collapse
Affiliation(s)
- Joshua Millward
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia.
| | - Zhen He
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia
| | - Aiden Nibali
- School of Computing, Engineering and Mathematical Sciences, La Trobe University, Melbourne, Australia
| | - Dmitri Mouradov
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
| | - Lisa A Mielke
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Melbourne, Australia
| | - Kelly Tran
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Melbourne, Australia
| | - Angela Chou
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | | | - Robyn L Ward
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Anthony J Gill
- Department of Anatomical Pathology, NSW Health Pathology, Royal North Shore Hospital, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, Australia
| | - Oliver M Sieber
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, Australia
- Department of Surgery, The University of Melbourne, Melbourne, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - David S Williams
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
- La Trobe University School of Cancer Medicine, Melbourne, Australia
- Department of Anatomical Pathology, Austin Health, Melbourne, Australia
| |
Collapse
|
5
|
Kiraz U, Rewcastle E, Pettersen KB, Abono DM, Raghe SH, Gudlaugsson EG, Baak JPA, Janssen EAM. In triple-negative breast cancer, fibrotic focus, the mitotic activity index and tumour-infiltrating lymphocytes have independent prognostic value: an observational population-based cohort study with very long follow-up. J Clin Pathol 2025:jcp-2024-209855. [PMID: 39965885 DOI: 10.1136/jcp-2024-209855] [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: 09/10/2024] [Accepted: 01/23/2025] [Indexed: 02/20/2025]
Abstract
AIMS Triple-negative breast cancer (TNBC) is prognostically and therapeutically heterogeneous. The mitotic activity index (MAI) and fibrotic focus (FF) have been established as predictors in non-TNBC but not in TNBC. Late distant metastases occur in TNBC, but previous studies had short follow-up. High stromal tumour-infiltrating lymphocytes (sTILs) are prognostically favourable, but prognostic sTILs-thresholds are not well assessed. We evaluated prognostic/predictive characteristics in an observational population-based cohort of 231 consecutive TNBC patients with long follow-up. METHODS MAI, FF, sTILs and other characteristics were analysed with standard receiver operating characteristic curve analysis, percentile-derived prognostic thresholds, univariate and multivariate survival methods. A TNBC index and decision tree were assessed for distant metastasis-free survival. RESULTS Long follow-up was decisive: 7% of patients developed late distant metastases. In agreement with the aggressive nature of TNBC, the strongest prognostic MAI-threshold was 5 (p=0.001), lower than that for non-TNBC phenotypes. Lymph-node (LN) status (p=0.0003), FF (p=0.002), MAI5 (p=0.009) and sTILs (threshold 40%, p=0.003) were multivariable based significant and independent prognosticators, but no other characteristics (age, tumour size and grade). LN status was the strongest prognosticator, followed by FF, MAI5 and sTILs40. Subgroup analyses of patients undergoing adjuvant chemotherapy (ACT) showed that only FF and sTILs had significant prognostic value, while LN-positivity and the combination of LN-positivity and MAI≥5 could be a predictive factor for ACT outcome. CONCLUSIONS LN status, MAI5, FF and sTILs40 are prognostic factors in TNBC patients. In TNBC patients who have undergone ACT, the combination of LN-positivity and MAI5 is predictive for response to treatment.
Collapse
Affiliation(s)
- Umay Kiraz
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Emma Rewcastle
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | | | - Desmond M Abono
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Sadia H Raghe
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | | | - Jan P A Baak
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
| | - Emilius A M Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| |
Collapse
|
6
|
Skok K, Bräutigam K. Tumor infiltrating lymphocytes (TILs) - Pathologia, quo vadis? - A global survey. Pathol Res Pract 2025; 266:155775. [PMID: 39700663 DOI: 10.1016/j.prp.2024.155775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Tumor-infiltrating lymphocytes (TILs) and the tumor microenvironment have become increasingly important in cancer research, and immunotherapy has achieved major breakthroughs in improving patient outcomes. Despite the significant role of the pathologist in identifying, subtyping and reporting TILs, the implementation and assessment of TILs in pathology routine remains vague. To assess the actual use of TILs in routine clinical practice, a formal standardized questionnaire was disseminated on two social media platforms ("X" and LinkedIn) and by email in June 2024. Based on the results, we conducted a literature review on TILs via Medline/Pubmed in the two most scored and reported entities, namely malignant melanoma and colorectal cancer (CRC). 77 participants from 24 different countries around the world, mostly pathologists (n = 63, 82.0 %), completed the survey. More than half of the participants do not assess or report TILs in their daily (clinical) practice, a trend consistent across the countries included in the study. A variety of methods are used to report TILs, ranging from Artificial Intelligence (AI)-based scoring algorithms to quantification by eyeballing. Despite recognizing the importance of TIL assessment in clinical routine, many participants find it time-consuming and express a strong preference for AI-based quantification. Our survey reflects the perspective of mostly early career pathologists who recognize the importance of TILs in cancer but face challenges in implementation. The development of AI tools and consensus guidelines could alleviate these barriers. In addition, increasing the visibility and understanding of the role of pathologists within the medical community remains critical.
Collapse
Affiliation(s)
- Kristijan Skok
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Stiftingtalstraße 6, Graz 8010, Austria; Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Taborska Ulica 8, Maribor 2000, Slovenia
| | - Konstantin Bräutigam
- Centre for Evolution and Cancer, Institute of Cancer Research, London, SM2 5NG, United Kingdom.
| |
Collapse
|
7
|
Grindrod N, Cecchini M, Brackstone M. Concurrent Neoadjuvant Chemotherapy and Radiation in Locally Advanced Breast Cancer: Impact on Locoregional Recurrence Rates. Curr Oncol 2025; 32:85. [PMID: 39996885 PMCID: PMC11854545 DOI: 10.3390/curroncol32020085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Neoadjuvant chemoradiation therapy (NCRT) is an underutilized treatment in breast cancer but may improve outcomes by impacting the tumor immune microenvironment. The aim of this study was to evaluate NCRT's impact on recurrence and the role of tumor-infiltrating lymphocytes (TILs) in treatment response. We hypothesized that NCRT reduces recurrence by upregulating TILs. Patients with locally advanced breast cancer (LABC) were treated with NCRT. Stage IIB to III patients with any molecular subtypes were eligible. The patients were matched for age, stage, and molecular subtype by a propensity score to a concurrent cohort receiving standard neoadjuvant chemotherapy (NCT) followed by adjuvant radiation. The objective of this study was to assess the patients in terms of the pathological complete response (pCR), TIL counts prior to and following treatment, and locoregional recurrence. The median follow-up was 7.2 years. Thirty NCRT patients were successfully matched 1:3 to ninety NCT patients. The NCRT cohort had no regional and locoregional recurrences (p = 0.036, (hazard ratio) HR [0.25], 95% confidence interval (CI) [0.06-0.94] and p = 0.013, HR [0.25], 95% CI [0.08-0.76], respectively), compared to 17.8% of the NCT cohort. The NCRT group had significantly more pCRs, and TILs were increased in the post-treatment pCR specimens. NCRT can improve outcomes in LABC patients, with a higher pCR and significantly lower locoregional recurrence/higher recurrence-free survival. Further trials are needed to evaluate the role of NCRT in all breast cancer patients.
Collapse
Affiliation(s)
- Natalie Grindrod
- Schulich Faculty of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (N.G.); (M.C.)
- Department of Pathology, London Health Sciences Centre, London, ON N6A 5A5, Canada
| | - Matthew Cecchini
- Schulich Faculty of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (N.G.); (M.C.)
- Department of Pathology, London Health Sciences Centre, London, ON N6A 5A5, Canada
| | - Muriel Brackstone
- Schulich Faculty of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada; (N.G.); (M.C.)
- Department of Surgery, London Health Sciences Centre, London, ON N6A 5W9, Canada
| |
Collapse
|
8
|
Schuiveling M, Liu H, Eek D, Breimer GE, Suijkerbuijk KPM, Blokx WAM, Veta M. A novel dataset for nuclei and tissue segmentation in melanoma with baseline nuclei segmentation and tissue segmentation benchmarks. Gigascience 2025; 14:giaf011. [PMID: 39970004 PMCID: PMC11837757 DOI: 10.1093/gigascience/giaf011] [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: 10/07/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Melanoma is an aggressive form of skin cancer in which tumor-infiltrating lymphocytes (TILs) are a biomarker for recurrence and treatment response. Manual TIL assessment is prone to interobserver variability, and current deep learning models are not publicly accessible or have low performance. Deep learning models, however, have the potential of consistent spatial evaluation of TILs and other immune cell subsets with the potential of improved prognostic and predictive value. To make the development of these models possible, we created the Panoptic Segmentation of nUclei and tissue in advanced MelanomA (PUMA) dataset and assessed the performance of several state-of-the-art deep learning models. In addition, we show how to improve model performance further by using heuristic postprocessing in which nuclei classes are updated based on their tissue localization. RESULTS The PUMA dataset includes 155 primary and 155 metastatic melanoma hematoxylin and eosin-stained regions of interest with nuclei and tissue annotations from a single melanoma referral institution. The Hover-NeXt model, trained on the PUMA dataset, demonstrated the best performance for lymphocyte detection, approaching human interobserver agreement. In addition, heuristic postprocessing of deep learning models improved the detection of noncommon classes, such as epithelial nuclei. CONCLUSION The PUMA dataset is the first melanoma-specific dataset that can be used to develop melanoma-specific nuclei and tissue segmentation models. These models can, in turn, be used for prognostic and predictive biomarker development. Incorporating tissue and nuclei segmentation is a step toward improved deep learning nuclei segmentation performance. To support the development of these models, this dataset is used in the PUMA challenge.
Collapse
Affiliation(s)
- Mark Schuiveling
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Hong Liu
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Daniel Eek
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| | - Gerben E Breimer
- Department of Pathology, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Willeke A M Blokx
- Department of Pathology, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Mitko Veta
- Medical Image Analysis, Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, the Netherlands
| |
Collapse
|
9
|
Nakabayashi Y, Kiuchi J, Kubota T, Ohashi T, Nishibeppu K, Imamura T, Nanishi K, Shimizu H, Arita T, Yamamoto Y, Konishi H, Morimura R, Komatsu S, Shiozaki A, Ikoma H, Kuriu Y, Fujiwara H, Tsuda H, Otsuji E. A novel semi-quantitative scoring method for CD8+ tumor-infiltrating lymphocytes based on infiltration sites in gastric cancer. Am J Cancer Res 2024; 14:5965-5986. [PMID: 39803654 PMCID: PMC11711524 DOI: 10.62347/jkcu5881] [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: 10/25/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
No established method currently exists for evaluating tumor-infiltrating lymphocytes (TILs) in gastric cancer (GC), and their clinical significance based on infiltration site in GC remains unclear. In this study, we developed a method to evaluate TILs according to their infiltration site as a prognostic marker for GC. We retrospectively analyzed 103 patients with advanced GC who underwent curative resection. TILs located at the invasive margin (TILIM) and the center of tumors (TILCT) were scored semi-quantitatively using immunohistochemical staining of CD8+ T cells. The sum of the TILIM and TILCT scores was defined as the TILs score. Based on this score, patients were classified into low and high TILs groups. Quantitative TILs were also assessed to validate the semi-quantitative scoring method. Furthermore, we confirmed a tumor suppressive effect due to CD8+ T cells co-cultured in GC cell lines in vitro. In the univariate analysis, patients with low TILIM were significantly more likely to be female, younger, and have undifferentiated histological types and deeper tumor invasion compared to those with high TILIM. Similarly, patients with low TILCT had significantly more positive lymph node metastases than those with high TILCT. In the multivariate analysis, deeper tumor invasion and positive lymph node metastasis were identified as independent risk factors for patients with low TILIM and low TILCT, respectively. According to our semi-quantitative TILs scoring method, the low TILs group had significantly poorer prognoses compared to the high TILs group. This group had significantly larger tumor diameters, deeper tumor invasion, and more positive lymph node metastases. Additionally, deeper tumor invasion was an independent risk factor for the low TILs group. Quantitative TILs analysis revealed that the low TILs group had significantly lower TIL levels compared to the high TILs group. In vitro, CD8+ T cells induced apoptosis in GC cells in a concentration-dependent manner. Furthermore, these cells significantly suppressed the proliferative, migratory, and invasive capacities of GC cells. Our simple and versatile semi-quantitative scoring method for CD8+ TILs indicates that CD8+ TILs are sensitive prognostic markers. The low TILs group accurately reflects the low quantitative TIL levels and is associated with poor oncological prognosis.
Collapse
Affiliation(s)
- Yudai Nakabayashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Jun Kiuchi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Takeshi Kubota
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Takuma Ohashi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Keiji Nishibeppu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Taisuke Imamura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Kenji Nanishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hiroki Shimizu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Tomohiro Arita
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yusuke Yamamoto
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Ryo Morimura
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hisashi Ikoma
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Yoshiaki Kuriu
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical CollegeTokorozawa, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of MedicineKawaramachi-Hirokoji, Kamigyo-ku, Kyoto, Japan
| |
Collapse
|
10
|
Capar A, Ekinci DA, Ertano M, Niazi MKK, Balaban EB, Aloglu I, Dogan M, Su Z, Aker FV, Gurcan MN. An interpretable framework for inter-observer agreement measurements in TILs scoring on histopathological breast images: A proof-of-principle study. PLoS One 2024; 19:e0314450. [PMID: 39636880 PMCID: PMC11620390 DOI: 10.1371/journal.pone.0314450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Breast cancer, a widespread and life-threatening disease, necessitates precise diagnostic tools for improved patient outcomes. Tumor-Infiltrating Lymphocytes (TILs), reflective of the immune response against cancer cells, are pivotal in understanding breast cancer behavior. However, inter-observer variability in TILs scoring methods poses challenges to reliable assessments. This study introduces a novel and interpretable proof-of-principle framework comprising two innovative inter-observer agreement measures. The first method, Boundary-Weighted Fleiss' Kappa (BWFK), addresses tissue segmentation predictions, focusing on mitigating disagreements along tissue boundaries. BWFK enhances the accuracy of stromal segmentation, providing a nuanced assessment of inter-observer agreement. The second proposed method, the Distance Based Cell Agreement Algorithm (DBCAA), eliminates the need for ground truth annotations in cell detection predictions. This innovative approach offers versatility across histopathological analyses, overcoming data availability challenges. Both methods were applied to assess inter-observer agreement using a clinical image dataset consisting of 25 images of invasive ductal breast carcinoma tissue, each annotated by four pathologists, serving as a proof-of-principle. Experimental investigations demonstrated that the BWFK method yielded gains of up to 32% compared to the standard Fleiss' Kappa model. Furthermore, a procedure for conducting clinical validations of artificial intelligence (AI) based cell detection methods was elucidated. Thoroughly validated on a clinical dataset, the framework contributes to standardized, reliable, and interpretable inter-observer agreement assessments. This study is the first examination of inter-observer agreements in stromal segmentation and lymphocyte detection for the TILs scoring problem. The study emphasizes the potential impact of these measures in advancing histopathological image analysis, fostering consensus in TILs scoring, and ultimately improving breast cancer diagnostics and treatment planning. The source code and implementation guide for this study are accessible on our GitHub page, and the full clinical dataset is available for academic and research purposes on Kaggle.
Collapse
Affiliation(s)
- Abdulkerim Capar
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
- Informatics Institute, Istanbul Technical University, Istanbul, Turkiye
| | - Dursun Ali Ekinci
- Informatics Institute, Istanbul Technical University, Istanbul, Turkiye
| | - Mucahit Ertano
- Informatics Institute, Istanbul Technical University, Istanbul, Turkiye
| | - M. Khalid Khan Niazi
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Erva Bengu Balaban
- Department of Pathology, Haydarpasa Numune Education and Research Hospital, University of Health Sciences, Istanbul, Turkiye
| | - Ibrahim Aloglu
- Department of Pathology, Haydarpasa Numune Education and Research Hospital, University of Health Sciences, Istanbul, Turkiye
| | - Meryem Dogan
- Department of Pathology, Haydarpasa Numune Education and Research Hospital, University of Health Sciences, Istanbul, Turkiye
| | - Ziyu Su
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Fugen Vardar Aker
- Department of Pathology, Haydarpasa Numune Education and Research Hospital, University of Health Sciences, Istanbul, Turkiye
| | - Metin Nafi Gurcan
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| |
Collapse
|
11
|
Carvalho FM. Targeting low-risk triple-negative breast cancer: a review on de-escalation strategies for a new era. TRANSLATIONAL BREAST CANCER RESEARCH : A JOURNAL FOCUSING ON TRANSLATIONAL RESEARCH IN BREAST CANCER 2024; 6:4. [PMID: 39980807 PMCID: PMC11836748 DOI: 10.21037/tbcr-24-28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/18/2024] [Indexed: 02/22/2025]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer lacking estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Comprising 15-20% of breast cancers, TNBC is typically high-grade, affects younger women, and has a poor prognosis. However, TNBC is heterogeneous, encompassing different molecular subtypes and histological types with distinct molecular drivers, prognoses, and treatment responses. Among these, a subset of low-risk diseases associated with a lower risk of recurrence led to the exploration of de-escalation strategies. This review presents the clinicopathological characteristics of special TNBC with a better prognosis that could be spared from aggressive systemic treatment. We searched the PubMed database for articles that could support treatment de-escalation using the keywords "early-stage", "TNBC", and "low-risk". This article addresses four subgroups of low-risk TNBC: special histological types, tumors with high tumor-infiltrating lymphocytes (TILs), low Ki-67 levels, and early-stage tumors that achieved pathological complete response (pCR). The discussion explores the optimization of treatment options ranging from the omission of any systemic treatment to anthracycline-free possibilities and/or immunotherapies. Identifying these tumors can help personalize treatment, reduce costs and unnecessary toxicity, and contribute to a better quality of life for patients with favorable prognoses. Further studies should explore reliable biomarkers to identify these low-risk diseases precisely.
Collapse
Affiliation(s)
- Filomena Marino Carvalho
- Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
12
|
Uchikov P, Khalid U, Dedaj-Salad GH, Ghale D, Rajadurai H, Kraeva M, Kraev K, Hristov B, Doykov M, Mitova V, Bozhkova M, Markov S, Stanchev P. Artificial Intelligence in Breast Cancer Diagnosis and Treatment: Advances in Imaging, Pathology, and Personalized Care. Life (Basel) 2024; 14:1451. [PMID: 39598249 PMCID: PMC11595975 DOI: 10.3390/life14111451] [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: 10/20/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Breast cancer is the most prevalent cancer worldwide, affecting both low- and middle-income countries, with a growing number of cases. In 2024, about 310,720 women in the U.S. are projected to receive an invasive breast cancer diagnosis, alongside 56,500 cases of ductal carcinoma in situ (DCIS). Breast cancer occurs in every country of the world in women at any age after puberty but with increasing rates in later life. About 65% of women with the BRCA1 and 45% with the BRCA2 gene variants develop breast cancer by age 70. While these genes account for 5% of breast cancers, their prevalence is higher in certain populations. Advances in early detection, personalised medicine, and AI-driven diagnostics are improving outcomes by enabling a more precise analysis, reducing recurrence, and minimising treatment side effects. Our paper aims to explore the vast applications of artificial intelligence within the diagnosis and treatment of breast cancer and how these advancements can contribute to elevating patient care as well as discussing the potential drawbacks of such integrations into modern medicine. We structured our paper as a non-systematic review and utilised Google Scholar and PubMed databases to review literature regarding the incorporation of AI in the diagnosis and treatment of non-palpable breast masses. AI is revolutionising breast cancer management by enhancing imaging, pathology, and personalised treatment. In imaging, AI can improve the detection of cancer in mammography, MRIs, and ultrasounds, rivalling expert radiologists in accuracy. In pathology, AI enhances biomarker detection, improving HER2 and Ki67 assessments. Personalised medicine benefits from AI's predictive power, aiding risk stratification and treatment response. AI also shows promise in triple-negative breast cancer management, offering better prognosis and subtype classification. However, challenges include data variability, ethical concerns, and real-world validation. Despite limitations, AI integration offers significant potential in improving breast cancer diagnosis, prognosis, and treatment outcomes.
Collapse
Affiliation(s)
- Petar Uchikov
- Department of Special Surgery, Faculty of Medicine, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Usman Khalid
- Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria; (U.K.); (G.H.D.-S.); (D.G.); (H.R.)
| | - Granit Harris Dedaj-Salad
- Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria; (U.K.); (G.H.D.-S.); (D.G.); (H.R.)
| | - Dibya Ghale
- Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria; (U.K.); (G.H.D.-S.); (D.G.); (H.R.)
| | - Harney Rajadurai
- Faculty of Medicine, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria; (U.K.); (G.H.D.-S.); (D.G.); (H.R.)
| | - Maria Kraeva
- Department of Otorhinolaryngology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.K.); (S.M.)
| | - Krasimir Kraev
- Department of Propedeutics of Internal Diseases, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Bozhidar Hristov
- Second Department of Internal Diseases, Section “Gastroenterology”, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| | - Mladen Doykov
- Department of Urology and General Medicine, Medical Faculty, Medical University of Plovdiv, 4001 Plovdiv, Bulgaria;
| | - Vanya Mitova
- University Specialized Hospital for Active Oncology Treatment “Prof. Ivan Chernozemsky”, 1756 Sofia, Bulgaria;
| | - Maria Bozhkova
- Medical College, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria;
| | - Stoyan Markov
- Department of Otorhinolaryngology, Medical Faculty, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria; (M.K.); (S.M.)
| | - Pavel Stanchev
- Clinic of Endocrinology and Metabolic Diseases, St George University Hospital, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| |
Collapse
|
13
|
Thomas N, Garaud S, Langouo M, Sofronii D, Boisson A, De Wind A, Duwel V, Craciun L, Larsimont D, Awada A, Willard-Gallo K. Tumor-Infiltrating Lymphocyte Scoring in Neoadjuvant-Treated Breast Cancer. Cancers (Basel) 2024; 16:2895. [PMID: 39199667 PMCID: PMC11352458 DOI: 10.3390/cancers16162895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Neoadjuvant chemotherapy (NAC) is now the standard of care for patients with locally advanced breast cancer (BC). TIL scoring is prognostic and adds predictive value to the residual cancer burden evaluation after NAC. However, NAC induces changes in the tumor, and the reliability of TIL scoring in post-NAC samples has not yet been studied. H&E- and dual CD3/CD20 chromogenic IHC-stained tissues were scored for stromal and intra-tumoral TIL by two experienced pathologists on pre- and post-treatment BC tissues. Digital TIL scoring was performed using the HALO® image analysis software (version 2.2). In patients with residual disease, we show a good inter-pathologist correlation for stromal TIL on H&E-stained tissues (CCC value 0.73). A good correlation for scoring with both staining methods (CCC 0.81) and the digital TIL scoring (CCC 0.77) was also observed. Overall concordance for TIL scoring in patients with a complete response was however poor. This study reveals there is good reliability for TIL scoring in patients with detectable residual tumors after NAC treatment, which is comparable to the scoring of untreated breast cancer patients. Based on the good consistency observed with digital TIL scoring, the development of a validated algorithm in the future might be advantageous.
Collapse
Affiliation(s)
- Noémie Thomas
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Mireille Langouo
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Doïna Sofronii
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Anaïs Boisson
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| | - Alexandre De Wind
- Anantomical Pathology Department, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Valérie Duwel
- Anatomical Pathology Department, AZ Klina, 2930 Brasschaat, Belgium;
| | - Ligia Craciun
- Anantomical Pathology Department, Institut Jules Bordet, 1070 Brussels, Belgium
- Tumor Bank, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Dennis Larsimont
- Anantomical Pathology Department, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Ahmad Awada
- Medical Oncology, Institut Jules Bordet, 1070 Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, 1070 Brussels, Belgium (A.B.)
| |
Collapse
|
14
|
Hamada K, Murakami R, Ueda A, Kashima Y, Miyagawa C, Taki M, Yamanoi K, Yamaguchi K, Hamanishi J, Minamiguchi S, Matsumura N, Mandai M. A Deep Learning-Based Assessment Pipeline for Intraepithelial and Stromal Tumor-Infiltrating Lymphocytes in High-Grade Serous Ovarian Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1272-1284. [PMID: 38537936 DOI: 10.1016/j.ajpath.2024.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/22/2024] [Accepted: 02/21/2024] [Indexed: 04/07/2024]
Abstract
Tumor-infiltrating lymphocytes (TILs) are associated with improved survival in patients with epithelial ovarian cancer. However, TIL evaluation has not been used in routine clinical practice because of reproducibility issues. The current study developed two convolutional neural network models to detect TILs and to determine their spatial location in whole slide images, and established a spatial assessment pipeline to objectively quantify intraepithelial and stromal TILs in patients with high-grade serous ovarian carcinoma. The predictions of the established models showed a significant positive correlation with the number of CD8+ T cells and immune gene expressions. Patients with a higher density of intraepithelial TILs had a significantly prolonged overall survival and progression-free survival in multiple cohorts. On the basis of the density of intraepithelial and stromal TILs, patients were classified into three immunophenotypes: immune inflamed, excluded, and desert. The immune-desert subgroup showed the worst prognosis. Gene expression analysis showed that the immune-desert subgroup had lower immune cytolytic activity and T-cell-inflamed gene-expression profile scores, whereas the immune-excluded subgroup had higher expression of interferon-γ and programmed death 1 receptor signaling pathway. The established evaluation method provided detailed and comprehensive quantification of intraepithelial and stromal TILs throughout hematoxylin and eosin-stained slides. It has potential for clinical application for personalized treatment of patients with ovarian cancer.
Collapse
Affiliation(s)
- Kohei Hamada
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Akihiko Ueda
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoko Kashima
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Chiho Miyagawa
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
15
|
Wu R, Horimoto Y, Oshi M, Benesch MGK, Khoury T, Takabe K, Ishikawa T. Emerging measurements for tumor-infiltrating lymphocytes in breast cancer. Jpn J Clin Oncol 2024; 54:620-629. [PMID: 38521965 PMCID: PMC11144297 DOI: 10.1093/jjco/hyae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/01/2024] [Indexed: 03/25/2024] Open
Abstract
Tumor-infiltrating lymphocytes are a general term for lymphocytes or immune cells infiltrating the tumor microenvironment. Numerous studies have demonstrated tumor-infiltrating lymphocytes to be robust prognostic and predictive biomarkers in breast cancer. Recently, immune checkpoint inhibitors, which directly target tumor-infiltrating lymphocytes, have become part of standard of care treatment for triple-negative breast cancer. Surprisingly, tumor-infiltrating lymphocytes quantified by conventional methods do not predict response to immune checkpoint inhibitors, which highlights the heterogeneity of tumor-infiltrating lymphocytes and the complexity of the immune network in the tumor microenvironment. Tumor-infiltrating lymphocytes are composed of diverse immune cell populations, including cytotoxic CD8-positive T lymphocytes, B cells and myeloid cells. Traditionally, tumor-infiltrating lymphocytes in tumor stroma have been evaluated by histology. However, the standardization of this approach is limited, necessitating the use of various novel technologies to elucidate the heterogeneity in the tumor microenvironment. This review outlines the evaluation methods for tumor-infiltrating lymphocytes from conventional pathological approaches that evaluate intratumoral and stromal tumor-infiltrating lymphocytes such as immunohistochemistry, to the more recent advancements in computer tissue imaging using artificial intelligence, flow cytometry sorting and multi-omics analyses using high-throughput assays to estimate tumor-infiltrating lymphocytes from bulk tumor using immune signatures or deconvolution tools. We also discuss higher resolution technologies that enable the analysis of tumor-infiltrating lymphocytes heterogeneity such as single-cell analysis and spatial transcriptomics. As we approach the era of personalized medicine, it is important for clinicians to understand these technologies.
Collapse
Affiliation(s)
- Rongrong Wu
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Yoshiya Horimoto
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- Department of Breast Oncology, Juntendo University Hospital, Tokyo, Japan
| | - Masanori Oshi
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Matthew G K Benesch
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Thaer Khoury
- Department of Pathology & Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kazuaki Takabe
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
- Department of Surgical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Surgery, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, The State University of New York, Buffalo, NY, USA
- Department of Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
- Department of Breast Surgery, Fukushima Medical University, Fukushima, Japan
| | - Takashi Ishikawa
- Department of Breast Surgery and Oncology, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
16
|
Ivanova M, Pescia C, Trapani D, Venetis K, Frascarelli C, Mane E, Cursano G, Sajjadi E, Scatena C, Cerbelli B, d’Amati G, Porta FM, Guerini-Rocco E, Criscitiello C, Curigliano G, Fusco N. Early Breast Cancer Risk Assessment: Integrating Histopathology with Artificial Intelligence. Cancers (Basel) 2024; 16:1981. [PMID: 38893102 PMCID: PMC11171409 DOI: 10.3390/cancers16111981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Effective risk assessment in early breast cancer is essential for informed clinical decision-making, yet consensus on defining risk categories remains challenging. This paper explores evolving approaches in risk stratification, encompassing histopathological, immunohistochemical, and molecular biomarkers alongside cutting-edge artificial intelligence (AI) techniques. Leveraging machine learning, deep learning, and convolutional neural networks, AI is reshaping predictive algorithms for recurrence risk, thereby revolutionizing diagnostic accuracy and treatment planning. Beyond detection, AI applications extend to histological subtyping, grading, lymph node assessment, and molecular feature identification, fostering personalized therapy decisions. With rising cancer rates, it is crucial to implement AI to accelerate breakthroughs in clinical practice, benefiting both patients and healthcare providers. However, it is important to recognize that while AI offers powerful automation and analysis tools, it lacks the nuanced understanding, clinical context, and ethical considerations inherent to human pathologists in patient care. Hence, the successful integration of AI into clinical practice demands collaborative efforts between medical experts and computational pathologists to optimize patient outcomes.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.T.); (C.C.); (G.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Chiara Frascarelli
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Eltjona Mane
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Giulia Cursano
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Cristian Scatena
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Bruna Cerbelli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy;
| | - Giulia d’Amati
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00185 Rome, Italy;
| | - Francesca Maria Porta
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.T.); (C.C.); (G.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, 20141 Milan, Italy; (D.T.); (C.C.); (G.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (M.I.); (C.P.); (K.V.); (C.F.); (E.M.); (G.C.); (E.S.); (F.M.P.); (E.G.-R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
17
|
McCaffrey C, Jahangir C, Murphy C, Burke C, Gallagher WM, Rahman A. Artificial intelligence in digital histopathology for predicting patient prognosis and treatment efficacy in breast cancer. Expert Rev Mol Diagn 2024; 24:363-377. [PMID: 38655907 DOI: 10.1080/14737159.2024.2346545] [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: 12/07/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
INTRODUCTION Histological images contain phenotypic information predictive of patient outcomes. Due to the heavy workload of pathologists, the time-consuming nature of quantitatively assessing histological features, and human eye limitations to recognize spatial patterns, manually extracting prognostic information in routine pathological workflows remains challenging. Digital pathology has facilitated the mining and quantification of these features utilizing whole-slide image (WSI) scanners and artificial intelligence (AI) algorithms. AI algorithms to identify image-based biomarkers from the tumor microenvironment (TME) have the potential to revolutionize the field of oncology, reducing delays between diagnosis and prognosis determination, allowing for rapid stratification of patients and prescription of optimal treatment regimes, thereby improving patient outcomes. AREAS COVERED In this review, the authors discuss how AI algorithms and digital pathology can predict breast cancer patient prognosis and treatment outcomes using image-based biomarkers, along with the challenges of adopting this technology in clinical settings. EXPERT OPINION The integration of AI and digital pathology presents significant potential for analyzing the TME and its diagnostic, prognostic, and predictive value in breast cancer patients. Widespread clinical adoption of AI faces ethical, regulatory, and technical challenges, although prospective trials may offer reassurance and promote uptake, ultimately improving patient outcomes by reducing diagnosis-to-prognosis delivery delays.
Collapse
Affiliation(s)
- Christine McCaffrey
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Chowdhury Jahangir
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Clodagh Murphy
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Caoimbhe Burke
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Arman Rahman
- UCD School of Medicine, UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
18
|
Lopez-Gonzalez L, Sanchez Cendra A, Sanchez Cendra C, Roberts Cervantes ED, Espinosa JC, Pekarek T, Fraile-Martinez O, García-Montero C, Rodriguez-Slocker AM, Jiménez-Álvarez L, Guijarro LG, Aguado-Henche S, Monserrat J, Alvarez-Mon M, Pekarek L, Ortega MA, Diaz-Pedrero R. Exploring Biomarkers in Breast Cancer: Hallmarks of Diagnosis, Treatment, and Follow-Up in Clinical Practice. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:168. [PMID: 38256428 PMCID: PMC10819101 DOI: 10.3390/medicina60010168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/02/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024]
Abstract
Breast cancer is a prevalent malignancy in the present day, particularly affecting women as one of the most common forms of cancer. A significant portion of patients initially present with localized disease, for which curative treatments are pursued. Conversely, another substantial segment is diagnosed with metastatic disease, which has a worse prognosis. Recent years have witnessed a profound transformation in the prognosis for this latter group, primarily due to the discovery of various biomarkers and the emergence of targeted therapies. These biomarkers, encompassing serological, histological, and genetic indicators, have demonstrated their value across multiple aspects of breast cancer management. They play crucial roles in initial diagnosis, aiding in the detection of relapses during follow-up, guiding the application of targeted treatments, and offering valuable insights for prognostic stratification, especially for highly aggressive tumor types. Molecular markers have now become the keystone of metastatic breast cancer diagnosis, given the diverse array of chemotherapy options and treatment modalities available. These markers signify a transformative shift in the arsenal of therapeutic options against breast cancer. Their diagnostic precision enables the categorization of tumors with elevated risks of recurrence, increased aggressiveness, and heightened mortality. Furthermore, the existence of therapies tailored to target specific molecular anomalies triggers a cascade of changes in tumor behavior. Therefore, the primary objective of this article is to offer a comprehensive review of the clinical, diagnostic, prognostic, and therapeutic utility of the principal biomarkers currently in use, as well as of their clinical impact on metastatic breast cancer. In doing so, our goal is to contribute to a more profound comprehension of this complex disease and, ultimately, to enhance patient outcomes through more precise and effective treatment strategies.
Collapse
Affiliation(s)
- Laura Lopez-Gonzalez
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
| | - Alicia Sanchez Cendra
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | - Cristina Sanchez Cendra
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | | | - Javier Cassinello Espinosa
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
| | - Tatiana Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Oscar Fraile-Martinez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Cielo García-Montero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Ana María Rodriguez-Slocker
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
| | - Laura Jiménez-Álvarez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| | - Luis G. Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
| | - Soledad Aguado-Henche
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
| | - Jorge Monserrat
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Melchor Alvarez-Mon
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - Leonel Pekarek
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Oncology Service, Guadalajara University Hospital, 19002 Guadalajara, Spain; (A.S.C.); (C.S.C.); (E.D.R.C.); (J.C.E.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
| | - Miguel A. Ortega
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (T.P.); (L.J.-Á.)
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, 28806 Alcala de Henares, Spain
| | - Raul Diaz-Pedrero
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (L.L.-G.); (A.M.R.-S.); (S.A.-H.); (R.D.-P.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (O.F.-M.); (C.G.-M.); (L.G.G.); (M.A.-M.); (L.P.); (M.A.O.)
- Department of General and Digestive Surgery, General and Digestive Surgery, Príncipe de Asturias Universitary Hospital, 28805 Alcala de Henares, Spain
| |
Collapse
|
19
|
Jones JL, Poulsom R, Coates PJ. Recent Advances in Pathology: the 2023 Annual Review Issue of The Journal of Pathology. J Pathol 2023; 260:495-497. [PMID: 37580852 DOI: 10.1002/path.6192] [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: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/16/2023]
Abstract
The 2023 Annual Review Issue of The Journal of Pathology, Recent Advances in Pathology, contains 12 invited reviews on topics of current interest in pathology. This year, our subjects include immuno-oncology and computational pathology approaches for diagnostic and research applications in human disease. Reviews on the tissue microenvironment include the effects of apoptotic cell-derived exosomes, how understanding the tumour microenvironment predicts prognosis, and the growing appreciation of the diverse functions of fibroblast subtypes in health and disease. We also include up-to-date reviews of modern aspects of the molecular basis of malignancies, and our final review covers new knowledge of vascular and lymphatic regeneration in cardiac disease. All of the reviews contained in this issue are written by expert groups of authors selected to discuss the recent progress in their particular fields and all articles are freely available online (https://pathsocjournals.onlinelibrary.wiley.com/journal/10969896). © 2023 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- J Louise Jones
- Centre for Tumour Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Richard Poulsom
- The Pathological Society of Great Britain and Ireland, London, UK
| | - Philip J Coates
- Research Center for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|