1
|
Brogård MB, Steiniche T, Lade-Keller J, Wandler A, Christensen KB, Georgsen JB, Nielsen PS. Digital quantification of Ki67 and PRAME in challenging melanocytic lesions - A novel diagnostic tool. Pathol Res Pract 2025; 270:155953. [PMID: 40209567 DOI: 10.1016/j.prp.2025.155953] [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: 12/18/2024] [Revised: 03/06/2025] [Accepted: 03/28/2025] [Indexed: 04/12/2025]
Abstract
The interpretation of immunohistochemical markers in melanocytic lesions possesses difficulties due to expression in non-melanocytic cells and the time-consuming, non-reproducible nature of manual assessment. A digital tool that accurately quantifies Ki67 and PRAME may valuably aid pathologists in the diagnostic classification of melanocytic lesions. The aim of this study was to assess the diagnostic performance of digitally quantified Ki67 and PRAME in challenging melanocytic lesions utilizing double nuclear staining methods for accurate identification of melanocytic cells. We explored the difference in Ki67 and PRAME expression by WHO-lesion-groups and Melanocytic Pathology Assessment Tool and Hierarchy for Diagnosis version 2.0 (MPATH-Dx V2.0). Tissue slides from a cohort of 156 melanocytic lesions were stained with the Ki67/SOX10 double nuclear stain and the PRAME/SOX10 virtual double nuclear stain. Melanocytic cell specific Ki67/SOX10- and PRAME/SOX10-indexes were quantified by AI-driven digital image analysis (DIA) and compared to non-specific Ki67- and PRAME-indexes. The results showed that ROC AUC of the Ki67/SOX10-index was increased compared to the non-specific Ki67-index (p < 0.001), as opposed to the AUC of the PRAME/SOX10-index compared to non-specific PRAME-index (p = 0.090). The medians of digitally quantified Ki67- and PRAME-indexes differed significantly for the overall WHO-groups and MPATH-Dx V2.0 classes (p < 0.001). In conclusion, we found that double nuclear staining improved the diagnostic performance of Ki67, but not PRAME. The combination of digitally quantified Ki67- and PRAME-indexes may potentially serve as a tool for diagnostic classification of challenging melanocytic lesions. The proposed diagnostic tool presents the results visually, graphically, and quantitatively to optimally aid the pathologist.
Collapse
Affiliation(s)
- Mette Bak Brogård
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200 Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark.
| | - Torben Steiniche
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200 Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Johanne Lade-Keller
- Department of Pathology, Aalborg University Hospital, Ladegårdsgade 3, 9000 Aalborg, Denmark
| | - Anne Wandler
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200 Aarhus, Denmark
| | - Kristina Bang Christensen
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200 Aarhus, Denmark
| | - Jeanette Bæhr Georgsen
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200 Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| | - Patricia Switten Nielsen
- Department of Pathology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, 8200 Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 99, 8200 Aarhus, Denmark
| |
Collapse
|
2
|
Chow AK, Sharmin N. Zoom in on learning with a virtual microscope: A convergent parallel mixed-method study. PLoS One 2025; 20:e0323412. [PMID: 40334219 PMCID: PMC12058188 DOI: 10.1371/journal.pone.0323412] [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: 12/05/2024] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Knowledge of histology is essential for many disciplines in professional health education. Virtual Microscopes (VMs) are increasingly becoming popular as a cost-effective teaching tool for histology. However, a lack of hands-on experience with traditional light microscopes and glass slides concerns many educators. Although studies have reported improvement or no difference in students' knowledge and/or performance using either virtual or optical microscopes, reports on the impact of VMs on students' understanding of magnification and orientation are scarce. We conducted a convergent, parallel mixed-method study to assess dental students' understanding of magnification and orientation after using a virtual microscope to study oral histology. Six hours of mandatory lab sessions and critical thinking assignment questions were designed for the 1st year students in the Doctor of Dental Surgery (DDS) program. Quantitative data were collected from students' performance in orientation and magnification-related questions in the summative assessments. Students' written responses to reflective questions were the qualitative data, analyzed using manifest content analysis. All 32 students accurately answered 3 out of 5 questions, requiring them to apply the knowledge of magnification and orientation. 30 and 29 students correctly answered the remaining questions, respectively. 97% of the class agreed to improve their understanding of magnification and orientation after using the VM. All (100%) students (n = 32) completed the reflective assignment, generating 64 meaning units. 17 codes were generated and compiled into seven subcategories, which were further condensed into two categories: refinement of mental models and enhanced learning. Although our study is limited by a small sample size, it sheds light on the strategies adopted by dental students to improve their senses of magnification and orientation while using a VM.
Collapse
Affiliation(s)
- Ava K. Chow
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta,Edmonton, Canada
| | - Nazlee Sharmin
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta,Edmonton, Canada
| |
Collapse
|
3
|
Phillips R, Zakkaroff C, Dittmer K, Robilliard N, Baer K, Butler A. A Proof-of-Concept Solution for Co-locating 2D Histology Images in 3D for Histology-to-CT and MR Image Registration: Closing the Loop for Bone Sarcoma Treatment Planning. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025:10.1007/s10278-025-01426-5. [PMID: 40011346 DOI: 10.1007/s10278-025-01426-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
This work presents a proof-of-concept solution designed to facilitate more accurate radiographic feature characterisation in pre-surgical CT/MR volumes. The solution involves 3D co-location of 2D digital histology slides within ex-vivo, tumour tissue CT volumes. Initially, laboratory dissection measurements seed the placement of histology slices in corresponding CT volumes, followed by in-plane point-based registration of bone in histology images to the bone in CT. Validation using six bisected canine humerus ex-vivo CT datasets indicated a plane misalignment of 0.19 ± 1.8 mm. User input sensitivity was assessed at 0.08 ± 0.2 mm for plane translation and 0-1.6° deviation. These results show a similar magnitude of error to related prostate histology co-location work. Although demonstrated with a femoral canine sarcoma tumour, this solution can be generalised to various orthopaedic geometries and sites. It supports high-fidelity histology image co-location to improve understanding of tissue characterisation accuracy in clinical radiology. This solution requires only minimal adjustment to routine workflows. By integrating histology insights earlier in the presentation-diagnosis-planning-surgery-recovery loop, this solution guides data co-location to support the continued evaluation of safe pre-surgical margins.
Collapse
Affiliation(s)
- Robert Phillips
- The University of Otago - Canterbury, Christchurch, New Zealand.
| | | | | | | | - Kenzie Baer
- The University of Otago - Canterbury, Christchurch, New Zealand
| | | |
Collapse
|
4
|
Sisó S, Kavirayani AM, Couto S, Stierstorfer B, Mohanan S, Morel C, Marella M, Bangari DS, Clark E, Schwartz A, Carreira V. Trends and Challenges of the Modern Pathology Laboratory for Biopharmaceutical Research Excellence. Toxicol Pathol 2025; 53:5-20. [PMID: 39673215 DOI: 10.1177/01926233241303898] [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] [Indexed: 12/16/2024]
Abstract
Pathology, a fundamental discipline that bridges basic scientific discovery to the clinic, is integral to successful drug development. Intrinsically multimodal and multidimensional, anatomic pathology continues to be empowered by advancements in molecular and digital technologies enabling the spatial tissue detection of biomolecules such as genes, transcripts, and proteins. Over the past two decades, breakthroughs in spatial molecular biology technologies and advancements in automation and digitization of laboratory processes have enabled the implementation of higher throughput assays and the generation of extensive molecular data sets from tissue sections in biopharmaceutical research and development research units. It is our goal to provide readers with some rationale, advice, and ideas to help establish a modern molecular pathology laboratory to meet the emerging needs of biopharmaceutical research. This manuscript provides (1) a high-level overview of the current state and future vision for excellence in research pathology practice and (2) shared perspectives on how to optimally leverage the expertise of discovery, toxicologic, and translational pathologists to provide effective spatial, molecular, and digital pathology data to support modern drug discovery. It captures insights from the experiences, challenges, and solutions from pathology laboratories of various biopharmaceutical organizations, including their approaches to troubleshooting and adopting new technologies.
Collapse
Affiliation(s)
- Sílvia Sisó
- AbbVie Bioresearch Center, Worcester, Massachusetts, USA
| | | | | | | | | | | | - Mathiew Marella
- Janssen Research & Development, LLC, La Jolla, California, USA
| | | | - Elizabeth Clark
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | | | | |
Collapse
|
5
|
Mandal S, Baker AM, Graham TA, Bräutigam K. The tumour histopathology "glossary" for AI developers. PLoS Comput Biol 2025; 21:e1012708. [PMID: 39847582 PMCID: PMC11756763 DOI: 10.1371/journal.pcbi.1012708] [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] [Indexed: 01/25/2025] Open
Abstract
The applications of artificial intelligence (AI) and deep learning (DL) are leading to significant advances in cancer research, particularly in analysing histopathology images for prognostic and treatment-predictive insights. However, effective translation of these computational methods requires computational researchers to have at least a basic understanding of histopathology. In this work, we aim to bridge that gap by introducing essential histopathology concepts to support AI developers in their research. We cover the defining features of key cell types, including epithelial, stromal, and immune cells. The concepts of malignancy, precursor lesions, and the tumour microenvironment (TME) are discussed and illustrated. To enhance understanding, we also introduce foundational histopathology techniques, such as conventional staining with hematoxylin and eosin (HE), antibody staining by immunohistochemistry, and including the new multiplexed antibody staining methods. By providing this essential knowledge to the computational community, we aim to accelerate the development of AI algorithms for cancer research.
Collapse
Affiliation(s)
- Soham Mandal
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
- Data Science Team, Institute of Cancer Research, London, United Kingdom
| | - Ann-Marie Baker
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Trevor A. Graham
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| | - Konstantin Bräutigam
- Centre for Evolution and Cancer, Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
6
|
Vázquez-Lema D, Mosqueira-Rey E, Hernández-Pereira E, Fernandez-Lozano C, Seara-Romera F, Pombo-Otero J. Segmentation, classification and interpretation of breast cancer medical images using human-in-the-loop machine learning. Neural Comput Appl 2024. [DOI: 10.1007/s00521-024-10799-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/12/2024] [Indexed: 01/04/2025]
|
7
|
Rajendran R, Beck RC, Waskasi MM, Kelly BD, Bauer DR. Digital analysis of the prostate tumor microenvironment with high-order chromogenic multiplexing. J Pathol Inform 2024; 15:100352. [PMID: 38186745 PMCID: PMC10770522 DOI: 10.1016/j.jpi.2023.100352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/30/2023] [Accepted: 11/16/2023] [Indexed: 01/09/2024] Open
Abstract
As our understanding of the tumor microenvironment grows, the pathology field is increasingly utilizing multianalyte diagnostic assays to understand important characteristics of tumor growth. In clinical settings, brightfield chromogenic assays represent the gold-standard and have developed significant trust as the first-line diagnostic method. However, conventional brightfield tests have been limited to low-order assays that are visually interrogated. We have developed a hybrid method of brightfield chromogenic multiplexing that overcomes these limitations and enables high-order multiplex assays. However, how compatible high-order brightfield multiplexed images are with advanced analytical algorithms has not been extensively evaluated. In the present study, we address this gap by developing a novel 6-marker prostate cancer assay that targets diverse aspects of the tumor microenvironment such as prostate-specific biomarkers (PSMA and p504s), immune biomarkers (CD8 and PD-L1), a prognostic biomarker (Ki-67), as well as an adjunctive diagnostic biomarker (basal cell cocktail) and apply the assay to 143 differentially graded adenocarcinoma prostate tissues. The tissues were then imaged on our spectroscopic multiplexing imaging platform and mined for proteomic and spatial features that were correlated with cancer presence and disease grade. Extracted features were used to train a UMAP model that differentiated healthy from cancerous tissue with an accuracy of 89% and identified clusters of cells based on cancer grade. For spatial analysis, cell-to-cell distances were calculated for all biomarkers and differences between healthy and adenocarcinoma tissues were studied. We report that p504s positive cells were at least 2× closer to cells expressing PD-L1, CD8, Ki-67, and basal cell in adenocarcinoma tissues relative to the healthy control tissues. These findings offer a powerful insight to understand the fingerprint of the prostate tumor microenvironment and indicate that high-order chromogenic multiplexing is compatible with digital analysis. Thus, the presented chromogenic multiplexing system combines the clinical applicability of brightfield assays with the emerging diagnostic power of high-order multiplexing in a digital pathology friendly format that is well-suited for translational studies to better understand mechanisms of tumor development and growth.
Collapse
Affiliation(s)
- Rahul Rajendran
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Rachel C. Beck
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Morteza M. Waskasi
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Brian D. Kelly
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| | - Daniel R. Bauer
- Roche Diagnostics Solutions, (Ventana Medical Systems, Inc.), Tucson, AZ, USA
| |
Collapse
|
8
|
Pelosi G, Travis WD. Head-to-head: Should Ki67 proliferation index be included in the formal classification of pulmonary neuroendocrine neoplasms? Histopathology 2024; 85:535-548. [PMID: 38728050 DOI: 10.1111/his.15206] [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: 02/26/2024] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 08/31/2024]
Abstract
The reporting of lung neuroendocrine neoplasms (NENs) according to the 2021 World Health Organisation (WHO) is based on mitotic count per 2 mm2, necrosis assessment and a constellation of cytological and immunohistochemical details. Accordingly, typical carcinoid and atypical carcinoid are low- to intermediate-grade neuroendocrine tumours (NETs), while large-cell neuroendocrine carcinoma (NEC) and small-cell lung carcinoma are high-grade NECs. In small-sized diagnostic material (cytology and biopsy), the noncommittal term of carcinoid tumour/NET not otherwise specified (NOS) and metastatic carcinoid NOS have been introduced with regard to primary and metastatic diagnostic settings, respectively. Ki-67 antigen, a well-known marker of cell proliferation, has been included in the WHO classification as a non-essential but desirable criterion, especially to distinguish NETs from high-grade NECs and to delineate the provisional category of carcinoid tumours/NETs with elevated mitotic counts (> 10 mitoses per mm2) and/or Ki-67 proliferation index (≥ 30%). However, a wider use of this marker in the spectrum of lung NENs continues to be highly reported and debated, thus witnessing a never-subsided attention. Therefore, the arguments for and against incorporating Ki-67 in the classification and clinical practice of these neoplasms are discussed herein in detail.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Inter-Hospital Pathology Division, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, Milan, Italy
| | - William D Travis
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York City, NY, USA
| |
Collapse
|
9
|
Liu A, Zhao Y, Shen H, Ding Z, Deng HW. ResSAT: Enhancing Spatial Transcriptomics Prediction from H&E- Stained Histology Images with Interactive Spot Transformer. RESEARCH SQUARE 2024:rs.3.rs-4707959. [PMID: 39149477 PMCID: PMC11326376 DOI: 10.21203/rs.3.rs-4707959/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Spatial transcriptomics (ST) revolutionizes RNA quantification with high spatial resolution. Hematoxylin and eosin (H&E) images, the gold standard in medical diagnosis, offer insights into tissue structure, correlating with gene expression patterns. Current methods for predicting spatial gene expression from H&E images often overlook spatial relationships. We introduce ResSAT (Residual networks - Self-Attention Transformer), a framework generating spatially resolved gene expression profiles from H&E images by capturing tissue structures and using a self-attention transformer to enhance prediction.Benchmarking on 10× Visium datasets, ResSAT significantly outperformed existing methods, promising reduced ST profiling costs and rapid acquisition of numerous profiles.
Collapse
|
10
|
Ahmed SAA, Gad SEM, Eida OM, Makhlouf LM. Anti-fibrotic Effect of Oral Versus Intraperitoneal Administration of Gold Nanoparticles in Hepatic Schistosoma mansoni-Infected Mice. Acta Parasitol 2024; 69:190-202. [PMID: 37964174 PMCID: PMC11001733 DOI: 10.1007/s11686-023-00730-w] [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: 06/05/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Schistosomiasis significantly impacts public health, as it causes severe morbidity. Infections caused by Schistosoma mansoni (S. mansoni) can be treated with gold nanoparticles (AuNPs). This study aims to determine the most effective route of AuNPs administration and the magnitude of its anti-fibrotic effect. METHODS In the five groups' in vivo assay design, AuNPs were administered intraperitoneally (1 mg/kg) and orally (1 mg/100 g) to S. mansoni-infected mice. Biochemical parameters (serum levels of albumin and liver enzymes alanine aminotransferase (ALT), and aspartate aminotransferase (AST) were measured. The histological changes of the liver in distinct groups were evaluated using Hematoxylin and Eosin, Masson's trichrome, and immunohistochemical stains. RESULTS Infection with S. mansoni was associated with substantial changes in the histological architecture of liver tissue and abnormal levels of hepatic function tests (albumin, AST, and ALT). Schistosoma infected hepatocytes exhibited an abnormal microscopic morphology, granuloma formation and aggressive fibrosis. AuNPs restored the liver histological architecture with a highly significant anti-fibrotic effect and significantly corrected hepatic function test levels. Intraperitoneal administration of AuNPs resulted in the most significant anti-fibrotic effect against hepatic S. mansoni infection as observed in all histological sections with Masson's trichrome being the best stain to represent this fact. CONCLUSION For treating S. mansoni-induced chronic liver fibrosis, intraperitoneal administration of AuNPs is a successful and effective route of administration that can be recommended.
Collapse
Affiliation(s)
| | - Samer Eid Mohamed Gad
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Omima Mohamed Eida
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| | - Laila Mohamed Makhlouf
- Department of Parasitology, Faculty of Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
11
|
Wagner SJ, Matek C, Shetab Boushehri S, Boxberg M, Lamm L, Sadafi A, Winter DJE, Marr C, Peng T. Built to Last? Reproducibility and Reusability of Deep Learning Algorithms in Computational Pathology. Mod Pathol 2024; 37:100350. [PMID: 37827448 DOI: 10.1016/j.modpat.2023.100350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
Recent progress in computational pathology has been driven by deep learning. While code and data availability are essential to reproduce findings from preceding publications, ensuring a deep learning model's reusability is more challenging. For that, the codebase should be well-documented and easy to integrate into existing workflows and models should be robust toward noise and generalizable toward data from different sources. Strikingly, only a few computational pathology algorithms have been reused by other researchers so far, let alone employed in a clinical setting. To assess the current state of reproducibility and reusability of computational pathology algorithms, we evaluated peer-reviewed articles available in PubMed, published between January 2019 and March 2021, in 5 use cases: stain normalization; tissue type segmentation; evaluation of cell-level features; genetic alteration prediction; and inference of grading, staging, and prognostic information. We compiled criteria for data and code availability and statistical result analysis and assessed them in 160 publications. We found that only one-quarter (41 of 160 publications) made code publicly available. Among these 41 studies, three-quarters (30 of 41) analyzed their results statistically, half of them (20 of 41) released their trained model weights, and approximately a third (16 of 41) used an independent cohort for evaluation. Our review is intended for both pathologists interested in deep learning and researchers applying algorithms to computational pathology challenges. We provide a detailed overview of publications with published code in the field, list reusable data handling tools, and provide criteria for reproducibility and reusability.
Collapse
Affiliation(s)
- Sophia J Wagner
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Christian Matek
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Sayedali Shetab Boushehri
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technical University Munich, Munich, Germany; Institute of Pathology Munich-North, Munich, Germany
| | - Lorenz Lamm
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Helmholtz Pioneer Campus, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ario Sadafi
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Dominik J E Winter
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Life Sciences, Technical University of Munich, Weihenstephan, Germany
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| | - Tingying Peng
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
12
|
Li KYC, Dejea H, De Winne K, Bonnin A, D'Onofrio V, Cox JA, Garcia-Canadilla P, Lammens M, Cook AC, Bijnens B, Dendooven A. Feasibility and safety of synchrotron-based X-ray phase contrast imaging as a technique complementary to histopathology analysis. Histochem Cell Biol 2023; 160:377-389. [PMID: 37523091 DOI: 10.1007/s00418-023-02220-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2023] [Indexed: 08/01/2023]
Abstract
X-ray phase contrast imaging (X-PCI) is a powerful technique for high-resolution, three-dimensional imaging of soft tissue samples in a non-destructive manner. In this technical report, we assess the quality of standard histopathological techniques performed on formalin-fixed, paraffin-embedded (FFPE) human tissue samples that have been irradiated with different doses of X-rays in the context of an X-PCI experiment. The data from this study demonstrate that routine histochemical and immunohistochemical staining quality as well as DNA and RNA analyses are not affected by previous X-PCI on human FFPE samples. From these data we conclude it is feasible and acceptable to perform X-PCI on FFPE human biopsies.
Collapse
Affiliation(s)
- Kan Yan Chloe Li
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Hector Dejea
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- ETH Zurich, Zurich, Switzerland
- Department of Biomedical Engineering, Lund University, Lund, Sweden
- MAX IV Laboratory, Lund, Sweden
| | - Koen De Winne
- Department of Pathology, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Anne Bonnin
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
| | | | - Janneke A Cox
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Infectious Diseases and Immunity, Jessa Hospital, Hasselt, Belgium
| | - Patricia Garcia-Canadilla
- Interdisciplinary Cardiovascular Research Group, Sant Joan de Déu Research Institute (IRSJD), Barcelona, Spain
- BCNatal Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Sant Joan de Déu and Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Martin Lammens
- Department of Pathology, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Andrew C Cook
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Bart Bijnens
- ICREA, Barcelona, Spain
- IDIBAPS, Barcelona, Spain
| | - Amélie Dendooven
- Department of Pathology, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium.
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
13
|
Hortsch M. Histology as a paradigm for a science-based learning experience: Visits by histology education spirits of past, present, and future. ANATOMICAL SCIENCES EDUCATION 2023; 16:372-383. [PMID: 36453080 DOI: 10.1002/ase.2235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/31/2022] [Accepted: 11/17/2022] [Indexed: 05/11/2023]
Abstract
The term "histology" was coined a little over 200 years ago and the subject has always relied on microscopy as its defining technology. Microscopy was and still is an essential approach for the description of cellular components and their arrangements in living organisms. For more than a century and a half, histology or microanatomy has also been part of the basic science education for biomedical students. Traditionally, it has been taught in two major components, a didactic transfer of information, either in a lecture or self-learning format, and in active-learning laboratory sessions. These two modes of histology instruction conform with the dual-processing theory of learning, one being more automatic and depending mainly on rote memorization, whereas the other is analytical, requiring more advanced reasoning skills. However, these two components of histology education are not separate and independent, but rather complementary and part of a multi-step learning process that encourages a scientific analysis of visual information and involves higher-level learning skills. Conventional, as well as modern electronic instruction methods (e-learning) have been used in complementary ways to support the integrated succession of individual learning steps as outlined in this manuscript. However, as recent curricular reforms have curtailed instructional time, this traditional format of teaching histology is no longer sustainable and a reflective reassessment of the role of histology in modern biomedical education is a timely necessity.
Collapse
Affiliation(s)
- Michael Hortsch
- Department of Cell and Developmental Biology, University of Michigan Medical School, Michigan, Ann Arbor, USA
- Department of Learning Health Sciences, University of Michigan Medical School, Michigan, Ann Arbor, USA
| |
Collapse
|
14
|
Hortsch M, Koney NKK, Oommen AM, Yohannan DG, Li Y, de Melo Leite ACR, Girão-Carmona VCC. Virtual Microscopy Goes Global: The Images Are Virtual and the Problems Are Real. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1421:79-124. [PMID: 37524985 DOI: 10.1007/978-3-031-30379-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
For the last two centuries, the scholarly education of histology and pathology has been based on technology, initially on the availability of low-cost, high-quality light microscopes, and more recently on the introduction of computers and e-learning approaches to biomedical education. Consequently, virtual microscopy (VM) is replacing glass slides and the traditional light microscope as the main instruments of instruction in histology and pathology laboratories. However, as with most educational changes, there are advantages and disadvantages associated with a new technology. The use of VM for the teaching of histology and pathology requires an extensive infrastructure and the availability of computing devices to all learners, both posing a considerable financial strain on schools and students. Furthermore, there may be valid reasons for practicing healthcare professionals to maintain competency in using light microscopes. In addition, some educators may be reluctant to embrace new technologies. These are some of the reasons why the introduction of VM as an integral part of histology and pathology instruction has been globally uneven. This paper compares the teaching of histology and pathology using traditional or VM in five different countries and their adjacent regions, representing developed, as well as developing areas of the globe. We identify general and local roadblocks to the introduction of this still-emerging didactic technology and outline solutions for overcoming these barriers.
Collapse
Affiliation(s)
- Michael Hortsch
- Departments of Cell and Developmental Biology and of Learning Health Sciences, University of Michigan, Ann Arbor, MI, USA.
| | - Nii Koney-Kwaku Koney
- Department of Anatomy, University of Ghana Medical School, University of Ghana, Korle Bu, Accra, Ghana
| | - Aswathy Maria Oommen
- Government Medical College Thiruvananthapuram, Thiruvananthapuram, Kerala, India
- Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Doris George Yohannan
- Government Medical College Thiruvananthapuram, Thiruvananthapuram, Kerala, India
- Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Yan Li
- Department of Anatomy, Histology and Embryology, Fudan University, Shanghai, China
| | | | | |
Collapse
|
15
|
Pisapia P, L'Imperio V, Galuppini F, Sajjadi E, Russo A, Cerbelli B, Fraggetta F, d'Amati G, Troncone G, Fassan M, Fusco N, Pagni F, Malapelle U. The evolving landscape of anatomic pathology. Crit Rev Oncol Hematol 2022; 178:103776. [PMID: 35934262 DOI: 10.1016/j.critrevonc.2022.103776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/11/2022] Open
Abstract
Anatomic pathology has changed dramatically in recent years. Although the microscopic assessment of tissues and cells is and will remain the mainstay of cancer diagnosis molecular profiling has become equally relevant. Thus, to stay abreast of the evolving landscape of today's anatomic pathology, modern pathologists must be able to master the intricate world of predictive molecular pathology. To this aim, pathologists have had to acquire additional knowledge to bridge the gap between clinicians and molecular biologists. This new role is particularly important, as cases are now collegially discussed in molecular tumor boards (MTBs). Moreover, as opposed to traditional pathologists, modern pathologists have also adamantly embraced innovation while keeping a constant eye on tradition. In this article, we depict the highlights and shadows of the upcoming "Anatomic Pathology 2.0" by placing particular emphasis on the pathologist's growing role in the management of cancer patients.
Collapse
Affiliation(s)
- Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, University of Milan-Bicocca (UNIMIB), Monza, Italy
| | - Francesca Galuppini
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua, Padua, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Bruna Cerbelli
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Filippo Fraggetta
- Pathology Unit, Gravina Hospital Caltagirone, ASP Catania, Caltagirone, Italy
| | - Giulia d'Amati
- Department of Radiology, Oncology and Pathology, Sapienza, University of Rome, Rome, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Matteo Fassan
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua, Padua, Italy; Veneto Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Padua, Veneto, Italy.
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, University of Milan-Bicocca (UNIMIB), Monza, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
16
|
Machine learning-enabled cancer diagnostics with widefield polarimetric second-harmonic generation microscopy. Sci Rep 2022; 12:10290. [PMID: 35717344 PMCID: PMC9206659 DOI: 10.1038/s41598-022-13623-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/03/2022] [Indexed: 11/08/2022] Open
Abstract
The extracellular matrix (ECM) collagen undergoes major remodeling during tumorigenesis. However, alterations to the ECM are not widely considered in cancer diagnostics, due to mostly uniform appearance of collagen fibers in white light images of hematoxylin and eosin-stained (H&E) tissue sections. Polarimetric second-harmonic generation (P-SHG) microscopy enables label-free visualization and ultrastructural investigation of non-centrosymmetric molecules, which, when combined with texture analysis, provides multiparameter characterization of tissue collagen. This paper demonstrates whole slide imaging of breast tissue microarrays using high-throughput widefield P-SHG microscopy. The resulting P-SHG parameters are used in classification to differentiate tumor from normal tissue, resulting in 94.2% for both accuracy and F1-score, and 6.3% false discovery rate. Subsequently, the trained classifier is employed to predict tumor tissue with 91.3% accuracy, 90.7% F1-score, and 13.8% false omission rate. As such, we show that widefield P-SHG microscopy reveals collagen ultrastructure over large tissue regions and can be utilized as a sensitive biomarker for cancer diagnostics and prognostics studies.
Collapse
|
17
|
Su A, Lee H, Tan X, Suarez CJ, Andor N, Nguyen Q, Ji HP. A deep learning model for molecular label transfer that enables cancer cell identification from histopathology images. NPJ Precis Oncol 2022; 6:14. [PMID: 35236916 PMCID: PMC8891271 DOI: 10.1038/s41698-022-00252-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Deep-learning classification systems have the potential to improve cancer diagnosis. However, development of these computational approaches so far depends on prior pathological annotations and large training datasets. The manual annotation is low-resolution, time-consuming, highly variable and subject to observer variance. To address this issue, we developed a method, H&E Molecular neural network (HEMnet). HEMnet utilizes immunohistochemistry as an initial molecular label for cancer cells on a H&E image and trains a cancer classifier on the overlapping clinical histopathological images. Using this molecular transfer method, HEMnet successfully generated and labeled 21,939 tumor and 8782 normal tiles from ten whole-slide images for model training. After building the model, HEMnet accurately identified colorectal cancer regions, which achieved 0.84 and 0.73 of ROC AUC values compared to p53 staining and pathological annotations, respectively. Our validation study using histopathology images from TCGA samples accurately estimated tumor purity, which showed a significant correlation (regression coefficient of 0.8) with the estimation based on genomic sequencing data. Thus, HEMnet contributes to addressing two main challenges in cancer deep-learning analysis, namely the need to have a large number of images for training and the dependence on manual labeling by a pathologist. HEMnet also predicts cancer cells at a much higher resolution compared to manual histopathologic evaluation. Overall, our method provides a path towards a fully automated delineation of any type of tumor so long as there is a cancer-oriented molecular stain available for subsequent learning. Software, tutorials and interactive tools are available at: https://github.com/BiomedicalMachineLearning/HEMnet.
Collapse
Affiliation(s)
- Andrew Su
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xiao Tan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Carlos J Suarez
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Noemi Andor
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, 94304, USA.
| |
Collapse
|
18
|
Kang L, Li X, Zhang Y, Wong TTW. Deep learning enables ultraviolet photoacoustic microscopy based histological imaging with near real-time virtual staining. PHOTOACOUSTICS 2022; 25:100308. [PMID: 34703763 PMCID: PMC8521289 DOI: 10.1016/j.pacs.2021.100308] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 06/02/2023]
Abstract
Histological images can reveal rich cellular information of tissue sections, which are widely used by pathologists in disease diagnosis. However, the gold standard for histopathological examination is based on thin sections on slides, which involves inevitable time-consuming and labor-intensive tissue processing steps, hindering the possibility of intraoperative pathological assessment of the precious patient specimens. Here, by incorporating ultraviolet photoacoustic microscopy (UV-PAM) with deep learning, we show a rapid and label-free histological imaging method that can generate virtually stained histological images (termed Deep-PAM) for both thin sections and thick fresh tissue specimens. With the tissue non-destructive nature of UV-PAM, the imaged intact specimens can be reused for other ancillary tests. We demonstrated Deep-PAM on various tissue preparation protocols, including formalin-fixation and paraffin-embedding sections (7-µm thick) and frozen sections (7-µm thick) in traditional histology, and rapid assessment of intact fresh tissue (~ 2-mm thick, within 15 min for a tissue with a surface area of 5 mm × 5 mm). Deep-PAM potentially serves as a comprehensive histological imaging method that can be simultaneously applied in preoperative, intraoperative, and postoperative disease diagnosis.
Collapse
Affiliation(s)
- Lei Kang
- Translational and Advanced Bioimaging Laboratory, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Xiufeng Li
- Translational and Advanced Bioimaging Laboratory, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Yan Zhang
- Translational and Advanced Bioimaging Laboratory, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Terence T W Wong
- Translational and Advanced Bioimaging Laboratory, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| |
Collapse
|
19
|
Gibilisco F, Fraggetta F, Williams BJ. A commemoration of the "digital" side of Juan Rosai: a junior's perspective of the legacy of an all-round pathologist. Pathologica 2021; 113:305-306. [PMID: 34837088 PMCID: PMC8720403 DOI: 10.32074/1591-951x-444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 10/10/2021] [Indexed: 11/30/2022] Open
Abstract
Juan Rosai, the “Maradona” of surgical pathology, played a role not only as a diagnostician but also as a researcher, a consultant and a teacher, distinguishing himself as a real icon at all levels of modern pathology. He was an innovative promoter of emerging technologies including digital pathology. These few lines commemorate the digital side of the “Maestro” Juan Rosai from a junior’s perspective highlighting how Rosai supported digital pathology and remembering that, according to his own words, digital pathology “will revolutionize the field of pathology, if it is not doing that already”.
Collapse
Affiliation(s)
- Fabio Gibilisco
- Department of Medical and Surgical Sciences and Advanced Technologies, "G. F. Ingrassia", Anatomic Pathology, University of Catania, Catania, Italy
| | | | - Bethany Jill Williams
- Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.,Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
20
|
Agaimy A. The challenge of undifferentiated malignancies: Phenotype versus genotype! What matters most? Semin Diagn Pathol 2021; 38:117-118. [PMID: 34556375 DOI: 10.1053/j.semdp.2021.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, Krankenhausstrasse 8-10, 91054 Erlangen, Germany.
| |
Collapse
|
21
|
Chatzopoulos K, Sotiriou S, Collins AR, Kartsidis P, Schmitt AC, Chen X, Khazaie K, Hinni ML, Ramsower CA, Zarka MA, Patel SH, Garcia JJ. Transcriptomic and Immunophenotypic Characterization of Tumor Immune Microenvironment in Squamous Cell Carcinoma of the Oral Tongue. Head Neck Pathol 2021; 15:509-522. [PMID: 33010009 PMCID: PMC8134601 DOI: 10.1007/s12105-020-01229-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022]
Abstract
The tumor immune microenvironment of oral tongue squamous cell carcinoma may be accountable for differences in clinical behavior, particularly between different age groups. We performed RNA expression profiling and evaluated tumor infiltrating lymphocytes (TILs) and their T-cell subsets in order to assess the functional status of oral tongue squamous cell carcinoma tumor microenvironment and detect potentially clinically useful associations. Archival surgical pathology material from sixteen oral tongue squamous cell carcinoma patients was microscopically evaluated for TIL densities. RNA was extracted from macrodissected whole tumor sections and normal controls and RNA expression profiling was performed by the NanoString PanCancer IO 360 Gene Expression Panel. Immunostains for CD4, CD8 and FOXP3 were evaluated manually and by digital image analysis. Oral tongue squamous cell carcinomas had increased TIL densities, numerically dominated by CD4 + T cells, followed by CD8 + and FOXP3 + T cells. RNA expression profiling of tumors versus normal controls showed tumor signature upregulation in inhibitory immune signaling (CTLA4, TIGIT and PD-L2), followed by inhibitory tumor mechanisms (IDO1, TGF-β, B7-H3 and PD-L1). Patients older than 44 years showed a tumor microenvironment with increased Tregs and CTLA4 expression. Immunohistochemically assessed CD8% correlated well with molecular signatures related to CD8 + cytotoxic T-cell functions. FOXP3% correlated significantly with CTLA4 upregulation. CTLA4 molecular signature could be predicted by FOXP3% assessed by immunohistochemistry (R2 = 0.619, p = 0.026). Oral tongue squamous cell carcinoma hosts a complex inhibitory immune microenvironment, partially reflected in immunohistochemically quantified CD8 + and FOXP3 + T-cell subsets. Immunohistochemistry can be a useful screening tool for detecting tumors with upregulated expression of the targetable molecule CTLA4.
Collapse
Affiliation(s)
- Kyriakos Chatzopoulos
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
- Division of Anatomic Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Sotiris Sotiriou
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| | - Andrea R. Collins
- Mayo Clinic Alix School of Medicine, 200 1st St SW, Rochester, MN 55905 USA
| | - Panagiotis Kartsidis
- Laboratory of Medical Physics, Medical School, Faculty of Health Sciences, Aristotle University of Thessaloniki, P.O. Box 376, 54124 Thessaloníki, Greece
| | - Alessandra C. Schmitt
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259 USA
| | - Xianfeng Chen
- Department of Research Biostatistics, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259 USA
| | | | - Michael L. Hinni
- Department of Otolaryngology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259 USA
| | - Colleen A. Ramsower
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259 USA
| | - Matthew A. Zarka
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259 USA
| | - Samir H. Patel
- Department of Radiation Oncology, Mayo Clinic, 13400 E. Shea Blvd., Scottsdale, AZ 85259 USA
| | - Joaquin J. Garcia
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
- Division of Anatomic Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905 USA
| |
Collapse
|
22
|
Suarez-Zamora DA, Mustafa RA, Estrada-Orozco K, Rodriguez-Urrego PA, Torres-Franco F, Barreto-Hauzeur L, Mora-Ochoa H, Di Tanna GL, Yepes-Nuñez JJ. Intraoperative sub-areolar frozen section analysis for detecting nipple involvement in candidates for nipple-sparing mastectomy. Hippokratia 2021. [DOI: 10.1002/14651858.cd014702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- David A Suarez-Zamora
- Department of Pathology and Laboratories; Fundación Santa Fe de Bogotá University Hospital; Bogota Colombia
| | - Reem A Mustafa
- Department of Internal Medicine, Division of Nephrology and Hypertension; University of Kansas Medical Center; Kansas City Missouri USA
- Department of Health Research Methods, Evidence and Impact; McMaster University; Hamilton Canada
| | - Kelly Estrada-Orozco
- Health Technologies and Politics Assessment Group, Clinical Research Institute; National University of Colombia; Bogota Colombia
| | - Paula A Rodriguez-Urrego
- Department of Pathology and Laboratories; Fundación Santa Fe de Bogotá University Hospital; Bogota Colombia
| | - Fabio Torres-Franco
- Division of Breast Surgery; Fundación Santa Fe de Bogotá University Hospital; Bogota Colombia
| | - Lisette Barreto-Hauzeur
- Division of Plastic and Reconstructive Surgery; Fundación Santa Fe de Bogotá University Hospital; Bogota Colombia
| | | | - Gian Luca Di Tanna
- Faculty of Medicine; University of New South Wales; Sydney Australia
- Statistics Division; The George Institute for Global Health; Newtown Australia
| | | |
Collapse
|
23
|
Deepa PR, Nalini V, Surianarayanan M, Krishnakumar S. Towards safer non-volatile tissue fixatives: Evaluation of choline-based ionic liquids for fixing ocular tissues. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 209:111777. [PMID: 33352431 DOI: 10.1016/j.ecoenv.2020.111777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/19/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
Volatile organic chemicals (VOCs) are routinely used for processing biological tissue samples in clinical laboratories. Recognizing their serious health and environmental impacts, a few non-volatile green solvents (choline based ionic liquids, ILs) were evaluated as tissue fixatives here. Microscopic evaluation of histo-morphology, fixation and staining quality, and macromolecular integrity (DNA and proteins) were assessed in human eye tissues (sclera, choroid, retinal layers and retinal pigmented epithelium, eyelid and orbit) after IL-fixation. Formalin-fixed tissues were used as standard reference. Microscopic examination revealed favorable histomorphology, tissue fixation and staining characteristics in most tissues immersed in ILs. Time taken to fix, and stability over a period of time (24 h, 48 h, 1 week, 1 month) was also recorded. Electrophoretic analysis revealed stability of cellular proteins and nucleic acids in IL-fixed scleral tissues. Heterogeneity in tissue fixation property relative to the type of ocular tissue, duration of fixation and storage, warrant further design and optimization of ILs to fix biological tissues. The simple cholinium salts based ILs tested here show favorable potential for tissue fixation application, and as an alternative approach to the use of VOCs, towards sustainable biomedical practice.
Collapse
Affiliation(s)
- P R Deepa
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS), Pilani 333 031, Rajasthan, India.
| | - V Nalini
- Department of Biological Sciences, Birla Institute of Technology & Science (BITS), Pilani 333 031, Rajasthan, India; L&T Department of Ocular Pathology, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Sankara Nethralaya, 18, College Road, 600 006 Chennai, India
| | - M Surianarayanan
- Cell for Industrial Safety and Risk Analysis (CISRA), Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai, India
| | - S Krishnakumar
- L&T Department of Ocular Pathology, Kamalnayan Bajaj Institute for Research in Vision and Ophthalmology, Vision Research Foundation, Sankara Nethralaya, 18, College Road, 600 006 Chennai, India.
| |
Collapse
|
24
|
Chatzopoulos K, Kotoula V, Koliou GA, Giannoulatou E, Papadopoulou K, Karavasilis V, Pazarli E, Pervana S, Kafiri G, Tsoulfas G, Chrisafi S, Sgouramali H, Papakostas P, Pectasides D, Hytiroglou P, Pentheroudakis G, Fountzilas G. Genotype-phenotype associations in colorectal adenocarcinomas and their matched metastases. Hum Pathol 2021; 107:104-116. [PMID: 33161028 DOI: 10.1016/j.humpath.2020.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/03/2020] [Accepted: 10/30/2020] [Indexed: 02/05/2023]
Abstract
Although primary colorectal carcinomas (CRCs) frequently share genetic alterations with their metastases, morphologic surrogates reflecting the genotype contexture of metastases remain largely unknown. We investigated phenotype/genotype associations in paired primary and metastatic colorectal adenocarcinomas from 75 patients. Thirty-three (44%) metastatic lesions were synchronous and 42 (56%) were metachronous. Tumor budding, micronecrosis, and tumor-infiltrating lymphocyte (TIL) density were compared with matched next-generation sequencing genotypes. Micronecrosis in the primary were significantly associated with nodal status (P = 0.0054) and with micronecrosis in metastatic sites (P = 0.0216), particularly in metachronous metastases (P = 0.0033). With a 57-gene panel, one or more mutations were identified in 64 (85.3%) cases. In metastases, high (brisk) TILs were associated with overall mutational burden (P = 0.0058) and with mutations in EGF (P = 0.0325), RAS genes (P = 0.0043), and MMR genes (P = 0.0069), whereas high-level micronecrosis correlated with mutations in APC (P = 0.0004) and MSH6 (P = 0.0385) genes. Genomic alterations were shared in 90.1% of primary/metastatic pairs, but clonality of the same mutation was shared in only 57.1% of paired lesions. Compared with synchronous, metachronous metastases had more private clonal alterations (P = 0.0291); in this group, clonal alterations coincided with brisk TILs (P = 0.0334) and high micronecrosis (P = 0.0133). High TILs in metastatic lesions were predictive of favorable overall survival (log-rank P = 0.044). The observed phenotype/genotype associations favor the clonal evolution model in CRC metastases that seems accompanied by intense host immune response. If the role of micronecrosis and brisk TILs in metachronous metastases is validated in larger studies, these histologic parameters will be worth adding in the armamentarium for the evaluation of metastatic CRC.
Collapse
Affiliation(s)
- Kyriakos Chatzopoulos
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece; Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, 54124, Greece.
| | - Vassiliki Kotoula
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece; Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, 54124, Greece
| | - Georgia-Angeliki Koliou
- Section of Biostatistics, Hellenic Cooperative Oncology Group, Data Office, Athens, 11524, Greece
| | - Eleni Giannoulatou
- Computational Genomics Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia; The University of New South Wales, Kensington, NSW, 2052, Australia
| | - Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Vasilios Karavasilis
- Department of Medical Oncology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, 56403, Greece
| | - Elissavet Pazarli
- Department of Pathology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, 56403, Greece
| | - Stavroula Pervana
- Department of Pathology, Papageorgiou Hospital, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, 56403, Greece
| | - Georgia Kafiri
- Department of Pathology, Hippokration Hospital, Athens, 11527, Greece
| | - Georgios Tsoulfas
- Department of Surgery, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Sofia Chrisafi
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Helen Sgouramali
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Pavlos Papakostas
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, 11527, Greece
| | - Dimitrios Pectasides
- Oncology Section, Second Department of Internal Medicine, Hippokration Hospital, Athens, 11527, Greece
| | - Prodromos Hytiroglou
- Department of Pathology, Aristotle University of Thessaloniki, School of Health Sciences, Faculty of Medicine, Thessaloniki, 54124, Greece
| | - George Pentheroudakis
- Department of Medical Oncology, Medical School, University of Ioannina, Ioannina, 45500, Greece; Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), Ioannina, Greece
| | - George Fountzilas
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research/Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece; Aristotle University of Thessaloniki, Thessaloniki, Greece; German Oncology Center, Limassol, Cyprus
| |
Collapse
|
25
|
Ščupáková K, Balluff B, Tressler C, Adelaja T, Heeren RM, Glunde K, Ertaylan G. Cellular resolution in clinical MALDI mass spectrometry imaging: the latest advancements and current challenges. Clin Chem Lab Med 2020; 58:914-929. [PMID: 31665113 PMCID: PMC9867918 DOI: 10.1515/cclm-2019-0858] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/07/2019] [Indexed: 02/07/2023]
Abstract
Mass spectrometry (MS) is the workhorse of metabolomics, proteomics and lipidomics. Mass spectrometry imaging (MSI), its extension to spatially resolved analysis of tissues, is a powerful tool for visualizing molecular information within the histological context of tissue. This review summarizes recent developments in MSI and highlights current challenges that remain to achieve molecular imaging at the cellular level of clinical specimens. We focus on matrix-assisted laser desorption/ionization (MALDI)-MSI. We discuss the current status of each of the analysis steps and remaining challenges to reach the desired level of cellular imaging. Currently, analyte delocalization and degradation, matrix crystal size, laser focus restrictions and detector sensitivity are factors that are limiting spatial resolution. New sample preparation devices and laser optic systems are being developed to push the boundaries of these limitations. Furthermore, we review the processing of cellular MSI data and images, and the systematic integration of these data in the light of available algorithms and databases. We discuss roadblocks in the data analysis pipeline and show how technology from other fields can be used to overcome these. Finally, we conclude with curative and community efforts that are needed to enable contextualization of the information obtained.
Collapse
Affiliation(s)
- Klára Ščupáková
- Maastricht MultiModal Molecular Imaging Institute (M4I), University of Maastricht, Maastricht, The Netherlands
| | - Benjamin Balluff
- Maastricht MultiModal Molecular Imaging Institute (M4I), University of Maastricht, Maastricht, The Netherlands
| | - Caitlin Tressler
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tobi Adelaja
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ron M.A. Heeren
- Corresponding author: Ron M.A. Heeren, Maastricht MultiModal Molecular Imaging Institute (M4I), University of Maastricht, Maastricht, The Netherlands,
| | - Kristine Glunde
- Russell H. Morgan Department of Radiology and Radiological Science, Division of Cancer Imaging Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA; and The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Gökhan Ertaylan
- Unit Health, Flemish Institute for Technological Research (VITO), Mol, Belgium
| |
Collapse
|
26
|
|
27
|
Himmel LE, Hackett TA, Moore JL, Adams WR, Thomas G, Novitskaya T, Caprioli RM, Zijlstra A, Mahadevan-Jansen A, Boyd KL. Beyond the H&E: Advanced Technologies for in situ Tissue Biomarker Imaging. ILAR J 2018; 59:51-65. [PMID: 30462242 PMCID: PMC6645175 DOI: 10.1093/ilar/ily004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/27/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023] Open
Abstract
For decades, histopathology with routine hematoxylin and eosin staining has been and remains the gold standard for reaching a morphologic diagnosis in tissue samples from humans and veterinary species. However, within the past decade, there has been exponential growth in advanced techniques for in situ tissue biomarker imaging that bridge the divide between anatomic and molecular pathology. It is now possible to simultaneously observe localization and expression magnitude of multiple protein, nucleic acid, and molecular targets in tissue sections and apply machine learning to synthesize vast, image-derived datasets. As these technologies become more sophisticated and widely available, a team-science approach involving subspecialists with medical, engineering, and physics backgrounds is critical to upholding quality and validity in studies generating these data. The purpose of this manuscript is to detail the scientific premise, tools and training, quality control, and data collection and analysis considerations needed for the most prominent advanced imaging technologies currently applied in tissue sections: immunofluorescence, in situ hybridization, laser capture microdissection, matrix-assisted laser desorption ionization imaging mass spectrometry, and spectroscopic/optical methods. We conclude with a brief overview of future directions for ex vivo and in vivo imaging techniques.
Collapse
Affiliation(s)
- Lauren E Himmel
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Troy A Hackett
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Jessica L Moore
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Wilson R Adams
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Giju Thomas
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Tatiana Novitskaya
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Richard M Caprioli
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Andries Zijlstra
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Anita Mahadevan-Jansen
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| | - Kelli L Boyd
- Lauren E. Himmel, DVM, PhD, is an assistant professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee. Troy A. Hackett, PhD, is a professor in the Department of Hearing and Speech Sciences at Vanderbilt University Medical Center in Nashville, Tennessee. Jessica L. Moore, PhD, is a postdoctoral research fellow in the Mass Spectrometry Research Center at the Vanderbilt University School of Medicine in Nashville, Tennessee. Wilson R. Adams, BS, is graduate student in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Giju Thomas, PhD, is a post-doctoral researcher in the Biophotonics Center and Department of Biomedical Engineering at Vanderbilt University in Nashville, Tennessee. Tatiana Novitskaya, MD, PhD, is a staff scientist in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center. Richard M. Caprioli, PhD, is a professor in the Department of Chemistry at the Vanderbilt University School of Medicine in Nashville, Tennessee. Andries Zijlstra, PhD, is an associate professor in the Department of Pathology, Microbiology and Immunology at Vanderbilt University Medical Center in Nashville, Tennessee. Anita Mahadevan-Jansen, PhD, is a professor in the Department of Biomedical Engineering at the Vanderbilt University School of Engineering and Department of Neurosurgery at Vanderbilt University Medical Center in Nashville, Tennessee. Kelli L. Boyd, DVM, PhD, is a professor and veterinary pathologist in the Division of Comparative Medicine at Vanderbilt University Medical Center in Nashville, Tennessee
| |
Collapse
|
28
|
Ortiz-Hidalgo C, Pina-Oviedo S. Hematoxylin: Mesoamerica’s Gift to Histopathology. Palo de Campeche (Logwood Tree), Pirates’ Most Desired Treasure, and Irreplaceable Tissue Stain. Int J Surg Pathol 2018; 27:4-14. [DOI: 10.1177/1066896918787652] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hematoxylin is a basic dye derived from the heartwood of Palo de Campeche ( Haematoxylum campechianum), the logwood tree native to Mexico and Central America. Haematoxylum means “bloodwood” in reference to its dark-red heartwood and campechianum refers to its site of origin, the coastal city of Campeche on the Yucatan Peninsula, Mexico. Hematoxylin is colorless but it turns into the color dye hematein after oxidation (ripening). The dyeing property of logwood was well-known to the natives of the Yucatan Peninsula before the arrival of the Spaniards who brought it to Europe shortly after the discovery of the Americas. An important trade soon developed related to growing and preparing hematoxylin for dyeing fabrics. Pirates discovered that one shipload of logwood was equivalent to a year’s value from any other cargo, and by 1563, more than 400 pirate vessels wandered the Atlantic Ocean and attacked Spanish galleons transporting gold, silver, and logwood from the Americas to Europe. Hematoxylin and eosin is a staining method that dates back to the late 19th century. In 1865 and 1891, Böhmer and Meyer, respectively, first used hematoxylin in combination with a mordant (alum). Later, with the use of anilines by Ehrlich, the repertoire of stains expanded rapidly resulting in the microscopic descriptions of multiple diseases that were defined by their stainable features. Today hematoxylin, along with eosin, remains the most popular stain in histology.
Collapse
Affiliation(s)
- Carlos Ortiz-Hidalgo
- Hospital y Fundación Médica Sur, Mexico City, Mexico
- Universidad Panamericana, Mexico City, Mexico
| | | |
Collapse
|
29
|
Pichat J, Iglesias JE, Yousry T, Ourselin S, Modat M. A Survey of Methods for 3D Histology Reconstruction. Med Image Anal 2018; 46:73-105. [DOI: 10.1016/j.media.2018.02.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 02/02/2018] [Accepted: 02/14/2018] [Indexed: 02/08/2023]
|
30
|
Sinha N, Nayak MT, Sunitha JD, Dawar G, Rallan N, Gupta S. Comparitive efficacies of a natural fixative with a conventional fixative. J Oral Maxillofac Pathol 2018; 21:458. [PMID: 29391731 PMCID: PMC5763879 DOI: 10.4103/jomfp.jomfp_236_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Introduction: The quest for formalin substitutes has long been going on due to its health hazards. Honey has been recognized as a safe substitute for formalin. However, we explored jaggery as a natural substitute for formalin. The aim of this study was to compare the tissue fixation abilities of jaggery syrup (30%) with that of 10% neutral-buffered formalin (NBF) and to determine the best fixative among both. Materials and Methods: A study was conducted with 65 pathological tissues. Each specimen was divided into two equal parts. One part was fixed in 30% jaggery solution (Group A), while the other half was fixed in 10% NBF solution (Group B). 24 h tissue fixation was attained at room temperature followed by evaluation of pre- and post-fixation, tissue shrinkage, weight difference and ease of sectioning, followed by evaluation of conventional processing and staining. The histomorphological assessment for each slide was made based on evaluation of cellular outline, cytoplasmic details, nuclear details, staining quality and overall morphology under light microscopy. Each criterion was rated on a scale of 1–4. Nominal categorical data between the groups were compared using Chi-squared test. Results: The preservation of tissue specimen by jaggery syrup was comparable to that of formalin and surprisingly overall nuclear detail of the tissue was better than conventional formalin fixative. Conclusion: Jaggery can be successfully adopted in routine histopathology laboratories in place of formalin.
Collapse
Affiliation(s)
- Nidhi Sinha
- Department of Oral Pathology and Microbiology, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Meghanand T Nayak
- Department of Oral Pathology and Microbiology, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| | - J D Sunitha
- Department of Oral Pathology and Microbiology, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Geetanshu Dawar
- Department of Oral Pathology and Microbiology, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Neelakshi Rallan
- Department of Oral Pathology and Microbiology, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| | - Shreya Gupta
- Department of Oral Pathology and Microbiology, Teerthanker Mahaveer Dental College and Research Centre, Moradabad, Uttar Pradesh, India
| |
Collapse
|
31
|
Underwood JCE. More than meets the eye: the changing face of histopathology. Histopathology 2017; 70:4-9. [PMID: 27960234 PMCID: PMC7165712 DOI: 10.1111/his.13047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/24/2016] [Indexed: 01/25/2023]
Abstract
This personal reflection on trends in histopathology over the past 50 years draws upon experience of professional training and practice in the specialty in the UK. Developments during this period often resulted from new therapies (and their adverse effects) necessitating greater precision in the histopathological classification of disease, for which morphology alone can be insufficient. Conversely, histopathology has contributed to advances in our understanding of disease, leading directly to novel and more effective treatments. New infections, some involving histopathology in their discovery, have also led to fresh diagnostic challenges. Increasingly, patients have benefited from fundamental changes in professionalism in pathology. Through audit, external quality assurance, continuing professional development, standardized reporting, and increasing specialization, the consistency and reliability of histopathological diagnoses have steadily improved. Regarding the specialty's future, some now see rivalry between the morphological and molecular approaches to diagnosis and classification, particularly for neoplastic disease. An integrated strategy led by the specialty is more likely to strengthen histopathology and ultimately to have the greatest benefit for patients.
Collapse
|
32
|
Granter SR. Reports of the Death of the Microscope Have Been Greatly Exaggerated. Arch Pathol Lab Med 2016; 140:744-5. [DOI: 10.5858/arpa.2016-0046-ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Scott R. Granter
- From the Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
33
|
Treuting PM, Snyder JM, Ikeno Y, Schofield PN, Ward JM, Sundberg JP. The Vital Role of Pathology in Improving Reproducibility and Translational Relevance of Aging Studies in Rodents. Vet Pathol 2016; 53:244-9. [PMID: 26792843 PMCID: PMC4835687 DOI: 10.1177/0300985815620629] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Pathology is a discipline of medicine that adds great benefit to aging studies of rodents by integrating in vivo, biochemical, and molecular data. It is not possible to diagnose systemic illness, comorbidities, and proximate causes of death in aging studies without the morphologic context provided by histopathology. To date, many rodent aging studies do not utilize end points supported by systematic necropsy and histopathology, which leaves studies incomplete, contradictory, and difficult to interpret. As in traditional toxicity studies, if the effect of a drug, dietary treatment, or altered gene expression on aging is to be studied, systematic pathology analysis must be included to determine the causes of age-related illness, moribundity, and death. In this Commentary, the authors discuss the factors that should be considered in the design of aging studies in mice, with the inclusion of robust pathology practices modified after those developed by toxicologic and discovery research pathologists. Investigators in the field of aging must consider the use of histopathology in their rodent aging studies in this era of integrative and preclinical geriatric science (geroscience).
Collapse
Affiliation(s)
- P M Treuting
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - J M Snyder
- Department of Comparative Medicine, School of Medicine, University of Washington, Seattle, WA, USA
| | - Y Ikeno
- Barshop Institute and Department of Pathology, University of Texas Health Science Center at San Antonio; Research Service and Geriatric Research and Education Clinical Center, Audie L. Murphy VA Hospital, South Texas Veterans Health Care System, San Antonio, TX, USA
| | - P N Schofield
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK The Jackson Laboratory, Bar Harbor, ME, USA
| | - J M Ward
- Global VetPathology, Montgomery Village, MD, USA
| | | |
Collapse
|
34
|
Rodriguez-Canales J, Parra-Cuentas E, Wistuba II. Diagnosis and Molecular Classification of Lung Cancer. Cancer Treat Res 2016; 170:25-46. [PMID: 27535388 DOI: 10.1007/978-3-319-40389-2_2] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Lung cancer is a complex disease composed of diverse histological and molecular types with clinical relevance. The advent of large-scale molecular profiling has been helpful to identify novel molecular targets that can be applied to the treatment of particular lung cancer patients and has helped to reshape the pathological classification of lung cancer. Novel directions include the immunotherapy revolution, which has opened the door for new opportunities for cancer therapy and is also redefining the classification of multiple tumors, including lung cancer. In the present chapter, we will review the main current basis of the pathological diagnosis and classification of lung cancer incorporating the histopathological and molecular dimensions of the disease.
Collapse
Affiliation(s)
- Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA
| | - Edwin Parra-Cuentas
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, Unit 951, The University of Texas MD Anderson Cancer Center, 2130 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Ramírez-Romero R, Silva-Pérez RA, Lara-Arias J, Ramírez-Hernández C, Marino-Martínez IA, Barbosa-Quintana Á, López-Mayagoitia A. Coccidioidomycosis in Biopsies with Presumptive Diagnosis of Malignancy in Dogs: Report of Three Cases and Comparative Discussion of Published Reports. Mycopathologia 2015; 181:151-7. [PMID: 26419685 PMCID: PMC4676787 DOI: 10.1007/s11046-015-9948-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/16/2015] [Indexed: 12/01/2022]
Abstract
Coccidioidomycosis is a respiratory fungal infection with occasional systemic dissemination. The disseminated coccidioidomycosis is considered a multifaceted disease. In medicine, disseminated coccidioidomycosis is included within a group of infectious diseases that have been referred as the great imitators. In many cases, malignancies are included in the presumptive diagnosis. In veterinary medicine, disseminated coccidioidomycosis is common in dogs. Nonetheless, despite of being a diagnostic dilemma, disseminated coccidioidomycosis is underestimated and frequently not included into differentials, even in endemic zones. Herein, we describe three cases of granulomatous inflammation caused by Coccidioides spp. which were masquerading malignancies in dogs (0.39 %). The presumptive diagnoses in these cases were osteosarcoma, lymphoma and neurofibroma, respectively. A PCR assay employing tissues in paraffin blocks resulted positive for C. posadasii in one of these cases. A comparative discussion on the ambiguous clinic-pathological presentation of disseminated coccidioidomycosis in dogs and humans is included.
Collapse
Affiliation(s)
- Rafael Ramírez-Romero
- Posgrado Conjunto Agronomía-Veterinaria, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Agropecuarias, Universidad Autónoma de Nuevo León, Av. Francisco Villa s/n, Ex-Hacienda el Canadá, C.P. 66050, Gral. Escobedo, N.L., México.
| | - Rolando Antonio Silva-Pérez
- Posgrado Conjunto Agronomía-Veterinaria, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Agropecuarias, Universidad Autónoma de Nuevo León, Av. Francisco Villa s/n, Ex-Hacienda el Canadá, C.P. 66050, Gral. Escobedo, N.L., México
| | - Jorge Lara-Arias
- Banco de Hueso y Tejidos, Hospital Universitario "Dr. José E. González", Universidad Autónoma de Nuevo León, Monterrey, N.L., México
| | - Cecilia Ramírez-Hernández
- Posgrado Conjunto Agronomía-Veterinaria, Facultad de Medicina Veterinaria y Zootecnia, Campus de Ciencias Agropecuarias, Universidad Autónoma de Nuevo León, Av. Francisco Villa s/n, Ex-Hacienda el Canadá, C.P. 66050, Gral. Escobedo, N.L., México
| | - Iván Alberto Marino-Martínez
- Centro de Investigación y Desarrollo en Ciencias de la Salud (CIDICS), Universidad Autónoma de Nuevo León, Monterrey, N.L., México
| | - Álvaro Barbosa-Quintana
- Departamento de Anatomía Patológica, Hospital Universitario, Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, N.L., México
| | | |
Collapse
|
36
|
Does everything a surgeon takes out have to be seen by a pathologist? A review of the current pathology practice. Virchows Arch 2015; 468:69-74. [PMID: 26155913 DOI: 10.1007/s00428-015-1801-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 06/15/2015] [Indexed: 01/13/2023]
Abstract
Histopathologic examination of surgically removed tissues and organs is an important aspect of modern hospital quality health care. Most surgical specimens deserve to be submitted for pathologic examination, which may yield valuable new information relevant for the future treatment of the patient. A small number of specimens, recognized as providing limited or no valuable clinical data during pathologic examination, may be placed on the list of specimens "exempt from submission" or those that are labeled as "for gross examination only." Guidelines written by the committees of the national regulatory organizations provide general orientation on how to deal with various specimens, but the final decision on which type of specimen to eliminate and which ones to include for pathologic examination rests on local governing and advisory bodies of each institution. Particular lists of specimens exempt from pathologic examination are best generated through a consensus agreement of clinical and laboratory physicians. Even though there is general nationwide and even international consensus on which types of specimens deserve pathologic examination and which do not, there are still discussions about the necessity of some pathologic examinations.
Collapse
|
37
|
M R, A RP, G S, Jk SB, Ndvn S. Transit fixatives: an innovative study. J Clin Diagn Res 2015; 9:ZM01-3. [PMID: 25954725 DOI: 10.7860/jcdr/2015/11083.5615] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/22/2015] [Indexed: 11/24/2022]
Abstract
BACKGROUND Universally accepted fixative is 10% formalin which has been used for preserving the tissues and their architecture. In certain conditions, formalin might not be readily available for immediate fixation. We here by explore more economical, eco-friendly and easily available solutions that can be used as transit media/ transporting media for tissue specimens. MATERIALS AND METHODS The study included commonly available solutions like Spirit, Saline, Betadine solution, Hydrogen peroxide (H2O2), Local anesthesia (L.A), Rose water, Coconut oil, Coconut water, Ice cold water, Honey and Milk while keeping formalin as control. The fresh tissue sample was cut into multiple bits and placed in different containers for a period of 8 hours before transferring to formalin solution. CONCLUSION Transit fixatives are very important in certain situations where formalin is not readily available. These fixatives can be used to fix the tissues for a period of at least 8 hours without causing any damage or distortion before they are fixed in formalin solution.
Collapse
Affiliation(s)
- Rajanikanth M
- Reader, Department of Oral and Maxillofacial Pathology, G. Pulla Reddy Dental College and Hospital , India
| | - Ravi Prakash A
- Professor and HOD, Department of Oral and Maxillofacial Pathology, G. Pulla Reddy Dental College and Hospital , Kurnool, India
| | - Sreenath G
- Senior Lecturer, Department of Oral and Maxillofacial Pathology, G. Pulla Reddy Dental College and Hospital , Kurnool, India
| | - Sonia Bai Jk
- Post Graduate Student, Department of Oral and Maxillofacial Pathology, G. Pulla Reddy Dental College and Hospital , Kurnool, India
| | - Shyam Ndvn
- Professor, Department of Oral and Maxillofacial Pathology, Government Dental College- Hyderabad, India
| |
Collapse
|
38
|
Schöchlin M, Weissinger SE, Brandes AR, Herrmann M, Möller P, Lennerz JK. A nuclear circularity-based classifier for diagnostic distinction of desmoplastic from spindle cell melanoma in digitized histological images. J Pathol Inform 2014; 5:40. [PMID: 25379346 PMCID: PMC4221957 DOI: 10.4103/2153-3539.143335] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/06/2014] [Indexed: 01/12/2023] Open
Abstract
Context: Distinction of spindle cell melanoma (SM) and desmoplastic melanoma (DM) is clinically important due to differences in metastatic rate and prognosis; however, histological distinction is not always straightforward. During a routine review of cases, we noted differences in nuclear circularity between SM and DM. Aim: The primary aim in our study was to determine whether these differences in nuclear circularity, when assessed using a basic ImageJ-based threshold extraction, can serve as a diagnostic classifier to distinguish DM from SM. Settings and Design: Our retrospective analysis of an established patient cohort (SM n = 9, DM n = 9) was employed to determine discriminatory power. Subjects and Methods: Regions of interest (total n = 108; 6 images per case) were selected from scanned H and E-stained histological sections, and nuclear circularity was extracted and quantified by computational image analysis using open source tools (plugins for ImageJ). Statistical Analysis: Using analysis of variance, t-tests, and Fisher's exact tests, we compared extracted quantitative shape measures; statistical significance was defined as P < 0.05. Results: Classifying circularity values into four shape categories (spindled, elongated, oval, round) demonstrated significant differences in the spindled and round categories. Paradoxically, DM contained more spindled nuclei than SM (P = 0.011) and SM contained more round nuclei than DM (P = 0.026). Performance assessment using a combined shape-classification of the round and spindled fractions showed 88.9% accuracy and a Youden index of 0.77. Conclusions: Spindle cell melanoma and DM differ significantly in their nuclear morphology with respect to fractions of round and spindled nuclei. Our study demonstrates that quantifying nuclear circularity can be used as an adjunct diagnostic tool for distinction of DM and SM.
Collapse
Affiliation(s)
| | | | - Arnd R Brandes
- Institut für Lasertechnologien in der Medizin und Meβtechnik, University Ulm, Ulm, Germany
| | - Markus Herrmann
- Institute of Pathology, University Ulm, Ulm, Germany ; Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Peter Möller
- Institute of Pathology, University Ulm, Ulm, Germany
| | | |
Collapse
|
39
|
Kang YK. Diminutive and Small Colorectal Polyps: The Pathologist's Perspective. Clin Endosc 2014; 47:404-8. [PMID: 25324998 PMCID: PMC4198555 DOI: 10.5946/ce.2014.47.5.404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/05/2014] [Indexed: 01/09/2023] Open
Abstract
Recent progress in advanced endoscopic imaging and electronic chromoendoscopy allows the real-time endoscopic estimation of the histologic type of polyps, mainly for the differentiation of adenomas from hyperplastic polyps. Accordingly, a "resect-and-discard" strategy applied to diminutive colorectal polyps is now one of the emerging issues among gastroenterologists. The strategy has a practical advantage in terms of the potential cost savings. However, it has a number of limitations in the medical, academic, and legal aspects. The major pitfalls include the endoscopic investigation of colorectal polyps with a wide variety of histogenetic origins, including serrated polyps, and the lack of a standardized method for polyp size measurement. Another issue is the importance of the pathologic diagnosis for legal purposes and medical research. Moreover, it is not certain whether the implementation of the strategy has economic benefit in countries with an undervalued reimbursement system for pathologic examination. There is no doubt that a highly confident optical diagnosis of polyp type is a novel valuable tool. It can provide a more steady symbiosis between gastroenterologists and pathologists to allow a more evident diagnosis and management of patients with colorectal polyps.
Collapse
Affiliation(s)
- Yun Kyung Kang
- Department of Pathology, Inje University Seoul Paik Hospital, Inje University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
|
41
|
ΔNp63 (p40) and thyroid transcription factor-1 immunoreactivity on small biopsies or cellblocks for typing non-small cell lung cancer: a novel two-hit, sparing-material approach. J Thorac Oncol 2012; 7:281-90. [PMID: 22071786 DOI: 10.1097/jto.0b013e31823815d3] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Diagnosing non-small cell lung cancer on biopsy/cellblock samples by morphology may be demanding. As sparing material for molecular testing is mandatory, a minimalist immunohistochemistry (IHC)-based diagnostic approach is warranted by means of novel, reliable, and easy-to-assess biomarkers. METHODS Forty-six consecutive biopsy/cellblock samples and the corresponding resection specimens (as the gold standard for morphology and IHC) from 30 adenocarcinomas (AD), 10 squamous carcinomas (SQC), 5 adenosquamous carcinomas (ADSQC), and 1 sarcomatoid carcinoma (SC) were IHC-evaluated for p40 [corresponding to nontransactivating ΔNp63 isoforms] and thyroid transcription factor-1 (TTF1) by semiquantitative assessment. For p40, also immunodecoration intensity was taken into account and dichotomized as strong or low. RESULTS Nonrandom and overlapping distributions of the relevant markers were found in biopsy/cellblock and surgical specimens, which closely correlated with each other and the diverse tumor categories, with no differences in area under curve-receiver-operating-characteristic curves for each marker between any two samples, including p40 and p63. Diagnostic combinations were p40-/TTF1+ or TTF1- for AD (where p40 was negative, apart from 5/30 AD showing at the best 1-2% tumor cells with low intensity); p40+/TTF1- (p40 strong and by far higher than 50%) for SQC; and p40+/TTF1+ or p40+/TTF1- (p40 strong and less than 50%) for ADSQC. The single SC case was p40-/TTF1-, suggesting glandular lineage. Practically, 41/46 (89%) tumors were correctly classified by IHC on small samples, including 30 AD, 10 SQC, 1/5 ADSQC, and no SC. Underdiagnosis of ADSQC was actually because of sampling error of biopsies/cellblocks rather than insufficient biomarker robustness, whereas underdiagnosis of SC was really because of the failure of either marker to highlight epithelial-mesenchymal transition. CONCLUSIONS This minimalist IHC-based model of p40 and TTF1 on biopsy/cellblock samples was effective to correctly subtype most cases of lung cancer.
Collapse
|
42
|
Yi M, Meric-Bernstam F, Kuerer HM, Mittendorf EA, Bedrosian I, Lucci A, Hwang RF, Crow JR, Luo S, Hunt KK. Evaluation of a breast cancer nomogram for predicting risk of ipsilateral breast tumor recurrences in patients with ductal carcinoma in situ after local excision. J Clin Oncol 2012; 30:600-7. [PMID: 22253459 DOI: 10.1200/jco.2011.36.4976] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Prediction of patients at highest risk for ipsilateral breast tumor recurrence (IBTR) after local excision of ductal carcinoma in situ (DCIS) remains a clinical concern. The aim of our study was to evaluate a published nomogram from Memorial Sloan-Kettering Cancer Center to predict for risk of IBTR in patients with DCIS from our institution. PATIENTS AND METHODS We retrospectively identified 794 patients with a diagnosis of DCIS who had undergone local excision from 1990 through 2007 at the MD Anderson Cancer Center (MDACC). Clinicopathologic factors and the performance of the Memorial Sloan-Kettering Cancer Center nomogram for prediction of IBTR were assessed for 734 patients who had complete data. RESULTS There was a marked difference with respect to tumor grade, prevalence of necrosis, initial presentation, final margins, and receipt of endocrine therapy between the two cohorts. The biggest difference was that more patients received radiation in the MDACC cohort (75% at MDACC v 49% at MSKCC; P < .001). Follow-up time in the MDACC cohort was longer than in the MSKCC cohort (median 7.1 years v 5.6 years), and the recurrence rate was lower in the MDACC cohort (7.9% v 11%). The median 5-year probability of recurrence was 5%, and the median 10-year probability of recurrence was 7%. The nomogram for prediction of 5- and 10-year IBTR probabilities demonstrated imperfect calibration and discrimination, with a concordance index of 0.63. CONCLUSION Predictive models for IBTR in patients with DCIS who were treated with local excision are imperfect. Our current ability to accurately predict recurrence on the basis of clinical parameters alone is limited.
Collapse
Affiliation(s)
- Min Yi
- Department of Surgical Oncology, Unit 1484, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Costa J. Systems pathology: a critical review. Mol Oncol 2011; 6:27-32. [PMID: 22178234 DOI: 10.1016/j.molonc.2011.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 11/16/2011] [Accepted: 11/17/2011] [Indexed: 01/31/2023] Open
Abstract
The technological advances of the last twenty years together with the dramatic increase in computational power have injected new life into systems-level thinking in Medicine. This review emphasizes the close relationship of Systems Pathology to Systems Biology and delineates the differences between Systems Pathology and Clinical Systems Pathology. It also suggests an algorithm to support the application of systems-level thinking to clinical research, proposes applying systems-level thinking to the health care systems and forecasts an acceleration of preventive medicine as a result of the coupling of personal genomics with systems pathology.
Collapse
Affiliation(s)
- Jose Costa
- Yale University School of Medicine, New Haven, CT 06510, United States.
| |
Collapse
|
45
|
Rodriguez-Canales J, Eberle FC, Jaffe ES, Emmert-Buck MR. Why is it crucial to reintegrate pathology into cancer research? Bioessays 2011; 33:490-8. [PMID: 21590787 PMCID: PMC6377259 DOI: 10.1002/bies.201100017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The integration of pathology with molecular biology is vital if we are to enhance the translational value of cancer research. Pathology represents a bridge between medicine and basic biology, it remains the gold standard for cancer diagnosis, and it plays an important role in discovery studies. In the past, pathology and cancer research were closely associated; however, the molecular biology revolution has shifted the focus of investigators toward the molecular alterations of tumors. The reductionist approach taken in molecular studies is producing great insight into the inner workings of neoplasia, but it can also minimize the importance of histopathology and of understanding the disease as a whole. In turn, pathologists can underestimate the role of molecular studies in developing new ancillary techniques for clinical diagnosis. A multidisciplinary approach that integrates pathology and molecular biology within a translational research system is needed. This process will require overcoming cultural barriers and can be achieved through education, a more effective incorporation of pathology into biological research, and conversely an integration of biological research into the pathology laboratory.
Collapse
Affiliation(s)
- Jaime Rodriguez-Canales
- Laser Capture Microdissection Core and Pathogenetics Unit, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Franziska C. Eberle
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
- Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Elaine S. Jaffe
- Hematopathology Section, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michael R. Emmert-Buck
- Laser Capture Microdissection Core and Pathogenetics Unit, Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
46
|
Pelosi G, Rossi G, Bianchi F, Maisonneuve P, Galetta D, Sonzogni A, Veronesi G, Spaggiari L, Papotti M, Barbareschi M, Graziano P, Decensi A, Cavazza A, Viale G. Immunhistochemistry by Means of Widely Agreed-Upon Markers (Cytokeratins 5/6 and 7, p63, Thyroid Transcription Factor-1, and Vimentin) on Small Biopsies of Non-small Cell Lung Cancer Effectively Parallels the Corresponding Profiling and Eventual Diagnoses on Surgical Specimens. J Thorac Oncol 2011; 6:1039-49. [DOI: 10.1097/jto.0b013e318211dd16] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
47
|
Righi L, Graziano P, Fornari A, Rossi G, Barbareschi M, Cavazza A, Pelosi G, Scagliotti GV, Papotti M. Immunohistochemical subtyping of nonsmall cell lung cancer not otherwise specified in fine-needle aspiration cytology: a retrospective study of 103 cases with surgical correlation. Cancer 2011; 117:3416-23. [PMID: 21246522 DOI: 10.1002/cncr.25830] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 09/23/2010] [Accepted: 09/27/2010] [Indexed: 01/04/2023]
Abstract
BACKGROUND Histopathological subtyping of nonsmall cell lung cancer (NSCLC) is currently relevant in treatment decision because of a differential activity of specific therapeutic agents. Immunohistochemistry highlights cell differentiation lineages and, in this study, it was applied to maximize the proportion of accurately subtyped NSCLC not otherwise specified (NOS) on fine-needle aspiration cytology (FNAC) samples. METHODS Cell blocks from 103 FNAC samples with a morphological diagnosis of NSCLC-NOS were immunostained for cytokeratin (CK) 7, CK5, TTF1, and p63, whereas p40, napsin A (Naps-A), and desmocollin-3 (DSC-3) were only assessed in a subgroup of cases with discordant (CK7 and TTF1+ for nonsquamous, CK5 and p63+ for squamous) findings. Results were correlated with surgical specimens evaluated by morphology alone. RESULTS Thirty-seven (36%) tumors with CK7/TTF1+ and CK5/p63- corresponded to 35 cases of adenocarcinoma (ADC) and 2 cases of large cell carcinoma, whereas 9 (9%) cases with the reverse immunoprofile were squamous cell carcinoma (SQCC) at surgery (P < .001). Although the remaining 57 cases had different marker combinations, a correlation was found with ADC histology for TTF1+ samples (independent of other markers) and with SQCC for p63+/TTF1- immunophenotype (P < .001). p40 was never expressed in p63+ ADC, whereas Naps-A was restricted to ADC and DSC-3 to SQCC lineage. The percentage of unclassified NSCLC-NOS decreased from 36% to 14%. Combinations of 2 antibodies (TTF1/DSC-3 or p63/Naps-A) in the same section allowed diagnostic optimization in scant cytological samples. CONCLUSIONS [corrected] This 4-antibody panel approach may contribute to refine lung cancer classification in FNAC cell blocks, remarkably reducing the NSCLC-NOS diagnostic category.
Collapse
Affiliation(s)
- Luisella Righi
- Department of Clinical and Biological Sciences, University of Turin at San Luigi Hospital, Orbassano, Turin, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Pelosi G, Sonzogni A, Viale G. The Classification of Lung Carcinoma: Time to Change the Morphology-Based Approach? Int J Surg Pathol 2010; 18:161-72. [DOI: 10.1177/1066896910361736] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Morphology still remains the cornerstone in lung cancer classification and always has been accompanying pathologists in their daily activity, even though several ancillary techniques have been incorporated over time to improve diagnostic, prognostic, and predictive capabilities in lung cancer. Currently, we are also faced with a global rethinking of lung cancer care, especially once novel therapy strategies have been made available on the basis of the diverse characteristics of tumors. Although morphology still remains a not easily replaceable tool for lung cancer classification, we are now challenged by the need of offering clinicians more detailed subtyping of non-small-cell lung cancer especially in event of limited diagnostic material, poorly differentiated tumors, or unresectable lesions. Close integration of improved morphology, immunohistochemistry, and molecular tests will be able to not only sharpen our diagnostic algorithms and prognostic and predictive potentialities but also get insights into several lung cancer biology issues, such as histogenesis and new classification schemes.
Collapse
Affiliation(s)
- Giuseppe Pelosi
- European Institute of Oncology, Milan, Italy, , University of Milan School of Medicine, Milan, Italy
| | | | - Giuseppe Viale
- European Institute of Oncology, Milan, Italy, University of Milan School of Medicine, Milan, Italy
| |
Collapse
|
49
|
Khoury JD, Coffin CM, Spunt SL, Anderson JR, Meyer WH, Parham DM. Grading of nonrhabdomyosarcoma soft tissue sarcoma in children and adolescents: a comparison of parameters used for the Fédération Nationale des Centers de Lutte Contre le Cancer and Pediatric Oncology Group Systems. Cancer 2010; 116:2266-74. [PMID: 20166208 PMCID: PMC2987713 DOI: 10.1002/cncr.24929] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Two systems for grading soft tissue sarcoma are widely used currently: the National Cancer Institute (NCI) and the Fédération Nationale des Centers de Lutte Contre le Cancer (FNCLCC) systems. Both were developed using cohorts of predominantly adult patients. The Pediatric Oncology Group (POG) system, based on the NCI system, was adapted for grading pediatric nonrhabdomyosarcoma soft tissue sarcoma (NRSTS). The applicability and prognostic utility of the FNCLCC system in pediatric NRSTS has not been assessed or compared with the POG system. METHODS Tumors from 130 patients with malignant NRSTS enrolled on 3 completed multi-institutional clinical trials were assessed. Of 130 tumors, 102 (78%) were localized and 28 (22%) metastatic. Of the localized tumors, 55 of 102 (54%) were >5 cm. The estimated 5-year event-free survival (EFS) for the entire group was 47%. RESULTS As expected, stage and tumor sizes were predictive of EFS (P < .001). Both systems were predictive of 5-year EFS (POG, P = .0095 and FNCLCC, P = .0075). Patients whose tumors received discrepant grades (POG-G3 vs FNCLCC-G2/G1) (n = 44) had an intermediate outcome between those with concordant (G3 [n = 44] or G1/G2 [n = 42]) grades on both systems (P = .0018). By multivariate analysis, the mitotic index was predictive of EFS, using a cutoff of 10 mitotic figures per 10 high-power fields (P < .001). CONCLUSIONS In conclusion, both FNCLCC and POG systems provide an adequate prognostic measure of outcome for pediatric NRSTS; albeit, a sizeable subset of cases with apparently intermediate prognosis was graded differently by the 2 systems. The mitotic index appears to be a key parameter in grading pediatric NRSTS.
Collapse
Affiliation(s)
- Joseph D Khoury
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Would Virchow be a systems biologist? A discourse on the philosophy of science with implications for pathological research. Virchows Arch 2010; 456:599-607. [PMID: 20422212 DOI: 10.1007/s00428-010-0920-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 04/01/2010] [Indexed: 10/19/2022]
Abstract
Research in pathology spans from merely descriptive work to functional studies, "-omics" approaches and, more recently, systems biology. The work presented here aims at placing pathological research into an epistemological context. Aided by Rudolf Virchow, we give an overview on the philosophy of science including the Wiener Kreis, Popper, Kuhn, Fleck and Rheinberger and demonstrate their implications for routine diagnostics and science in pathology. A focus is on the fields of "-omics" and systems pathology.
Collapse
|