101
|
Burrows L, Sculthorpe D, Zhang H, Rehman O, Mukherjee A, Chen K. Mathematical modelling and deep learning algorithms to automate assessment of single and digitally multiplexed immunohistochemical stains in tumoural stroma. J Pathol Inform 2024; 15:100351. [PMID: 38186746 PMCID: PMC10770531 DOI: 10.1016/j.jpi.2023.100351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 01/09/2024] Open
Abstract
Whilst automated analysis of immunostains in pathology research has focused predominantly on the epithelial compartment, automated analysis of stains in the stromal compartment is challenging and therefore requires time-consuming pathological input and guidance to adjust to tissue morphometry as perceived by pathologists. This study aimed to develop a robust method to automate stromal stain analyses using 2 of the commonest stromal stains (SMA and desmin) employed in clinical pathology practice as examples. An effective computational method capable of automatically assessing and quantifying tumour-associated stromal stains was developed and applied on cores of colorectal cancer tissue microarrays. The methodology combines both mathematical models and deep learning techniques with the former requiring no training data and the latter as many inputs as possible. The novel mathematical model was used to produce a digital double marker overlay allowing for fast automated digital multiplex analysis of stromal stains. The results show that deep learning methodologies in combination with mathematical modelling allow for an accurate means of quantifying stromal stains whilst also opening up new possibilities of digital multiplex analyses.
Collapse
Affiliation(s)
- Liam Burrows
- Department of Mathematical Sciences and Centre for Mathematical Imaging Techniques, University of Liverpool, Liverpool, United Kingdom
| | - Declan Sculthorpe
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Hongrun Zhang
- Department of Eye and Vision Science, University of Liverpool, Liverpool, United Kingdom
| | - Obaid Rehman
- Department of Histopathology, Nottingham University Hospitals NHS, Nottingham, United Kingdom
| | - Abhik Mukherjee
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Department of Histopathology, Nottingham University Hospitals NHS, Nottingham, United Kingdom
| | - Ke Chen
- Department of Mathematical Sciences and Centre for Mathematical Imaging Techniques, University of Liverpool, Liverpool, United Kingdom
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
102
|
Chen Y, Du P, Zhang Y, Guo X, Song Y, Wang J, Yang LL, He W. Image-based multi-omics analysis for oral science: Recent progress and perspectives. J Dent 2024; 151:105425. [PMID: 39427959 DOI: 10.1016/j.jdent.2024.105425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVES The diagnosis and treatment of oral and dental diseases rely heavily on various types of medical imaging. Deep learning-mediated multi-omics analysis can extract more representative features than those identified through traditional diagnostic methods. This review aims to discuss the applications and recent advances in image-based multi-omics analysis in oral science and to highlight its potential to enhance traditional diagnostic approaches for oral diseases. STUDY SELECTION, DATA, AND SOURCES A systematic search was conducted in the PubMed, Web of Science, and Google Scholar databases, covering all available records. This search thoroughly examined and summarized advances in image-based multi-omics analysis in oral and maxillofacial medicine. CONCLUSIONS This review comprehensively summarizes recent advancements in image-based multi-omics analysis for oral science, including radiomics, pathomics, and photographic-based omics analysis. It also discusses the ongoing challenges and future perspectives that could provide new insights into exploiting the potential of image-based omics analysis in the field of oral science. CLINICAL SIGNIFICANCE This review article presents the state of image-based multi-omics analysis in stomatology, aiming to help oral clinicians recognize the utility of combining omics analyses with imaging during diagnosis and treatment, which can improve diagnostic accuracy, shorten times to diagnosis, save medical resources, and reduce disparity in professional knowledge among clinicians.
Collapse
Affiliation(s)
- Yizhuo Chen
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Pengxi Du
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yinyin Zhang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xin Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yujing Song
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jianhua Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Lei-Lei Yang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Wei He
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
103
|
Khosravi M, Jasemi SK, Hayati P, Javar HA, Izadi S, Izadi Z. Transformative artificial intelligence in gastric cancer: Advancements in diagnostic techniques. Comput Biol Med 2024; 183:109261. [PMID: 39488054 DOI: 10.1016/j.compbiomed.2024.109261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024]
Abstract
Gastric cancer represents a significant global health challenge with elevated incidence and mortality rates, highlighting the need for advancements in diagnostic and therapeutic strategies. This review paper addresses the critical need for a thorough synthesis of the role of artificial intelligence (AI) in the management of gastric cancer. It provides an in-depth analysis of current AI applications, focusing on their contributions to early diagnosis, treatment planning, and outcome prediction. The review identifies key gaps and limitations in the existing literature by examining recent studies and technological developments. It aims to clarify the evolution of AI-driven methods and their impact on enhancing diagnostic accuracy, personalizing treatment strategies, and improving patient outcomes. The paper emphasizes the transformative potential of AI in overcoming the challenges associated with gastric cancer management and proposes future research directions to further harness AI's capabilities. Through this synthesis, the review underscores the importance of integrating AI technologies into clinical practice to revolutionize gastric cancer management.
Collapse
Affiliation(s)
- Mobina Khosravi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Seyedeh Kimia Jasemi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Parsa Hayati
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Hamid Akbari Javar
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Saadat Izadi
- Department of Computer Engineering and Information Technology, Razi University, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
104
|
Lee S, Arffman RK, Komsi EK, Lindgren O, Kemppainen J, Kask K, Saare M, Salumets A, Piltonen TT. Dynamic changes in AI-based analysis of endometrial cellular composition: Analysis of PCOS and RIF endometrium. J Pathol Inform 2024; 15:100364. [PMID: 38445292 PMCID: PMC10914580 DOI: 10.1016/j.jpi.2024.100364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/07/2024] Open
Abstract
Background The human endometrium undergoes a monthly cycle of tissue growth and degeneration. During the mid-secretory phase, the endometrium establishes an optimal niche for embryo implantation by regulating cellular composition (e.g., epithelial and stromal cells) and differentiation. Impaired endometrial development observed in conditions such as polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF) contributes to infertility. Surprisingly, despite the importance of the endometrial lining properly developing prior to pregnancy, precise measures of endometrial cellular composition in these two infertility-associated conditions are entirely lacking. Additionally, current methods for measuring the epithelial and stromal area have limitations, including intra- and inter-observer variability and efficiency. Methods We utilized a deep-learning artificial intelligence (AI) model, created on a cloud-based platform and developed in our previous study. The AI model underwent training to segment both areas populated by epithelial and stromal endometrial cells. During the training step, a total of 28.36 mm2 areas were annotated, comprising 2.56 mm2 of epithelium and 24.87 mm2 of stroma. Two experienced pathologists validated the performance of the AI model. 73 endometrial samples from healthy control women were included in the sample set to establish cycle phase-dependent dynamics of the endometrial epithelial-to-stroma ratio from the proliferative (PE) to secretory (SE) phases. In addition, 91 samples from PCOS cases, accounting for the presence or absence of ovulation and representing all menstrual cycle phases, and 29 samples from RIF patients on day 5 after progesterone administration in the hormone replacement treatment cycle were also included and analyzed in terms of cellular composition. Results Our AI model exhibited reliable and reproducible performance in delineating epithelial and stromal compartments, achieving an accuracy of 92.40% and 99.23%, respectively. Moreover, the performance of the AI model was comparable to the pathologists' assessment, with F1 scores exceeding 82% for the epithelium and >96% for the stroma. Next, we compared the endometrial epithelial-to-stromal ratio during the menstrual cycle in women with PCOS and in relation to endometrial receptivity status in RIF patients. The ovulatory PCOS endometrium exhibited epithelial cell proportions similar to those of control and healthy women's samples in every cycle phase, from the PE to the late SE, correlating with progesterone levels (control SE, r2 = 0.64, FDR < 0.001; PCOS SE, r2 = 0.52, FDR < 0.001). The mid-SE endometrium showed the highest epithelial percentage compared to both the early and late SE endometrium in both healthy women and PCOS patients. Anovulatory PCOS cases showed epithelial cellular fractions comparable to those of PCOS cases in the PE (Anovulatory, 14.54%; PCOS PE, 15.56%, p = 1.00). We did not observe significant differences in the epithelial-to-stroma ratio in the hormone-induced endometrium in RIF patients with different receptivity statuses. Conclusion The AI model rapidly and accurately identifies endometrial histology features by calculating areas occupied by epithelial and stromal cells. The AI model demonstrates changes in epithelial cellular proportions according to the menstrual cycle phase and reveals no changes in epithelial cellular proportions based on PCOS and RIF conditions. In conclusion, the AI model can potentially improve endometrial histology assessment by accelerating the analysis of the cellular composition of the tissue and by ensuring maximal objectivity for research and clinical purposes.
Collapse
Affiliation(s)
- Seungbaek Lee
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Riikka K. Arffman
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| | - Elina K. Komsi
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| | - Outi Lindgren
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Janette Kemppainen
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Keiu Kask
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm 14152, Sweden
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynaecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu 90220, Finland
| |
Collapse
|
105
|
Lee S, Arffman RK, Komsi EK, Lindgren O, Kemppainen JA, Metsola H, Rossi HR, Ahtikoski A, Kask K, Saare M, Salumets A, Piltonen TT. AI-algorithm training and validation for identification of endometrial CD138+ cells in infertility-associated conditions; polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF). J Pathol Inform 2024; 15:100380. [PMID: 38827567 PMCID: PMC11140811 DOI: 10.1016/j.jpi.2024.100380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/20/2024] [Accepted: 04/26/2024] [Indexed: 06/04/2024] Open
Abstract
Background Endometrial CD138+ plasma cells serve as a diagnostic biomarker for endometrial inflammation, and their elevated occurrence correlates positively with adverse pregnancy outcomes. Infertility-related conditions like polycystic ovary syndrome (PCOS) and recurrent implantation failure (RIF) are closely associated with systemic and local chronic inflammatory status, wherein endometrial CD138+ plasma cell accumulation could also contribute to endometrial pathology. Current methods for quantifying CD138+ cells typically involve laborious and time-consuming microscopic assessments of only a few random areas from a slide. These methods have limitations in accurately representing the entire slide and are susceptible to significant biases arising from intra- and interobserver variations. Implementing artificial intelligence (AI) for CD138+ cell identification could enhance the accuracy, reproducibility, and reliability of analysis. Methods Here, an AI algorithm was developed to identify CD138+ plasma cells within endometrial tissue. The AI model comprised two layers of convolutional neural networks (CNNs). CNN1 was trained to segment epithelium and stroma across 28,363 mm2 (2.56 mm2 of epithelium and 24.87 mm2 of stroma), while CNN2 was trained to distinguish stromal cells based on CD138 staining, encompassing 7345 cells in the object layers (6942 CD138- cells and 403 CD138+ cells). The training and performance of the AI model were validated by three experienced pathologists. We collected 193 endometrial tissues from healthy controls (n = 73), women with PCOS (n = 91), and RIF patients (n = 29) and compared the CD138+ cell percentages based on cycle phases, ovulation status, and endometrial receptivity utilizing the AI model. Results The AI algorithm consistently and reliably distinguished CD138- and CD138+ cells, with total error rates of 6.32% and 3.23%, respectively. During the training validation, there was a complete agreement between the decisions made by the pathologists and the AI algorithm, while the performance validation demonstrated excellent accuracy between the AI and human evaluation methods (intraclass correlation; 0.76, 95% confidence intervals; 0.36-0.93, p = 0.002) and a positive correlation (Spearman's rank correlation coefficient: 0.79, p < 0.01). In the AI analysis, the AI model revealed higher CD138+ cell percentages in the proliferative phase (PE) endometrium compared to the secretory phase or anovulatory PCOS endometrium, irrespective of PCOS diagnosis. Interestingly, CD138+ percentages differed according to PCOS phenotype in the PE (p = 0.03). On the other hand, the receptivity status had no impact on the cell percentages in RIF samples. Conclusion Our findings emphasize the potential and accuracy of the AI algorithm in detecting endometrial CD138+ plasma cells, offering distinct advantages over manual inspection, such as rapid analysis of whole slide images, reduction of intra- and interobserver variations, sparing the valuable time of trained specialists, and consistent productivity. This supports the application of AI technology to help clinical decision-making, for example, in understanding endometrial cycle phase-related dynamics, as well as different reproductive disorders.
Collapse
Affiliation(s)
- Seungbaek Lee
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
| | - Riikka K. Arffman
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Elina K. Komsi
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Outi Lindgren
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Janette A. Kemppainen
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Hanna Metsola
- Department of Pathology, Oulu University Hospital, Cancer and Translational Medicine Research Unit, University of Oulu, Oulu 90220, Finland
| | - Henna-Riikka Rossi
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| | - Anne Ahtikoski
- Department of Pathology, Turku University Hospital, Turku 20521, Finland
| | - Keiu Kask
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Merli Saare
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
| | - Andres Salumets
- Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu 50406, Estonia
- Competence Centre on Health Technologies, Tartu 51014, Estonia
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm 14152, Sweden
| | - Terhi T. Piltonen
- Department of Obstetrics and Gynaecology, Medical Research Center Oulu, Research Unit of Clinical Medicine, University of Oulu and Oulu University Hospital, Oulu 90220, Finland
| |
Collapse
|
106
|
Wang YL, Gao S, Xiao Q, Li C, Grzegorzek M, Zhang YY, Li XH, Kang Y, Liu FH, Huang DH, Gong TT, Wu QJ. Role of artificial intelligence in digital pathology for gynecological cancers. Comput Struct Biotechnol J 2024; 24:205-212. [PMID: 38510535 PMCID: PMC10951449 DOI: 10.1016/j.csbj.2024.03.007] [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: 12/28/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 03/22/2024] Open
Abstract
The diagnosis of cancer is typically based on histopathological sections or biopsies on glass slides. Artificial intelligence (AI) approaches have greatly enhanced our ability to extract quantitative information from digital histopathology images as a rapid growth in oncology data. Gynecological cancers are major diseases affecting women's health worldwide. They are characterized by high mortality and poor prognosis, underscoring the critical importance of early detection, treatment, and identification of prognostic factors. This review highlights the various clinical applications of AI in gynecological cancers using digitized histopathology slides. Particularly, deep learning models have shown promise in accurately diagnosing, classifying histopathological subtypes, and predicting treatment response and prognosis. Furthermore, the integration with transcriptomics, proteomics, and other multi-omics techniques can provide valuable insights into the molecular features of diseases. Despite the considerable potential of AI, substantial challenges remain. Further improvements in data acquisition and model optimization are required, and the exploration of broader clinical applications, such as the biomarker discovery, need to be explored.
Collapse
Affiliation(s)
- Ya-Li Wang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Information Center, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Song Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qian Xiao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chen Li
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Marcin Grzegorzek
- Institute for Medical Informatics, University of Luebeck, Luebeck, Germany
| | - Ying-Ying Zhang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiao-Han Li
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ye Kang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dong-Hui Huang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Shenyang, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China
| |
Collapse
|
107
|
Ghete T, Kock F, Pontones M, Pfrang D, Westphal M, Höfener H, Metzler M. Models for the marrow: A comprehensive review of AI-based cell classification methods and malignancy detection in bone marrow aspirate smears. Hemasphere 2024; 8:e70048. [PMID: 39629240 PMCID: PMC11612571 DOI: 10.1002/hem3.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Accepted: 10/26/2024] [Indexed: 12/07/2024] Open
Abstract
Given the high prevalence of artificial intelligence (AI) research in medicine, the development of deep learning (DL) algorithms based on image recognition, such as the analysis of bone marrow aspirate (BMA) smears, is rapidly increasing in the field of hematology and oncology. The models are trained to identify the optimal regions of the BMA smear for differential cell count and subsequently detect and classify a number of cell types, which can ultimately be utilized for diagnostic purposes. Moreover, AI is capable of identifying genetic mutations phenotypically. This pipeline has the potential to offer an accurate and rapid preliminary analysis of the bone marrow in the clinical routine. However, the intrinsic complexity of hematological diseases presents several challenges for the automatic morphological assessment. To ensure general applicability across multiple medical centers and to deliver high accuracy on prospective clinical data, AI models would require highly heterogeneous training datasets. This review presents a systematic analysis of models for cell classification and detection of hematological malignancies published in the last 5 years (2019-2024). It provides insight into the challenges and opportunities of these DL-assisted tasks.
Collapse
Affiliation(s)
- Tabita Ghete
- Department of Pediatrics and Adolescent MedicineUniversity Hospital ErlangenErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Farina Kock
- Computational PathologyFraunhofer Institute for Digital Medicine (MEVIS)BremenGermany
| | - Martina Pontones
- Department of Pediatrics and Adolescent MedicineUniversity Hospital ErlangenErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - David Pfrang
- Computational PathologyFraunhofer Institute for Digital Medicine (MEVIS)BremenGermany
| | - Max Westphal
- Computational PathologyFraunhofer Institute for Digital Medicine (MEVIS)BremenGermany
| | - Henning Höfener
- Computational PathologyFraunhofer Institute for Digital Medicine (MEVIS)BremenGermany
| | - Markus Metzler
- Department of Pediatrics and Adolescent MedicineUniversity Hospital ErlangenErlangenGermany
- Bavarian Cancer Research Center (BZKF)ErlangenGermany
- Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)ErlangenGermany
| |
Collapse
|
108
|
Brevet M, Li Z, Parwani A. Computational pathology in the identification of HER2-low breast cancer: Opportunities and challenges. J Pathol Inform 2024; 15:100343. [PMID: 38125925 PMCID: PMC10730362 DOI: 10.1016/j.jpi.2023.100343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/18/2023] [Accepted: 11/01/2023] [Indexed: 12/23/2023] Open
Abstract
For the past 2 decades, pathologists have been accustomed to reporting the HER2 status of breast cancer as either positive or negative, based on HER2 IHC. Today, however, there is a clinical imperative to employ a 3-tier approach to interpreting HER2 IHC that can also identify tumours categorised as HER2-low. Meeting this need for a finer degree of discrimination may be challenging, and in this article, we consider the potential for the integration of computational approaches to support pathologists in achieving accurate and reproducible HER2 IHC scoring as well as outlining some of the practicalities involved.
Collapse
Affiliation(s)
| | - Zaibo Li
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Anil Parwani
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
109
|
Buja LM. Pathobiology of myocardial and cardiomyocyte injury in ischemic heart disease: Perspective from seventy years of cell injury research. Exp Mol Pathol 2024; 140:104944. [PMID: 39577392 DOI: 10.1016/j.yexmp.2024.104944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/11/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
This review presents a perspective on the pathobiology of acute myocardial infarction, a major manifestation of ischemic heart disease, and related mechanisms of ischemic and toxic cardiomyocyte injury, based on advances and insights that have accrued over the last seventy years, including my sixty years of involvement in the field as a physician-scientist-pathologist. This analysis is based on integration of my research within the broader context of research in the field. A particular focus has been on direct measurements in cardiomyocytes of electrolyte content by electron probe X-ray microanalysis (EPXMA) and Ca2+ fluxes by fura-2 microspectrofluorometry. These studies established that increased intracellular Ca2+ develops at a transitional stage in the progression of cardiomyocyte injury in association with ATP depletion, other electrolyte alterations, altered cell volume regulation, and altered membrane phospholipid composition. Subsequent increase in total calcium with mitochondrial calcium accumulation can occur. These alterations are characteristic of oncosis, which is an initial pre-lethal state of cell injury with cell swelling due to cell membrane dysfunction in ATP depleted cells; oncosis rapidly progresses to necrosis/necroptosis with physical disruption of the cell membrane, unless the adverse stimulus is rapidly reversed. The observed sequential changes fit a three-stage model of membrane injury leading to irreversible cell injury. The data establish oncosis as the primary mode of cardiomyocyte injury in evolving myocardial infarcts. Oncosis also has been documented to be the typical form of non-ischemic cell injury due to toxins. Cardiomyocytes with less energy impairment have the capability of undergoing apoptosis and autophagic death as well as oncosis, as is seen in pathological remodeling in chronic heart failure. Work is ongoing to apply the insights from experimental studies to better understand and ameliorate myocardial ischemia and reperfusion injury in patients. The perspective and insights in this review are derived from basic principles of pathology, an integrative discipline focused on mechanisms of disease affecting the cell, the organizing unit of living organisms.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, TX, United States of America.
| |
Collapse
|
110
|
Santa-Rosario JC, Gustafson EA, Sanabria Bellassai DE, Gustafson PE, de Socarraz M. Validation and three years of clinical experience in using an artificial intelligence algorithm as a second read system for prostate cancer diagnosis-real-world experience. J Pathol Inform 2024; 15:100378. [PMID: 38868487 PMCID: PMC11166872 DOI: 10.1016/j.jpi.2024.100378] [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: 03/22/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/14/2024] Open
Abstract
Background Prostate cancer ranks as the most frequently diagnosed cancer in men in the USA, with significant mortality rates. Early detection is pivotal for optimal patient outcomes, providing increased treatment options and potentially less invasive interventions. There remain significant challenges in prostate cancer histopathology, including the potential for missed diagnoses due to pathologist variability and subjective interpretations. Methods To address these challenges, this study investigates the ability of artificial intelligence (AI) to enhance diagnostic accuracy. The Galen™ Prostate AI algorithm was validated on a cohort of Puerto Rican men to demonstrate its efficacy in cancer detection and Gleason grading. Subsequently, the AI algorithm was integrated into routine clinical practice during a 3-year period at a CLIA certified precision pathology laboratory. Results The Galen™ Prostate AI algorithm showed a 96.7% (95% CI 95.6-97.8) specificity and a 96.6% (95% CI 93.3-98.8) sensitivity for prostate cancer detection and 82.1% specificity (95% CI 73.9-88.5) and 81.1% sensitivity (95% CI 73.7-87.2) for distinction of Gleason Grade Group 1 from Grade Group 2+. The subsequent AI integration into routine clinical use examined prostate cancer diagnoses on >122,000 slides and 9200 cases over 3 years and had an overall AI Impact ™ factor of 1.8%. Conclusions The potential of AI to be a powerful, reliable, and effective diagnostic tool for pathologists is highlighted, while the AI Impact™ in a real-world setting demonstrates the ability of AI to standardize prostate cancer diagnosis at a high level of performance across pathologists.
Collapse
Affiliation(s)
- Juan Carlos Santa-Rosario
- CorePlus Servicios Clínicos y Patológicos; Plazoleta la Cerámica, Suite 2-6 Ave. Sánchez Vilella, Esq, PR-190, Carolina, PR 00983, USA
| | - Erik A. Gustafson
- CorePlus Servicios Clínicos y Patológicos; Plazoleta la Cerámica, Suite 2-6 Ave. Sánchez Vilella, Esq, PR-190, Carolina, PR 00983, USA
| | - Dario E. Sanabria Bellassai
- CorePlus Servicios Clínicos y Patológicos; Plazoleta la Cerámica, Suite 2-6 Ave. Sánchez Vilella, Esq, PR-190, Carolina, PR 00983, USA
| | - Phillip E. Gustafson
- CorePlus Servicios Clínicos y Patológicos; Plazoleta la Cerámica, Suite 2-6 Ave. Sánchez Vilella, Esq, PR-190, Carolina, PR 00983, USA
| | - Mariano de Socarraz
- CorePlus Servicios Clínicos y Patológicos; Plazoleta la Cerámica, Suite 2-6 Ave. Sánchez Vilella, Esq, PR-190, Carolina, PR 00983, USA
| |
Collapse
|
111
|
Deng B, Tian Y, Zhang Q, Wang Y, Chai Z, Ye Q, Yao S, Liang T, Li J. NecroGlobalGCN: Integrating micronecrosis information in HCC prognosis prediction via graph convolutional neural networks. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 257:108435. [PMID: 39357091 DOI: 10.1016/j.cmpb.2024.108435] [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: 06/05/2024] [Revised: 09/13/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND AND OBJECTIVE Hepatocellular carcinoma (HCC) ranks fourth in cancer mortality, underscoring the importance of accurate prognostic predictions to improve postoperative survival rates in patients. Although micronecrosis has been shown to have high prognostic value in HCC, its application in clinical prognosis prediction requires specialized knowledge and complex calculations, which poses challenges for clinicians. It would be of interest to develop a model to help clinicians make full use of micronecrosis to assess patient survival. METHODS To address these challenges, we propose a HCC prognosis prediction model that integrates pathological micronecrosis information through Graph Convolutional Neural Networks (GCN). This approach enables GCN to utilize micronecrosis, which has been shown to be highly correlated with prognosis, thereby significantly enhancing prognostic stratification quality. We developed our model using 3622 slides from 752 patients with primary HCC from the FAH-ZJUMS dataset and conducted internal and external validations on the FAH-ZJUMS and TCGA-LIHC datasets, respectively. RESULTS Our method outperformed the baseline by 8.18% in internal validation and 9.02% in external validations. Overall, this paper presents a deep learning research paradigm that integrates HCC micronecrosis, enhancing both the accuracy and interpretability of prognostic predictions, with potential applicability to other pathological prognostic markers. CONCLUSIONS This study proposes a composite GCN prognostic model that integrates information on HCC micronecrosis, collecting large dataset of HCC histopathological images. This approach could assist clinicians in analyzing HCC patient survival and precisely locating and visualizing necrotic tissues that affect prognosis. Following the research paradigm outlined in this paper, other prognostic biomarker integration models with GCN could be developed, significantly enhancing the predictive performance and interpretability of prognostic model.
Collapse
Affiliation(s)
- Boyang Deng
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, No. 38 Zheda Road, Hangzhou 310027, China
| | - Yu Tian
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, No. 38 Zheda Road, Hangzhou 310027, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, and also with Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Yangyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenxin Chai
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, No. 38 Zheda Road, Hangzhou 310027, China
| | - Qiancheng Ye
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, No. 38 Zheda Road, Hangzhou 310027, China
| | - Shang Yao
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, No. 38 Zheda Road, Hangzhou 310027, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang University Cancer Center, and also with Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
| | - Jingsong Li
- Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, No. 38 Zheda Road, Hangzhou 310027, China; Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou 311100, China.
| |
Collapse
|
112
|
Hosseini MS, Bejnordi BE, Trinh VQH, Chan L, Hasan D, Li X, Yang S, Kim T, Zhang H, Wu T, Chinniah K, Maghsoudlou S, Zhang R, Zhu J, Khaki S, Buin A, Chaji F, Salehi A, Nguyen BN, Samaras D, Plataniotis KN. Computational pathology: A survey review and the way forward. J Pathol Inform 2024; 15:100357. [PMID: 38420608 PMCID: PMC10900832 DOI: 10.1016/j.jpi.2023.100357] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 03/02/2024] Open
Abstract
Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field's future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to GitHub. Updated version of this draft can also be found from arXiv.
Collapse
Affiliation(s)
- Mahdi S. Hosseini
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | | | - Vincent Quoc-Huy Trinh
- Institute for Research in Immunology and Cancer of the University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Lyndon Chan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Danial Hasan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Xingwen Li
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Stephen Yang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Taehyo Kim
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Haochen Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Theodore Wu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Kajanan Chinniah
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Sina Maghsoudlou
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ryan Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jiadai Zhu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Samir Khaki
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Andrei Buin
- Huron Digitial Pathology, St. Jacobs, ON N0B 2N0, Canada
| | - Fatemeh Chaji
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ala Salehi
- Department of Electrical and Computer Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Bich Ngoc Nguyen
- University of Montreal Hospital Center, Montreal, QC H2X 0C2, Canada
| | - Dimitris Samaras
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11794, United States
| | - Konstantinos N. Plataniotis
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| |
Collapse
|
113
|
Lobanova OA, Kolesnikova AO, Ponomareva VA, Vekhova KA, Shaginyan AL, Semenova AB, Nekhoroshkov DP, Kochetkova SE, Kretova NV, Zanozin AS, Peshkova MA, Serezhnikova NB, Zharkov NV, Kogan EA, Biryukov AA, Rudenko EE, Demura TA. Artificial intelligence (AI) for tumor microenvironment (TME) and tumor budding (TB) identification in colorectal cancer (CRC) patients: A systematic review. J Pathol Inform 2024; 15:100353. [PMID: 39712977 PMCID: PMC11662261 DOI: 10.1016/j.jpi.2023.100353] [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: 07/03/2023] [Revised: 09/19/2023] [Accepted: 11/16/2023] [Indexed: 12/24/2024] Open
Abstract
Evaluation of the parameters such as tumor microenvironment (TME) and tumor budding (TB) is one of the most important steps in colorectal cancer (CRC) diagnosis and cancer development prognosis. In recent years, artificial intelligence (AI) has been successfully used to solve such problems. In this paper, we summarize the latest data on the use of artificial intelligence to predict tumor microenvironment and tumor budding in histological scans of patients with colorectal cancer. We performed a systematic literature search using 2 databases (Medline and Scopus) with the following search terms: ("tumor microenvironment" OR "tumor budding") AND ("colorectal cancer" OR CRC) AND ("artificial intelligence" OR "machine learning " OR "deep learning"). During the analysis, we gathered from the articles performance scores such as sensitivity, specificity, and accuracy of identifying TME and TB using artificial intelligence. The systematic review showed that machine learning and deep learning successfully cope with the prediction of these parameters. The highest accuracy values in TB and TME prediction were 97.7% and 97.3%, respectively. This review led us to the conclusion that AI platforms can already be used as diagnostic aids, which will greatly facilitate the work of pathologists in detection and estimation of TB and TME as instruments and second-opinion services. A key limitation in writing this systematic review was the heterogeneous use of performance metrics for machine learning models by different authors, as well as relatively small datasets used in some studies.
Collapse
Affiliation(s)
- Olga Andreevna Lobanova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Anastasia Olegovna Kolesnikova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | | | - Ksenia Andreevna Vekhova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Anaida Lusparonovna Shaginyan
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Alisa Borisovna Semenova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | | | - Svetlana Evgenievna Kochetkova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Natalia Valeryevna Kretova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Alexander Sergeevich Zanozin
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Maria Alekseevna Peshkova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Natalia Borisovna Serezhnikova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Nikolay Vladimirovich Zharkov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Evgeniya Altarovna Kogan
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Alexander Alekseevich Biryukov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Ekaterina Evgenievna Rudenko
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| | - Tatiana Alexandrovna Demura
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Trubetskaya street, 8, p. 2, 119991 Moscow, Russia
| |
Collapse
|
114
|
Vidal JM, Tsiknakis N, Staaf J, Bosch A, Ehinger A, Nimeus E, Salgado R, Bai Y, Rimm DL, Hartman J, Acs B. The analytical and clinical validity of AI algorithms to score TILs in TNBC: can we use different machine learning models interchangeably? EClinicalMedicine 2024; 78:102928. [PMID: 39634035 PMCID: PMC11615110 DOI: 10.1016/j.eclinm.2024.102928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/24/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
Background Pathologist-read tumor-infiltrating lymphocytes (TILs) have showcased their predictive and prognostic potential for early and metastatic triple-negative breast cancer (TNBC) but it is still subject to variability. Artificial intelligence (AI) is a promising approach toward eliminating variability and objectively automating TILs assessment. However, demonstrating robust analytical and prognostic validity is the key challenge currently preventing their integration into clinical workflows. Methods We evaluated the impact of ten AI models on TILs scoring, emphasizing their distinctions in TILs analytical and prognostic validity. Several AI-based TILs scoring models (seven developed and three previously validated AI models) were tested in a retrospective analytical cohort and in an independent prospective cohort to compare prognostic validation against invasive disease-free survival endpoint with 4 years median follow-up. The development and analytical validity set consisted of diagnostic tissue slides of 79 women with surgically resected primary invasive TNBC tumors diagnosed between 2012 and 2016 from the Yale School of Medicine. An independent set comprising of 215 TNBC patients from Sweden diagnosed between 2010 and 2015, was used for testing prognostic validity. Findings A significant difference in analytical validity (Spearman's r = 0.63-0.73, p < 0.001) is highlighted across AI methodologies and training strategies. Interestingly, the prognostic performance of digital TILs is demonstrated for eight out of ten AI models, even less extensively trained ones, with similar and overlapping hazard ratios (HR) in the external validation cohort (Cox regression analysis based on IDFS-endpoint, HR = 0.40-0.47; p < 0.004). Interpretation The demonstrated prognostic validity for most of the AI TIL models can be attributed to the intrinsic robustness of host anti-tumor immunity (measured by TILs) as a biomarker. However, the discrepancies between AI models should not be overlooked; rather, we believe that there is a critical need for an accessible, large, multi-centric dataset that will serve as a benchmark ensuring the comparability and reliability of different AI tools in clinical implementation. Funding Nikos Tsiknakis is supported by the Swedish Research Council (Grant Number 2021-03061, Theodoros Foukakis). Balazs Acs is supported by The Swedish Society for Medical Research (Svenska Sällskapet för Medicinsk Forskning) postdoctoral grant. Roberto Salgado is supported by a grant from Breast Cancer Research Foundation (BCRF).
Collapse
Affiliation(s)
- Joan Martínez Vidal
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Nikos Tsiknakis
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Ana Bosch
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Region Skåne, Lund, Sweden
| | - Anna Ehinger
- Department of Genetics, Pathology and Molecular Diagnostics, Laboratory Medicine, Region Skåne, Lund, Sweden
| | - Emma Nimeus
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
- Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Yalai Bai
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - David L. Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Internal Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Balazs Acs
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
115
|
Kim H, Kim J, Yeon SY, You S. Machine learning approaches for spatial omics data analysis in digital pathology: tools and applications in genitourinary oncology. Front Oncol 2024; 14:1465098. [PMID: 39678498 PMCID: PMC11638011 DOI: 10.3389/fonc.2024.1465098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Recent advances in spatial omics technologies have enabled new approaches for analyzing tissue morphology, cell composition, and biomolecule expression patterns in situ. These advances are promoting the development of new computational tools and quantitative techniques in the emerging field of digital pathology. In this review, we survey current trends in the development of computational methods for spatially mapped omics data analysis using digitized histopathology slides and supplementary materials, with an emphasis on tools and applications relevant to genitourinary oncological research. The review contains three sections: 1) an overview of image processing approaches for histopathology slide analysis; 2) machine learning integration with spatially resolved omics data analysis; 3) a discussion of current limitations and future directions for integration of machine learning in the clinical decision-making process.
Collapse
Affiliation(s)
- Hojung Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Jina Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Su Yeon Yeon
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
116
|
Zheng J, Wang T, Wang H, Yan B, Lai J, Qiu K, Zhou X, Tan J, Wang S, Ji H, Feng M, Jiang W, Wang H, Yan J. Use of a Pathomics Nomogram to Predict Postoperative Liver Metastasis in Patients with Stage III Colorectal Cancer. Ann Surg Oncol 2024:10.1245/s10434-024-16519-8. [PMID: 39614006 DOI: 10.1245/s10434-024-16519-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/30/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Approximately 25% of patients with stage III colorectal cancer experience liver metastasis after radical resection; however, there is currently a lack of methods to predict liver metastasis. This study aims to develop and validate a pathomics nomogram to predict liver metastasis in patients with stage III colorectal cancer. METHODS A total of 318 enrolled patients were divided into three cohorts: a training cohort (n = 139), a validation cohort (n = 69), and an external cohort (n = 110). A competitive risk nomogram was established by the pathomics signature and clinicopathological characteristics and assessed by calibration, discrimination, and clinical usefulness. RESULTS A significant correlation between the pathomics signature and liver metastasis in stage III colorectal cancer was found. Multivariate Fine-Gray analysis indicated that preoperative carcinoembryonic antigen level, postoperative chemotherapy, and pathomics signature were independent predictors of liver metastasis. A competitive risk nomogram was developed to predict liver metastasis in patients with stage III colorectal cancer. The predicting nomogram shows good discrimination and calibration, with C-indexes of 0.811 (95% confidence interval [CI] 0.651-0.971), 0.759 (95% CI 0.531-0.987), and 0.845 (95% CI 0.641-0.999), with area under the receiver operating characteristic (AUROC) curves at 5 years of 0.833 (95% CI 0.742-0.925), 0.760 (95% CI 0.652-0.893), and 0.812 (95% CI 0.692-0.931) in the training, validation, and external cohorts, respectively. Compared with the clinicopathological nomogram, the nomogram combined with the pathomics signature had better performance (AUROC 0.823 [95% CI 0.764-0.881] vs. 0.678 [95% CI 0.606-0.751]; p < 0.001). CONCLUSIONS The pathomics signature is a predictive indicator for liver metastasis in patients with stage III colorectal cancer, and the integrated nomogram can be used to predict liver metastasis better than the clinicopathological nomogram alone.
Collapse
Affiliation(s)
- Jixiang Zheng
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ting Wang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Huaiming Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China
| | - Botao Yan
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jianbo Lai
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Kemao Qiu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xinyi Zhou
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jie Tan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shijie Wang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hongli Ji
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Mingyuan Feng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Wei Jiang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China.
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Hui Wang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, People's Republic of China.
| | - Jun Yan
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China.
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
117
|
Zhang Y, Xue Y, Gao Y, Zhang Y. Prognostic and predictive value of pathohistological features in gastric cancer and identification of SLITRK4 as a potential biomarker for gastric cancer. Sci Rep 2024; 14:29241. [PMID: 39587240 PMCID: PMC11589652 DOI: 10.1038/s41598-024-80292-7] [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: 09/07/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024] Open
Abstract
The aim of this study was to develop a quantitative feature-based model from histopathologic images to assess the prognosis of patients with gastric cancer. Whole slide image (WSI) images of H&E-stained histologic specimens of gastric cancer patients from The Cancer Genome Atlas were included and randomly assigned to training and test groups in a 7:3 ratio. A systematic preprocessing approach was employed as well as a non-overlapping segmentation method that combined patch-level prediction with a multi-instance learning approach to integrate features across the slide images. Subjects were categorized into high- or low-risk groups based on the median risk score derived from the model, and the significance of this stratification was assessed using a log-rank test. In addition, combining transcriptomic data from patients and data from other large cohort studies, we further searched for genes associated with pathological features and their prognostic value. A total of 165 gastric cancer patients were included for model training, and a total of 26 features were integrated through multi-instance learning, with each process generating 11 probabilistic features and 2 predictive labeling features. We applied a 10-fold Lasso-Cox regression model to achieve dimensionality reduction of these features. The predictive accuracy of the model was verified using Kaplan-Meyer (KM) curves for stratification with a consistency index of 0.741 for the training set and 0.585 for the test set. Deep learning-based resultant supervised pathohistological features have the potential for superior prognostic stratification of gastric cancer patients, transforming image pixels into an effective and labor-saving tool to optimize the clinical management of gastric cancer patients. Also, SLITRK4 was identified as a prognostic marker for gastric cancer.
Collapse
Affiliation(s)
- Yuzhe Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China
| | - Yuhang Xue
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Yongju Gao
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Hospital of China Medical University, NO.155, North Nanjing Street, Heping District, Shenyang City, 110001, China.
| |
Collapse
|
118
|
Zheng J, Wang T, Wang S, Ji H, Yan J. ASO Author Reflections: Use of a Pathomics Nomogram to Predict Postoperative Liver Metastasis in Patients with Stage III Colorectal Cancer. Ann Surg Oncol 2024:10.1245/s10434-024-16576-z. [PMID: 39565491 DOI: 10.1245/s10434-024-16576-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024]
Affiliation(s)
- Jixiang Zheng
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Ting Wang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Shijie Wang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Hongli Ji
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jun Yan
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China.
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Cancer, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
119
|
AlDoughaim M, AlSuhebany N, AlZahrani M, AlQahtani T, AlGhamdi S, Badreldin H, Al Alshaykh H. Cancer Biomarkers and Precision Oncology: A Review of Recent Trends and Innovations. Clin Med Insights Oncol 2024; 18:11795549241298541. [PMID: 39559827 PMCID: PMC11571259 DOI: 10.1177/11795549241298541] [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: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
The discovery of cancer-specific biomarkers has resulted in major advancements in the field of cancer diagnostics and therapeutics, therefore significantly lowering cancer-related morbidity and mortality. Cancer biomarkers can be generally classified as prognostic biomarkers that predict specific disease outcomes and predictive biomarkers that predict disease response to targeted therapeutic interventions. As research in the area of predictive biomarkers continues to grow, precision medicine becomes far more integrated in cancer treatment. This article presents a general overview on the most recent advancements in the area of cancer biomarkers, immunotherapy, artificial intelligence, and pharmacogenomics of the Middle East.
Collapse
Affiliation(s)
- Maha AlDoughaim
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Nada AlSuhebany
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Mohammed AlZahrani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Tariq AlQahtani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Sahar AlGhamdi
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hisham Badreldin
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hana Al Alshaykh
- Pharmaceutical Care Devision, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh, Saudi Arabia
| |
Collapse
|
120
|
Ercan C, Renne SL, Di Tommaso L, Ng CKY, Piscuoglio S, Terracciano LM. Hepatocellular Carcinoma Immune Microenvironment Analysis: A Comprehensive Assessment with Computational and Classical Pathology. Clin Cancer Res 2024; 30:5105-5115. [PMID: 39264292 DOI: 10.1158/1078-0432.ccr-24-0960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/16/2024] [Accepted: 09/10/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE The spatial variability and clinical relevance of the tumor immune microenvironment (TIME) are still poorly understood for hepatocellular carcinoma (HCC). In this study, we aim to develop a deep learning (DL)-based image analysis model for the spatial analysis of immune cell biomarkers and microscopically evaluate the distribution of immune infiltration. EXPERIMENTAL DESIGN Ninety-two HCC surgical liver resections and 51 matched needle biopsies were histologically classified according to their immunophenotypes: inflamed, immune-excluded, and immune-desert. To characterize the TIME on immunohistochemistry (IHC)-stained slides, we designed a multistage DL algorithm, IHC-TIME, to automatically detect immune cells and their localization in the TIME in tumor-stroma and center-border segments. RESULTS Two models were trained to detect and localize the immune cells on IHC-stained slides. The framework models (i.e., immune cell detection models and tumor-stroma segmentation) reached 98% and 91% accuracy, respectively. Patients with inflamed tumors showed better recurrence-free survival than those with immune-excluded or immune-desert tumors. Needle biopsies were found to be 75% accurate in representing the immunophenotypes of the main tumor. Finally, we developed an algorithm that defines immunophenotypes automatically based on the IHC-TIME analysis, achieving an accuracy of 80%. CONCLUSIONS Our DL-based tool can accurately analyze and quantify immune cells on IHC-stained slides of HCC. Microscopic classification of the TIME can stratify HCC according to the patient prognosis. Needle biopsies can provide valuable insights for TIME-related prognostic prediction, albeit with specific constraints. The computational pathology tool provides a new way to study the HCC TIME.
Collapse
Affiliation(s)
- Caner Ercan
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Salvatore Lorenzo Renne
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Luca Di Tommaso
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Charlotte K Y Ng
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Salvatore Piscuoglio
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Luigi M Terracciano
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
121
|
Blechman SE, Wright ES. Applications of Machine Learning on Electronic Health Record Data to Combat Antibiotic Resistance. J Infect Dis 2024; 230:1073-1082. [PMID: 38995050 PMCID: PMC11565868 DOI: 10.1093/infdis/jiae348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 06/05/2024] [Accepted: 07/10/2024] [Indexed: 07/13/2024] Open
Abstract
There is growing excitement about the clinical use of artificial intelligence and machine learning (ML) technologies. Advancements in computing and the accessibility of ML frameworks enable researchers to easily train predictive models using electronic health record data. However, several practical factors must be considered when employing ML on electronic health record data. We provide a primer on ML and approaches commonly taken to address these challenges. To illustrate how these approaches have been applied to address antimicrobial resistance, we review the use of electronic health record data to construct ML models for predicting pathogen carriage or infection, optimizing empiric therapy, and aiding antimicrobial stewardship tasks. ML shows promise in promoting the appropriate use of antimicrobials, although clinical deployment is limited. We conclude by describing the potential dangers of, and barriers to, implementation of ML models in the clinic.
Collapse
Affiliation(s)
- Samuel E Blechman
- Department of Biomedical Informatics, University of Pittsburgh, Pennsylvania
| | - Erik S Wright
- Department of Biomedical Informatics, University of Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pennsylvania
| |
Collapse
|
122
|
Tsou P, Wu CJ. Classifying driver mutations of papillary thyroid carcinoma on whole slide image: an automated workflow applying deep convolutional neural network. Front Endocrinol (Lausanne) 2024; 15:1395979. [PMID: 39564124 PMCID: PMC11573888 DOI: 10.3389/fendo.2024.1395979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 10/17/2024] [Indexed: 11/21/2024] Open
Abstract
Background Informative biomarkers play a vital role in guiding clinical decisions regarding management of cancers. We have previously demonstrated the potential of a deep convolutional neural network (CNN) for predicting cancer driver gene mutations from expert-curated histopathologic images in papillary thyroid carcinomas (PTCs). Recognizing the importance of whole slide image (WSI) analysis for clinical application, we aimed to develop an automated image preprocessing workflow that uses WSI inputs to categorize PTCs based on driver mutations. Methods Histopathology slides from The Cancer Genome Atlas (TCGA) repository were utilized for diagnostic purposes. These slides underwent an automated tile extraction and preprocessing pipeline to ensure analysis-ready quality. Next, the extracted image tiles were utilized to train a deep learning CNN model, specifically Google's Inception v3, for the classification of PTCs. The model was trained to distinguish between different groups based on BRAFV600E or RAS mutations. Results The newly developed pipeline performed equally well as the expert-curated image classifier. The best model achieved Area Under the Curve (AUC) values of 0.86 (ranging from 0.847 to 0.872) for validation and 0.865 (ranging from 0.854 to 0.876) for the final testing subsets. Notably, it accurately predicted 90% of tumors in the validation set and 84.2% in the final testing set. Furthermore, the performance of our new classifier showed a strong correlation with the expert-curated classifier (Spearman rho = 0.726, p = 5.28 e-08), and correlated with the molecular expression-based classifier, BRS (BRAF-RAS scores) (Spearman rho = 0.418, p = 1.92e-13). Conclusions Utilizing WSIs, we implemented an automated workflow with deep CNN model that accurately classifies driver mutations in PTCs.
Collapse
Affiliation(s)
- Peiling Tsou
- Department of Genomic Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| | - Chang-Jiun Wu
- Department of Genomic Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
123
|
Fu M, Lin Y, Yang J, Cheng J, Lin L, Wang G, Long C, Xu S, Lu J, Li G, Yan J, Chen G, Zhuo S, Chen D. Multitask machine learning-based tumor-associated collagen signatures predict peritoneal recurrence and disease-free survival in gastric cancer. Gastric Cancer 2024; 27:1242-1257. [PMID: 39271552 DOI: 10.1007/s10120-024-01551-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Accurate prediction of peritoneal recurrence for gastric cancer (GC) is crucial in clinic. The collagen alterations in tumor microenvironment affect the migration and treatment response of cancer cells. Herein, we proposed multitask machine learning-based tumor-associated collagen signatures (TACS), which are composed of quantitative collagen features derived from multiphoton imaging, to simultaneously predict peritoneal recurrence (TACSPR) and disease-free survival (TACSDFS). METHODS Among 713 consecutive patients, with 275 in training cohort, 222 patients in internal validation cohort, and 216 patients in external validation cohort, we developed and validated a multitask machine learning model for simultaneously predicting peritoneal recurrence (TACSPR) and disease-free survival (TACSDFS). The accuracy of the model for prediction of peritoneal recurrence and prognosis as well as its association with adjuvant chemotherapy were evaluated. RESULTS The TACSPR and TACSDFS were independently associated with peritoneal recurrence and disease-free survival in three cohorts, respectively (all P < 0.001). The TACSPR demonstrated a favorable performance for peritoneal recurrence in all three cohorts. In addition, the TACSDFS also showed a satisfactory accuracy for disease-free survival among included patients. For stage II and III diseases, adjuvant chemotherapy improved the survival of patients with low TACSPR and low TACSDFS, or high TACSPR and low TACSDFS, or low TACSPR and high TACSDFS, but had no impact on patients with high TACSPR and high TACSDFS. CONCLUSIONS The multitask machine learning model allows accurate prediction of peritoneal recurrence and survival for GC and could distinguish patients who might benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Meiting Fu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Guangzhou, 510515, People's Republic of China
- School of Science, Jimei University, Xiamen, 361021, People's Republic of China
| | - Yuyu Lin
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Junyao Yang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jiaxin Cheng
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Liyan Lin
- Department of Pathology, Fujian Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Guangxing Wang
- School of Science, Jimei University, Xiamen, 361021, People's Republic of China
| | - Chenyan Long
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Shuoyu Xu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jianping Lu
- Department of Pathology, Fujian Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Guoxin Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Gang Chen
- Department of Pathology, Fujian Key Laboratory of Translational Cancer Medicine, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, People's Republic of China
| | - Shuangmu Zhuo
- School of Science, Jimei University, Xiamen, 361021, People's Republic of China
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Normal University, Fuzhou, 350007, People's Republic of China
| | - Dexin Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
124
|
Reis‐Filho JS, Scaltriti M, Kapil A, Sade H, Galbraith S. Shifting the paradigm in personalized cancer care through next-generation therapeutics and computational pathology. Mol Oncol 2024; 18:2607-2611. [PMID: 39214683 PMCID: PMC11547233 DOI: 10.1002/1878-0261.13724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The incorporation of novel therapeutic agents such as antibody-drug conjugates, radio-conjugates, T-cell engagers, and chimeric antigen receptor cell therapies represents a paradigm shift in oncology. Cell-surface target quantification, quantitative assessment of receptor internalization, and changes in the tumor microenvironment (TME) are essential variables in the development of biomarkers for patient selection and therapeutic response. Assessing these parameters requires capabilities that transcend those of traditional biomarker approaches based on immunohistochemistry, in situ hybridization and/or sequencing assays. Computational pathology is emerging as a transformative solution in this new therapeutic landscape, enabling detailed assessment of not only target presence, expression levels, and intra-tumor distribution but also of additional phenotypic features of tumor cells and their surrounding TME. Here, we delineate the pivotal role of computational pathology in enhancing the efficacy and specificity of these advanced therapeutics, underscoring the integration of novel artificial intelligence models that promise to revolutionize biomarker discovery and drug development.
Collapse
Affiliation(s)
- Jorge S. Reis‐Filho
- Cancer Biomarker Development, Oncology Research and DevelopmentAstraZenecaGaithersburgMDUSA
| | - Maurizio Scaltriti
- Translational Medicine, Oncology Research and DevelopmentAstraZenecaGaithersburgMDUSA
| | - Ansh Kapil
- Oncology Research and DevelopmentAstraZeneca Computational Pathology GmbH, AstraZenecaMunichGermany
| | - Hadassah Sade
- Oncology Research and DevelopmentAstraZeneca Computational Pathology GmbH, AstraZenecaMunichGermany
| | - Susan Galbraith
- Oncology Research and DevelopmentAstraZenecaGaithersburgMarylandUSA
| |
Collapse
|
125
|
Ding S, Li J, Wang J, Ying S, Shi J. Multimodal Co-Attention Fusion Network With Online Data Augmentation for Cancer Subtype Classification. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:3977-3989. [PMID: 38801690 DOI: 10.1109/tmi.2024.3405535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
It is an essential task to accurately diagnose cancer subtypes in computational pathology for personalized cancer treatment. Recent studies have indicated that the combination of multimodal data, such as whole slide images (WSIs) and multi-omics data, could achieve more accurate diagnosis. However, robust cancer diagnosis remains challenging due to the heterogeneity among multimodal data, as well as the performance degradation caused by insufficient multimodal patient data. In this work, we propose a novel multimodal co-attention fusion network (MCFN) with online data augmentation (ODA) for cancer subtype classification. Specifically, a multimodal mutual-guided co-attention (MMC) module is proposed to effectively perform dense multimodal interactions. It enables multimodal data to mutually guide and calibrate each other during the integration process to alleviate inter- and intra-modal heterogeneities. Subsequently, a self-normalizing network (SNN)-Mixer is developed to allow information communication among different omics data and alleviate the high-dimensional small-sample size problem in multi-omics data. Most importantly, to compensate for insufficient multimodal samples for model training, we propose an ODA module in MCFN. The ODA module leverages the multimodal knowledge to guide the data augmentations of WSIs and maximize the data diversity during model training. Extensive experiments are conducted on the public TCGA dataset. The experimental results demonstrate that the proposed MCFN outperforms all the compared algorithms, suggesting its effectiveness.
Collapse
|
126
|
Li X, Li L, He N, Kou D, Chen S, Song H, Yan X. Pathomics signatures and cuproptosis-related genes signatures for prediction of prognosis in patients with hepatocellular carcinoma. Transl Cancer Res 2024; 13:5473-5483. [PMID: 39525011 PMCID: PMC11543060 DOI: 10.21037/tcr-24-350] [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: 03/04/2024] [Accepted: 08/14/2024] [Indexed: 11/16/2024]
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignant tumor with high heterogeneity and poor prognosis, so early prediction and treatment are still difficult. Cuproptosis is a newly discovered type of programmed cell death that has been shown to be closely related to the occurrence and progression of HCC. Cancer morphology is influenced by genetic drivers, and computational pathology methods typically use tissue images such as entire slide images as input to predict clinical or genetic features. Therefore, the comprehensive analysis of pathological features and genomic data provides a feasible way to explore the potential mechanism of the tumor. The objective of this study was to develop a prediction model for HCC prognosis based on the pathomics signatures (PS) and the genomics signatures (GS). Methods A dataset comprising 315 HCC patients was randomly divided into a training set (n=200) and a validation set (n=115). Prognostic models related to PS and GS were constructed by univariate and multivariate Cox regression analyses and least absolute shrinkage and selection operator (LASSO) regression analysis. Kaplan-Meier survival analysis, receiver operating characteristic (ROC) curve, univariate and multivariate Cox analyses, and nomogram were used to evaluate the predictive performance of the prognostic model. The prognostic value of the model was internally validated. Results A prognostic model incorporating clinical features, PS, and GS was developed using Cox regression analysis and LASSO regression analyses. Kaplan-Meier survival analysis revealed statistically significant differences in survival time between high-risk and low-risk subgroups in both the training and validation datasets (PS: P=0.003 and <0.001, respectively; GS: P=0.008 and 0.004, respectively). The time-dependent ROC curve showed favorable predictive value for survival in both the training and validation sets. The area under the ROC curves at 1, 3, and 5 years was 0.750, 0.830, and 0.870 in the training set, and 0.780, 0.810, and 0.760 in the validation set, respectively. A nomogram model based on the risk model score could effectively predict the survival probability of HCC patients. The calibration curves further demonstrated the good predictive capability of the nomogram model. Conclusions The prognostic model incorporating PS and GS could effectively predict the prognosis of HCC patients.
Collapse
Affiliation(s)
- Xiaoliang Li
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Lina Li
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Nan He
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Dan Kou
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Shizhao Chen
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Hui Song
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| | - Xiang Yan
- Department of Geriatric, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
127
|
Zhou X, Man M, Cui M, Zhou X, Hu Y, Liu Q, Deng Y. Relationship between EZH2 expression and prognosis of patients with hepatocellular carcinoma using a pathomics predictive model. Heliyon 2024; 10:e38562. [PMID: 39640777 PMCID: PMC11619983 DOI: 10.1016/j.heliyon.2024.e38562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/04/2024] [Accepted: 09/26/2024] [Indexed: 12/07/2024] Open
Abstract
Background Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) is overexpressed in hepatocellular carcinoma, promoting tumorigenesis and correlating with poor prognosis. Traditional histopathological examinations are insufficient to accurately predict hepatocellular carcinoma (HCC) survival; however, pathomics models can predict EZH2 expression and HCC prognosis. This study aimed to investigate the relationship between pathomics features and EZH2 expression for predicting overall survival of patients with HCC. Methods We analyzed 267 patients with HCC from the Cancer Genome Atlas database, with available pathological images and gene expression data. RNA sequencing data were divided into high and low EZH2 expression groups for prognosis and survival analysis. Pathological image features were screened using mRMR_RFE. A pathological model was constructed using a gradient boosting machine (GBM) algorithm, and efficiency evaluation and survival analysis of the model were performed. The R package "survminer" took the pathomics score (PS) cutoff value of 0.4628 to divide the patients into two groups: high and low PS expression. Survival analyses included Kaplan-Meier curve analysis, univariate and multivariate Cox regression analyses, and interaction tests. Potential pathomechanisms were explored through enrichment, differential, immune cell infiltration abundance, and gene mutation analyses. Result EZH2 was highly expressed in tumor samples but poorly expressed in normal tissue samples. Univariate and multivariate Cox regression analyses revealed that EZH2 was an independent risk factor for HCC (hazard ratio [HR], 2.792 and 3.042, respectively). Seven imaging features were selected to construct a pathomics model to predict EZH2. Decision curve analysis showed that the model had high clinical utility. Multivariate Cox regression analysis showed that high PS expression was an independent risk factor for HCC prognosis (HR, 2.446). The Kaplan-Meier curve showed that high PS expression was a risk factor for overall survival. Conclusion EZH2 expression can affect the prognosis of patients with liver cancer. Our pathological model could predict EZH2 expression and prognosis of patients with HCC with high accuracy and robustness, making it a new and potentially valuable tool.
Collapse
Affiliation(s)
- Xulin Zhou
- Department of Oncology, Hefei BOE Hospital, Hefei, PR China
| | - Muran Man
- Department of Oncology, People's Hospital of Shizhong District, Zaozhuang City, Shandong Province, PR China
| | - Min Cui
- Affiliated Hospital Of Jining Medical University (Shanxian Central Hospital), Heze City, Shandong Province, PR China
| | - Xiang Zhou
- People's Hospital of Xinjiang Uygur Autonomous Region Urumqi, Xinjiang, CN, PR China
| | - Yan Hu
- Department of Oncology, Hefei BOE Hospital, Hefei, PR China
| | - Qinghua Liu
- Department of Oncology, Deyang People's Hospital, Deyang, Sichuan, CN, PR China
| | - Youxing Deng
- Department of Oncology, Hefei BOE Hospital, Hefei, PR China
| |
Collapse
|
128
|
Xiaojian Y, Zhanbo Q, Jian C, Zefeng W, Jian L, Jin L, Yuefen P, Shuwen H. Deep learning application in prediction of cancer molecular alterations based on pathological images: a bibliographic analysis via CiteSpace. J Cancer Res Clin Oncol 2024; 150:467. [PMID: 39422817 PMCID: PMC11489169 DOI: 10.1007/s00432-024-05992-z] [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: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The advancements in artificial intelligence (AI) technology for image recognition were propelling molecular pathology research into a new era. OBJECTIVE To summarize the hot spots and research trends in the field of molecular pathology image recognition. METHODS Relevant articles from January 1st, 2010, to August 25th, 2023, were retrieved from the Web of Science Core Collection. Subsequently, CiteSpace was employed for bibliometric and visual analysis, generating diverse network diagrams illustrating keywords, highly cited references, hot topics, and research trends. RESULTS A total of 110 relevant articles were extracted from a pool of 10,205 articles. The overall publication count exhibited a rising trend each year. The leading contributors in terms of institutions, countries, and authors were Maastricht University (11 articles), the United States (38 articles), and Kather Jacob Nicholas (9 articles), respectively. Half of the top ten research institutions, based on publication volume, were affiliated with Germany. The most frequently cited article was authored by Nicolas Coudray et al. accumulating 703 citations. The keyword "Deep learning" had the highest frequency in 2019. Notably, the highlighted keywords from 2022 to 2023 included "microsatellite instability", and there were 21 articles focusing on utilizing algorithms to recognize microsatellite instability (MSI) in colorectal cancer (CRC) pathological images. CONCLUSION The use of DL is expected to provide a new strategy to effectively solve the current problem of time-consuming and expensive molecular pathology detection. Therefore, further research is needed to address issues, such as data quality and standardization, model interpretability, and resource and infrastructure requirements.
Collapse
Affiliation(s)
- Yu Xiaojian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Wang Zefeng
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jin
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Pan Yuefen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France.
| |
Collapse
|
129
|
Xu D, Li H, Su F, Qiu S, Tong H, Huang M, Yao J. Identification of middle cerebral artery stenosis in transcranial Doppler using a modified VGG-16. Front Neurol 2024; 15:1394435. [PMID: 39479004 PMCID: PMC11521853 DOI: 10.3389/fneur.2024.1394435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/29/2024] [Indexed: 11/02/2024] Open
Abstract
Objectives The diagnosis of intracranial atherosclerotic stenosis (ICAS) is of great significance for the prevention of stroke. Deep learning (DL)-based artificial intelligence techniques may aid in the diagnosis. The study aimed to identify ICAS in the middle cerebral artery (MCA) based on a modified DL model. Methods This retrospective study included two datasets. Dataset1 consisted of 3,068 transcranial Doppler (TCD) images of the MCA from 1,729 patients, which were assessed as normal or stenosis by three physicians with varying levels of experience, in conjunction with other medical imaging data. The data were used to improve and train the VGG16 models. Dataset2 consisted of TCD images of 90 people who underwent physical examination, which were used to verify the robustness of the model and compare the consistency between the model and human physicians. Results The accuracy, precision, specificity, sensitivity, and area under curve (AUC) of the best model VGG16 + Squeeze-and-Excitation (SE) + skip connection (SC) on dataset1 reached 85.67 ± 0.43(%),87.23 ± 1.17(%),87.73 ± 1.47(%),83.60 ± 1.60(%), and 0.857 ± 0.004, while those of dataset2 were 93.70 ± 2.80(%),62.65 ± 11.27(%),93.00 ± 3.11(%),100.00 ± 0.00(%), and 0.965 ± 0.016. The kappa coefficient showed that it reached the recognition level of senior doctors. Conclusion The improved DL model has a good diagnostic effect for MCV stenosis in TCD images and is expected to help in ICAS screening.
Collapse
Affiliation(s)
- Dong Xu
- Department of Neuroelectrophysiology, Anyang People's Hospital, Anyang, China
| | - Hao Li
- Shenzhen Institute for Advanced Study, University of Electronic Science and Technology of China, Shenzhen, China
| | - Fanghui Su
- Department of Neuroelectrophysiology, Anyang People's Hospital, Anyang, China
| | - Sizheng Qiu
- Department of Neuroelectrophysiology, Anyang People's Hospital, Anyang, China
| | - Huixia Tong
- Department of Neuroelectrophysiology, Anyang People's Hospital, Anyang, China
| | - Meifeng Huang
- Department of Neuroelectrophysiology, Anyang People's Hospital, Anyang, China
| | - Jianzhong Yao
- Department of Neuroelectrophysiology, Anyang People's Hospital, Anyang, China
| |
Collapse
|
130
|
Pareja F, Dopeso H, Wang YK, Gazzo AM, Brown DN, Banerjee M, Selenica P, Bernhard JH, Derakhshan F, da Silva EM, Colon-Cartagena L, Basili T, Marra A, Sue J, Ye Q, Da Cruz Paula A, Yildirim SY, Pei X, Safonov A, Green H, Gill KY, Zhu Y, Lee MCH, Godrich RA, Casson A, Weigelt B, Riaz N, Wen HY, Brogi E, Mandelker DL, Hanna MG, Kunz JD, Rothrock B, Chandarlapaty S, Kanan C, Oakley J, Klimstra DS, Fuchs TJ, Reis-Filho JS. A Genomics-Driven Artificial Intelligence-Based Model Classifies Breast Invasive Lobular Carcinoma and Discovers CDH1 Inactivating Mechanisms. Cancer Res 2024; 84:3478-3489. [PMID: 39106449 PMCID: PMC11479818 DOI: 10.1158/0008-5472.can-24-1322] [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: 04/23/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/09/2024]
Abstract
Artificial intelligence (AI) systems can improve cancer diagnosis, yet their development often relies on subjective histologic features as ground truth for training. Herein, we developed an AI model applied to histologic whole-slide images using CDH1 biallelic mutations, pathognomonic for invasive lobular carcinoma (ILC) in breast neoplasms, as ground truth. The model accurately predicted CDH1 biallelic mutations (accuracy = 0.95) and diagnosed ILC (accuracy = 0.96). A total of 74% of samples classified by the AI model as having CDH1 biallelic mutations but lacking these alterations displayed alternative CDH1 inactivating mechanisms, including a deleterious CDH1 fusion gene and noncoding CDH1 genetic alterations. Analysis of internal and external validation cohorts demonstrated 0.95 and 0.89 accuracy for ILC diagnosis, respectively. The latent features of the AI model correlated with human-explainable histopathologic features. Taken together, this study reports the construction of an AI algorithm trained using a genetic rather than histologic ground truth that can robustly classify ILCs and uncover CDH1 inactivating mechanisms, providing the basis for orthogonal ground truth utilization for development of diagnostic AI models applied to whole-slide image. Significance: Genetic alterations linked to strong genotypic-phenotypic correlations can be utilized to develop AI systems applied to pathology that facilitate cancer diagnosis and biologic discoveries.
Collapse
Affiliation(s)
- Fresia Pareja
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Higinio Dopeso
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Andrea M. Gazzo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David N. Brown
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Fatemeh Derakhshan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Edaise M. da Silva
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lorraine Colon-Cartagena
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Thais Basili
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Antonio Marra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Qiqi Ye
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Selma Yeni Yildirim
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xin Pei
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anton Safonov
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hunter Green
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kaitlyn Y. Gill
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yingjie Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Hannah Y. Wen
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Edi Brogi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Diana L. Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Matthew G. Hanna
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | - Sarat Chandarlapaty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christopher Kanan
- Department of Computer Science, University of Rochester, Rochester, NY
| | | | | | | | - Jorge S. Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Current employment: AstraZeneca
| |
Collapse
|
131
|
Luvhengo TE, Moeng MS, Sishuba NT, Makgoka M, Jonas L, Mamathuntsha TG, Mbambo T, Kagodora SB, Dlamini Z. Holomics and Artificial Intelligence-Driven Precision Oncology for Medullary Thyroid Carcinoma: Addressing Challenges of a Rare and Aggressive Disease. Cancers (Basel) 2024; 16:3469. [PMID: 39456563 PMCID: PMC11505703 DOI: 10.3390/cancers16203469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objective: Medullary thyroid carcinoma (MTC) is a rare yet aggressive form of thyroid cancer comprising a disproportionate share of thyroid cancer-related mortalities, despite its low prevalence. MTC differs from other differentiated thyroid malignancies due to its heterogeneous nature, presenting complexities in both hereditary and sporadic cases. Traditional management guidelines, which are designed primarily for papillary thyroid carcinoma (PTC), fall short in providing the individualized care required for patients with MTC. In recent years, the sheer volume of data generated from clinical evaluations, radiological imaging, pathological assessments, genetic mutations, and immunological profiles has made it humanly impossible for clinicians to simultaneously analyze and integrate these diverse data streams effectively. This data deluge necessitates the adoption of advanced technologies to assist in decision-making processes. Holomics, which is an integrated approach that combines various omics technologies, along with artificial intelligence (AI), emerges as a powerful solution to address these challenges. Methods: This article reviews how AI-driven precision oncology can enhance the diagnostic workup, staging, risk stratification, management, and follow-up care of patients with MTC by processing vast amounts of complex data quickly and accurately. Articles published in English language and indexed in Pubmed were searched. Results: AI algorithms can identify patterns and correlations that may not be apparent to human clinicians, thereby improving the precision of personalized treatment plans. Moreover, the implementation of AI in the management of MTC enables the collation and synthesis of clinical experiences from across the globe, facilitating a more comprehensive understanding of the disease and its treatment outcomes. Conclusions: The integration of holomics and AI in the management of patients with MTC represents a significant advancement in precision oncology. This innovative approach not only addresses the complexities of a rare and aggressive disease but also paves the way for global collaboration and equitable healthcare solutions, ultimately transforming the landscape of treatment and care of patients with MTC. By leveraging AI and holomics, we can strive toward making personalized healthcare accessible to every individual, regardless of their economic status, thereby improving overall survival rates and quality of life for MTC patients worldwide. This global approach aligns with the United Nations Sustainable Development Goal 3, which aims to ensure healthy lives and promote well-being at all ages.
Collapse
Affiliation(s)
| | - Maeyane Stephens Moeng
- Department of Surgery, University of the Witwatersrand, Johannesburg 2193, South Africa; (M.S.M.); (N.T.S.)
| | - Nosisa Thabile Sishuba
- Department of Surgery, University of the Witwatersrand, Johannesburg 2193, South Africa; (M.S.M.); (N.T.S.)
| | - Malose Makgoka
- Department of Surgery, University of Pretoria, Pretoria 0002, South Africa;
| | - Lusanda Jonas
- Department of Surgery, University of Limpopo, Mankweng 4062, South Africa; (L.J.); (T.G.M.)
| | | | - Thandanani Mbambo
- Department of Surgery, University of KwaZulu-Natal, Durban 2025, South Africa;
| | | | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI, Precision Oncology and Cancer Prevention (POCP), University of Pretoria, Pretoria 0028, South Africa;
| |
Collapse
|
132
|
Wu IC, Chen YC, Karmakar R, Mukundan A, Gabriel G, Wang CC, Wang HC. Advancements in Hyperspectral Imaging and Computer-Aided Diagnostic Methods for the Enhanced Detection and Diagnosis of Head and Neck Cancer. Biomedicines 2024; 12:2315. [PMID: 39457627 PMCID: PMC11504349 DOI: 10.3390/biomedicines12102315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/12/2024] [Accepted: 09/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Head and neck cancer (HNC), predominantly squamous cell carcinoma (SCC), presents a significant global health burden. Conventional diagnostic approaches often face challenges in terms of achieving early detection and accurate diagnosis. This review examines recent advancements in hyperspectral imaging (HSI), integrated with computer-aided diagnostic (CAD) techniques, to enhance HNC detection and diagnosis. Methods: A systematic review of seven rigorously selected studies was performed. We focused on CAD algorithms, such as convolutional neural networks (CNNs), support vector machines (SVMs), and linear discriminant analysis (LDA). These are applicable to the hyperspectral imaging of HNC tissues. Results: The meta-analysis findings indicate that LDA surpasses other algorithms, achieving an accuracy of 92%, sensitivity of 91%, and specificity of 93%. CNNs exhibit moderate performance, with an accuracy of 82%, sensitivity of 77%, and specificity of 86%. SVMs demonstrate the lowest performance, with an accuracy of 76% and sensitivity of 48%, but maintain a high specificity level at 89%. Additionally, in vivo studies demonstrate superior performance when compared to ex vivo studies, reporting higher accuracy (81%), sensitivity (83%), and specificity (79%). Conclusion: Despite these promising findings, challenges persist, such as HSI's sensitivity to external conditions, the need for high-resolution and high-speed imaging, and the lack of comprehensive spectral databases. Future research should emphasize dimensionality reduction techniques, the integration of multiple machine learning models, and the development of extensive spectral libraries to enhance HSI's clinical utility in HNC diagnostics. This review underscores the transformative potential of HSI and CAD techniques in revolutionizing HNC diagnostics, facilitating more accurate and earlier detection, and improving patient outcomes.
Collapse
Affiliation(s)
- I-Chen Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, No. 100, Tzyou 1st Rd., Sanmin Dist., Kaohsiung City 80756, Taiwan;
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, No. 100, Tzyou 1st Rd., Sanmin Dist., Kaohsiung City 80756, Taiwan
| | - Yen-Chun Chen
- Department of Gastroenterology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 2, Minsheng Road, Dalin, Chiayi 62247, Taiwan;
| | - Riya Karmakar
- Department of Mechanical Engineering, National Chung Cheng University, 168, University Rd., Min Hsiung, Chiayi 62102, Taiwan; (R.K.); (A.M.)
| | - Arvind Mukundan
- Department of Mechanical Engineering, National Chung Cheng University, 168, University Rd., Min Hsiung, Chiayi 62102, Taiwan; (R.K.); (A.M.)
| | - Gahiga Gabriel
- Vel Tech Rangarajan Dr. Sagunthala R&D Institute of Science and Technology, No. 42, Avadi-Vel Tech Road Vel Nagar, Avadi, Chennai 600062, Tamil Nadu, India;
| | - Chih-Chiang Wang
- Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, 2, Zhongzheng 1st Rd., Lingya District, Kaohsiung City 80284, Taiwan
- School of Medicine, National Defense Medical Center, No. 161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City 11490, Taiwan
| | - Hsiang-Chen Wang
- Department of Mechanical Engineering, National Chung Cheng University, 168, University Rd., Min Hsiung, Chiayi 62102, Taiwan; (R.K.); (A.M.)
- Hitspectra Intelligent Technology Co., Ltd., 8F. 11-1, No. 25, Chenggong 2nd Rd., Qianzhen Dist., Kaohsiung City 80661, Taiwan
| |
Collapse
|
133
|
Harder C, Pryalukhin A, Quaas A, Eich ML, Tretiakova M, Klein S, Seper A, Heidenreich A, Netto GJ, Hulla W, Büttner R, Bozek K, Tolkach Y. Enhancing Prostate Cancer Diagnosis: Artificial Intelligence-Driven Virtual Biopsy for Optimal Magnetic Resonance Imaging-Targeted Biopsy Approach and Gleason Grading Strategy. Mod Pathol 2024; 37:100564. [PMID: 39029903 DOI: 10.1016/j.modpat.2024.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/28/2024] [Accepted: 07/06/2024] [Indexed: 07/21/2024]
Abstract
An optimal approach to magnetic resonance imaging fusion targeted prostate biopsy (PBx) remains unclear (number of cores, intercore distance, Gleason grading [GG] principle). The aim of this study was to develop a precise pixel-wise segmentation diagnostic artificial intelligence (AI) algorithm for tumor detection and GG as well as an algorithm for virtual prostate biopsy that are used together to systematically investigate and find an optimal approach to targeted PBx. Pixel-wise AI algorithms for tumor detection and GG were developed using a high-quality, manually annotated data set (slides n = 442) after fast-track annotation transfer into segmentation style. To this end, a virtual biopsy algorithm was developed that can perform random biopsies from tumor regions in whole-mount whole-slide images with predefined parameters. A cohort of 115 radical prostatectomy (RP) patient cases with clinically significant, magnetic resonance imaging-visible tumors (n = 121) was used for systematic studies of the optimal biopsy approach. Three expert genitourinary (GU) pathologists (Y.T., A.P., A.Q.) participated in the validation. The tumor detection algorithm (aware version sensitivity/specificity 0.99/0.90, balanced version 0.97/0.97) and GG algorithm (quadratic kappa range vs pathologists 0.77-0.78) perform on par with expert GU pathologists. In total, 65,340 virtual biopsies were performed to study different biopsy approaches with the following results: (1) 4 biopsy cores is the optimal number for a targeted PBx, (2) cumulative GG strategy is superior to using maximal Gleason score for single cores, (3) controlling for minimal intercore distance does not improve the predictive accuracy for the RP Gleason score, (4) using tertiary Gleason pattern principle (for AI tool) in cumulative GG strategy might allow better predictions of final RP Gleason score. The AI algorithm (based on cumulative GG strategy) predicted the RP Gleason score of the tumor better than 2 of the 3 expert GU pathologists. In this study, using an original approach of virtual prostate biopsy on the real cohort of patient cases, we find the optimal approach to the biopsy procedure and the subsequent GG of a targeted PBx. We publicly release 2 large data sets with associated expert pathologists' GG and our virtual biopsy algorithm.
Collapse
Affiliation(s)
- Christian Harder
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Alexey Pryalukhin
- Institute of Pathology, Wiener Neustadt State Hospital, Wiener Neustadt, Austria
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Marie-Lisa Eich
- Institute of Pathology, University Hospital Cologne, Cologne, Germany; Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humbolt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Maria Tretiakova
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington
| | - Sebastian Klein
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Alexander Seper
- Institute of Pathology, Wiener Neustadt State Hospital, Wiener Neustadt, Austria; Danube Private University, Austria
| | - Axel Heidenreich
- Department of Urology, Pro-Oncology, Robot-Assisted and Specialized Urologic Surgery, University Hospital Cologne, Cologne, Germany; Department of Urology, Medical University Vienna, Austria
| | - George Jabboure Netto
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadephia, Pennsylvania
| | - Wolfgang Hulla
- Institute of Pathology, Wiener Neustadt State Hospital, Wiener Neustadt, Austria
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Kasia Bozek
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Yuri Tolkach
- Institute of Pathology, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
134
|
Hu CL, Wang YC, Wu WF, Xi Y. Evaluation of AI-enhanced non-mydriatic fundus photography for diabetic retinopathy screening. Photodiagnosis Photodyn Ther 2024; 49:104331. [PMID: 39245303 DOI: 10.1016/j.pdpdt.2024.104331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE To assess the feasibility of using non-mydriatic fundus photography in conjunction with an artificial intelligence (AI) reading platform for large-scale screening of diabetic retinopathy (DR). METHODS In this study, we selected 120 patients with diabetes hospitalized in our institution from December 2019 to April 2021. Retinal imaging of 240 eyes was obtained using non-mydriatic fundus photography. The fundus images of these patients were divided into two groups based on different interpretation methods. In Experiment Group 1, the images were analyzed and graded for DR diagnosis using an AI reading platform. In Experiment Group 2, the images were analyzed and graded for DR diagnosis by an associate chief physician in ophthalmology, specializing in fundus diseases. Concurrently, all patients underwent the gold standard for DR diagnosis and grading-fundus fluorescein angiography (FFA)-with the outcomes serving as the Control Group. The diagnostic value of the two methods was assessed by comparing the results of Experiment Groups 1 and 2 with those of the Control Group. RESULTS Keeping the control group (FFA results) as the gold standard, no significant differences were observed between the two experimental groups regarding diagnostic sensitivity, specificity, false positive rate, false negative rate, positive predictive value, negative predictive value, Youden's index, Kappa value, and diagnostic accuracy (X2 = 0.371, P > 0.05). CONCLUSION Compared with the manual reading group, the AI reading group revealed no significant differences across all diagnostic indicators, exhibiting high sensitivity and specificity, as well as a relatively high positive predictive value. Additionally, it demonstrated a high level of diagnostic consistency with the gold standard. This technology holds potential for suitability in large-scale screening of DR.
Collapse
Affiliation(s)
- Chen-Liang Hu
- Department of endocrinology, Huangshan city People's Hospital, Huangshan 245000, China.
| | - Yu-Chan Wang
- Department of endocrinology, Huangshan city People's Hospital, Huangshan 245000, China
| | - Wen-Fang Wu
- Department of ophthalmology, Huangshan city People's Hospital, Huangshan 245000, China
| | - Yu Xi
- Department of endocrinology, Huangshan city People's Hospital, Huangshan 245000, China
| |
Collapse
|
135
|
Weitz P, Valkonen M, Solorzano L, Carr C, Kartasalo K, Boissin C, Koivukoski S, Kuusela A, Rasic D, Feng Y, Pouplier SS, Sharma A, Eriksson KL, Robertson S, Marzahl C, Gatenbee CD, Anderson ARA, Wodzinski M, Jurgas A, Marini N, Atzori M, Müller H, Budelmann D, Weiss N, Heldmann S, Lotz J, Wolterink JM, De Santi B, Patil A, Sethi A, Kondo S, Kasai S, Hirasawa K, Farrokh M, Kumar N, Greiner R, Latonen L, Laenkholm AV, Hartman J, Ruusuvuori P, Rantalainen M. The ACROBAT 2022 challenge: Automatic registration of breast cancer tissue. Med Image Anal 2024; 97:103257. [PMID: 38981282 DOI: 10.1016/j.media.2024.103257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 05/17/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
The alignment of tissue between histopathological whole-slide-images (WSI) is crucial for research and clinical applications. Advances in computing, deep learning, and availability of large WSI datasets have revolutionised WSI analysis. Therefore, the current state-of-the-art in WSI registration is unclear. To address this, we conducted the ACROBAT challenge, based on the largest WSI registration dataset to date, including 4,212 WSIs from 1,152 breast cancer patients. The challenge objective was to align WSIs of tissue that was stained with routine diagnostic immunohistochemistry to its H&E-stained counterpart. We compare the performance of eight WSI registration algorithms, including an investigation of the impact of different WSI properties and clinical covariates. We find that conceptually distinct WSI registration methods can lead to highly accurate registration performances and identify covariates that impact performances across methods. These results provide a comparison of the performance of current WSI registration methods and guide researchers in selecting and developing methods.
Collapse
Affiliation(s)
- Philippe Weitz
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden.
| | - Masi Valkonen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Leslie Solorzano
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden
| | - Circe Carr
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kimmo Kartasalo
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden
| | - Constance Boissin
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden
| | - Sonja Koivukoski
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Aino Kuusela
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Dusan Rasic
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Yanbo Feng
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden
| | | | - Abhinav Sharma
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden
| | - Kajsa Ledesma Eriksson
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden
| | - Stephanie Robertson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | - Chandler D Gatenbee
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, USA
| | | | - Marek Wodzinski
- Informatics Institute, University of Applied Sciences Western Switzerland, Switzerland; Department of Measurement and Electronics, AGH University of Kraków, Poland
| | - Artur Jurgas
- Informatics Institute, University of Applied Sciences Western Switzerland, Switzerland; Department of Measurement and Electronics, AGH University of Kraków, Poland
| | - Niccolò Marini
- Informatics Institute, University of Applied Sciences Western Switzerland, Switzerland; Department of Computer Science, University of Geneva, Geneva, Switzerland
| | - Manfredo Atzori
- Informatics Institute, University of Applied Sciences Western Switzerland, Switzerland; Department of Neuroscience, University of Padova, Italy
| | - Henning Müller
- Informatics Institute, University of Applied Sciences Western Switzerland, Switzerland; Medical Faculty, University of Geneva, Switzerland
| | - Daniel Budelmann
- Fraunhofer Institute for Digital Medicine MEVIS, Lübeck, Germany
| | - Nick Weiss
- Fraunhofer Institute for Digital Medicine MEVIS, Lübeck, Germany
| | - Stefan Heldmann
- Fraunhofer Institute for Digital Medicine MEVIS, Lübeck, Germany
| | - Johannes Lotz
- Fraunhofer Institute for Digital Medicine MEVIS, Lübeck, Germany
| | - Jelmer M Wolterink
- Department of Applied Mathematics, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Bruno De Santi
- Multimodality Medical Imaging, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Abhijeet Patil
- Department of Electrical Engineering, Indian Institute of Technology, Bombay, India
| | - Amit Sethi
- Department of Electrical Engineering, Indian Institute of Technology, Bombay, India
| | - Satoshi Kondo
- Graduate School of Engineering, Muroran Institute of Technology, Hokkaido, Japan
| | - Satoshi Kasai
- Faculty of Medical Technology, Niigata University of Health and Welfare, Niigata, Japan
| | | | - Mahtab Farrokh
- Department of Computing Science, University of Alberta, Edmonton, Alberta
| | - Neeraj Kumar
- Department of Computing Science, University of Alberta, Edmonton, Alberta
| | - Russell Greiner
- Department of Computing Science, University of Alberta, Edmonton, Alberta; Alberta Machine Intelligence Institute, Edmonton, Canada
| | - Leena Latonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | | | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden; MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden
| | - Pekka Ruusuvuori
- Institute of Biomedicine, University of Turku, Turku, Finland; Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mattias Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Insitutet, Stockholm, Sweden; MedTechLabs, BioClinicum, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
136
|
Nambison NK, Singh DP, Mehar R, Nambison SN, Sharma H, Sharma D, Nambison E. Pathology Report Interpretation and Disease Diagnosis Using Fuzzy Logic Embedded in an Artificial Intelligence Framework: A New Paradigm for Digital Technologies. Cureus 2024; 16:e71148. [PMID: 39525187 PMCID: PMC11548983 DOI: 10.7759/cureus.71148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Accurate diagnosis is essential for effective treatment in healthcare settings. Digital technologies have revolutionized medical diagnostics, particularly in pathology laboratory report analyses, by enhancing speed, quality, and precision. Diagnostic pathology has advanced significantly with digital imaging, AI algorithms, and computer-assisted techniques. Traditional machine learning methods require extensive training data, struggle with decision-making flexibility, and lack interpretability when handling new data. Methods This paper introduces a novel approach to address these challenges: a fuzzy logic-based system for interpreting pathology laboratory reports and diagnosing diseases, while considering the restrictions of engineered interpretations so that the degree of reliability of the output of the system is at par with human perception. Results The analysis highlights the impressive accuracy of the fuzzy logic-based diagnosis system, which is closely aligned with professional diagnoses. Notably, it correctly identified "normal" with an 83% probability, showcasing its potential for early disease detection. Performance evaluations indicated that the precision, recall, and F-measure significantly improved as the number of probable diagnoses considered increased. Furthermore, the system exhibited enhanced predictive accuracy for disease recurrence when incorporating multiple probable diagnoses, underscoring its robust clinical effectiveness. Conclusion A fuzzy logic-based system within an AI framework for interpreting pathology laboratory reports and diagnosing 17 diseases across various demographics is presented. The performance of the algorithm, assessed using specialized classification metrics, showed stable and satisfactory results. The analysis revealed a direct correlation between the number of probable diagnoses considered and improved diagnostic accuracy with enhancements in precision, recall, and F-measure. Broadening the diagnostic scope optimizes immediate patient care and long-term treatment planning, potentially enhancing patient outcomes and healthcare quality.
Collapse
Affiliation(s)
- Nisanth K Nambison
- Research, Government Homeopathic Medical College and Hospital, Bhopal, IND
| | - D P Singh
- General Medicine, LN Medical College and Research Center, Bhopal, IND
| | - Rakesh Mehar
- Pathology, Mahatma Gandhi Memorial (MGM) Medical College, Indore, IND
| | - Smita N Nambison
- Medical Technology, Neshlin Medtech Private Limited, Bhopal, IND
| | - Hariom Sharma
- Civil Engineering, Maulana Azad National Institute of Technology Bhopal, Bhopal, IND
| | - Divyansh Sharma
- Biomedical Engineering, Indian Institute of Technology Varanasi, Varanasi, IND
| | - Eva Nambison
- Data Science, Indian Institute of Technology Madras, Chennai, IND
| |
Collapse
|
137
|
Kludt C, Wang Y, Ahmad W, Bychkov A, Fukuoka J, Gaisa N, Kühnel M, Jonigk D, Pryalukhin A, Mairinger F, Klein F, Schultheis AM, Seper A, Hulla W, Brägelmann J, Michels S, Klein S, Quaas A, Büttner R, Tolkach Y. Next-generation lung cancer pathology: Development and validation of diagnostic and prognostic algorithms. Cell Rep Med 2024; 5:101697. [PMID: 39178857 PMCID: PMC11524894 DOI: 10.1016/j.xcrm.2024.101697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/25/2024] [Accepted: 07/31/2024] [Indexed: 08/26/2024]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common malignant tumors. In this study, we develop a clinically useful computational pathology platform for NSCLC that can be a foundation for multiple downstream applications and provide immediate value for patient care optimization and individualization. We train the primary multi-class tissue segmentation algorithm on a substantial, high-quality, manually annotated dataset of whole-slide images with lung adenocarcinoma and squamous cell carcinomas. We investigate two downstream applications. NSCLC subtyping algorithm is trained and validated using a large, multi-institutional (n = 6), multi-scanner (n = 5), international cohort of NSCLC cases (slides/patients 4,097/1,527). Moreover, we develop four AI-derived, fully explainable, quantitative, prognostic parameters (based on tertiary lymphoid structure and necrosis assessment) and validate them for different clinical endpoints. The computational platform enables the high-precision, quantitative analysis of H&E-stained slides. The developed prognostic parameters facilitate robust and independent risk stratification of patients with NSCLC.
Collapse
Affiliation(s)
- Carina Kludt
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany
| | - Yuan Wang
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany
| | - Waleed Ahmad
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany
| | - Andrey Bychkov
- Department of Pathology, Kameda Medical Center, Kamogawa 296-0041, Japan; Department of Pathology Informatics, Nagasaki University, Nagasaki 852-8131, Japan
| | - Junya Fukuoka
- Department of Pathology, Kameda Medical Center, Kamogawa 296-0041, Japan; Department of Pathology Informatics, Nagasaki University, Nagasaki 852-8131, Japan
| | - Nadine Gaisa
- Institute of Pathology, University Hospital Aachen, 52074 Aachen, Germany; Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Mark Kühnel
- Institute of Pathology, University Hospital Aachen, 52074 Aachen, Germany
| | - Danny Jonigk
- Institute of Pathology, University Hospital Aachen, 52074 Aachen, Germany; German Center for Lung Research, DZL, BREATH, 30625 Hanover, Germany
| | - Alexey Pryalukhin
- Institute of Clinical Pathology and Molecular Pathology, Wiener Neustadt State Hospital, 2700 Wiener Neustadt, Austria
| | - Fabian Mairinger
- Institute of Pathology, University Hospital Essen, 45147 Essen, Germany
| | - Franziska Klein
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany
| | - Anne Maria Schultheis
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany; Medical Faculty University of Cologne, 50937 Cologne, Germany
| | - Alexander Seper
- Institute of Clinical Pathology and Molecular Pathology, Wiener Neustadt State Hospital, 2700 Wiener Neustadt, Austria; Danube Private University, 3500 Krems an der Donau, Austria
| | - Wolfgang Hulla
- Institute of Clinical Pathology and Molecular Pathology, Wiener Neustadt State Hospital, 2700 Wiener Neustadt, Austria
| | - Johannes Brägelmann
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, 50937 Cologne, Germany; Mildred Scheel School of Oncology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, 50937 Cologne, Germany
| | - Sebastian Michels
- University of Cologne, Faculty of Medicine and University Hospital of Colone, Lung Cancer Group Cologne, Department I for Internal Medicine and Center for Integrated Oncology Aachen Bonn Cologne Dusseldorf, 50937 Cologne, Germany
| | - Sebastian Klein
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany; Medical Faculty University of Cologne, 50937 Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany; Medical Faculty University of Cologne, 50937 Cologne, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany; Medical Faculty University of Cologne, 50937 Cologne, Germany.
| | - Yuri Tolkach
- Institute of Pathology, University Hospital Cologne, 50937 Cologne, Germany; Medical Faculty University of Cologne, 50937 Cologne, Germany.
| |
Collapse
|
138
|
Comes MC, Fucci L, Strippoli S, Bove S, Cazzato G, Colangiuli C, Risi ID, Roma ID, Fanizzi A, Mele F, Ressa M, Saponaro C, Soranno C, Tinelli R, Guida M, Zito A, Massafra R. An artificial intelligence-based model exploiting H&E images to predict recurrence in negative sentinel lymph-node melanoma patients. J Transl Med 2024; 22:838. [PMID: 39267101 PMCID: PMC11391752 DOI: 10.1186/s12967-024-05629-2] [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: 03/07/2024] [Accepted: 08/18/2024] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND Risk stratification and treatment benefit prediction models are urgent to improve negative sentinel lymph node (SLN-) melanoma patient selection, thus avoiding costly and toxic treatments in patients at low risk of recurrence. To this end, the application of artificial intelligence (AI) could help clinicians to better calculate the recurrence risk and choose whether to perform adjuvant therapy. METHODS We made use of AI to predict recurrence-free status (RFS) within 2-years from diagnosis in 94 SLN- melanoma patients. In detail, we detected quantitative imaging information from H&E slides of a cohort of 71 SLN- melanoma patients, who registered at Istituto Tumori "Giovanni Paolo II" in Bari, Italy (investigational cohort, IC). For each slide, two expert pathologists firstly annotated two Regions of Interest (ROIs) containing tumor cells alone (TUMOR ROI) or with infiltrating cells (TUMOR + INF ROI). In correspondence of the two kinds of ROIs, two AI-based models were developed to extract information directly from the tiles in which each ROI was automatically divided. This information was then used to predict RFS. Performances of the models were computed according to a 5-fold cross validation scheme. We further validated the prediction power of the two models on an independent external validation cohort of 23 SLN- melanoma patients (validation cohort, VC). RESULTS The TUMOR ROIs have revealed more informative than the TUMOR + INF ROIs. An Area Under the Curve (AUC) value of 79.1% and 62.3%, a sensitivity value of 81.2% and 76.9%, a specificity value of 70.0% and 43.3%, an accuracy value of 73.2% and 53.4%, were achieved on the TUMOR and TUMOR + INF ROIs extracted for the IC cohort, respectively. An AUC value of 76.5% and 65.2%, a sensitivity value of 66.7% and 41.6%, a specificity value of 70.0% and 55.9%, an accuracy value of 70.0% and 56.5%, were achieved on the TUMOR and TUMOR + INF ROIs extracted for the VC cohort, respectively. CONCLUSIONS Our approach represents a first effort to develop a non-invasive prognostic method to better define the recurrence risk and improve the management of SLN- melanoma patients.
Collapse
Affiliation(s)
- Maria Colomba Comes
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Livia Fucci
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Sabino Strippoli
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Samantha Bove
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Gerardo Cazzato
- Dipartimento di Medicina di Precisione e Rigenerativa e Area Jonica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Carmen Colangiuli
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Ivana De Risi
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Ileana De Roma
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Annarita Fanizzi
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Fabio Mele
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Maurizio Ressa
- Unità Operativa Complessa di Chirurgica Plastica e Ricostruttiva, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Concetta Saponaro
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | | | - Rosita Tinelli
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Michele Guida
- Unità Operativa Tumori Rari e Melanoma, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy.
| | - Alfredo Zito
- Unità Operativa Complessa di Anatomia Patologica, I.R.C.C.S. Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Raffaella Massafra
- Laboratorio di Biostatistica e Bioinformatica, I.R.C.C.S. Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| |
Collapse
|
139
|
Pham JH, Thongprayoon C, Miao J, Suppadungsuk S, Koirala P, Craici IM, Cheungpasitporn W. Large language model triaging of simulated nephrology patient inbox messages. Front Artif Intell 2024; 7:1452469. [PMID: 39315245 PMCID: PMC11417033 DOI: 10.3389/frai.2024.1452469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/29/2024] [Indexed: 09/25/2024] Open
Abstract
Background Efficient triage of patient communications is crucial for timely medical attention and improved care. This study evaluates ChatGPT's accuracy in categorizing nephrology patient inbox messages, assessing its potential in outpatient settings. Methods One hundred and fifty simulated patient inbox messages were created based on cases typically encountered in everyday practice at a nephrology outpatient clinic. These messages were triaged as non-urgent, urgent, and emergent by two nephrologists. The messages were then submitted to ChatGPT-4 for independent triage into the same categories. The inquiry process was performed twice with a two-week period in between. ChatGPT responses were graded as correct (agreement with physicians), overestimation (higher priority), or underestimation (lower priority). Results In the first trial, ChatGPT correctly triaged 140 (93%) messages, overestimated the priority of 4 messages (3%), and underestimated the priority of 6 messages (4%). In the second trial, it correctly triaged 140 (93%) messages, overestimated the priority of 9 (6%), and underestimated the priority of 1 (1%). The accuracy did not depend on the urgency level of the message (p = 0.19). The internal agreement of ChatGPT responses was 92% with an intra-rater Kappa score of 0.88. Conclusion ChatGPT-4 demonstrated high accuracy in triaging nephrology patient messages, highlighting the potential for AI-driven triage systems to enhance operational efficiency and improve patient care in outpatient clinics.
Collapse
Affiliation(s)
- Justin H Pham
- Mayo Clinic College of Medicine and Science, Mayo Clinic, Rochester, MN, United States
| | - Charat Thongprayoon
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Jing Miao
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Supawadee Suppadungsuk
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
- Faculty of Medicine Ramathibodi Hospital, Chakri Naruebodindra Medical Institute, Mahidol University, Samut Prakan, Thailand
| | - Priscilla Koirala
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Iasmina M Craici
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | - Wisit Cheungpasitporn
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
140
|
Chi J, Xue Y, Zhou Y, Han T, Ning B, Cheng L, Xie H, Wang H, Wang W, Meng Q, Fan K, Gong F, Fan J, Jiang N, Liu Z, Pan K, Sun H, Zhang J, Zheng Q, Wang J, Su M, Song Y. Perovskite Probe-Based Machine Learning Imaging Model for Rapid Pathologic Diagnosis of Cancers. ACS NANO 2024; 18:24295-24305. [PMID: 39164203 DOI: 10.1021/acsnano.4c06351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Accurately distinguishing tumor cells from normal cells is a key issue in tumor diagnosis, evaluation, and treatment. Fluorescence-based immunohistochemistry as the standard method faces the inherent challenges of the heterogeneity of tumor cells and the lack of big data analysis of probing images. Here, we have demonstrated a machine learning-driven imaging method for rapid pathological diagnosis of five types of cancers (breast, colon, liver, lung, and stomach) using a perovskite nanocrystal probe. After conducting the bioanalysis of survivin expression in five different cancers, high-efficiency perovskite nanocrystal probes modified with the survivin antibody can recognize the cancer tissue section at the single cell level. The tumor to normal (T/N) ratio is 10.3-fold higher than that of a conventional fluorescent probe, which can successfully differentiate between tumors and adjacent normal tissues within 10 min. The features of the fluorescence intensity and pathological texture morphology have been extracted and analyzed from 1000 fluorescence images by machine learning. The final integrated decision model makes the area under the receiver operating characteristic curve (area under the curve) value of machine learning classification of breast, colon, liver, lung, and stomach above 90% while predicting the tumor organ of 92% of positive patients. This method demonstrates a high T/N ratio probe in the precise diagnosis of multiple cancers, which will be good for improving the accuracy of surgical resection and reducing cancer mortality.
Collapse
Affiliation(s)
- Jimei Chi
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yonggan Xue
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yinying Zhou
- School of Software, Northwestern Polytechnical University, Xi'an, 710129, China
| | - Teng Han
- Institute of Software, Chinese Academy of Sciences, Beijing, 100191, China
| | - Bobin Ning
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Lijun Cheng
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Hongfei Xie
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Huadong Wang
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Wenchen Wang
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qingyu Meng
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Kaijie Fan
- Department of Thoracic Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Fangming Gong
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Junzhen Fan
- Department of Pathology, the Third Medical Center, Chinese PLA General Hospital, Beijing, 100089, China
| | - Nan Jiang
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zheng Liu
- Senior Department of Orthopedics, the Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Ke Pan
- Institute of Hepato-Pancreato-Biliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hongyu Sun
- Department of Gastroenterology, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiajin Zhang
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qian Zheng
- Department of Thoracic Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiandong Wang
- Department of General Surgery, the First Medical Centre, Chinese PLA General Hospital, Beijing, 100853, China
| | - Meng Su
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Yanlin Song
- Key Laboratory of Green Printing, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences (ICCAS), Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Beijing 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
141
|
Choudhury D, Dolezal JM, Dyer E, Kochanny S, Ramesh S, Howard FM, Margalus JR, Schroeder A, Schulte J, Garassino MC, Kather JN, Pearson AT. Developing a low-cost, open-source, locally manufactured workstation and computational pipeline for automated histopathology evaluation using deep learning. EBioMedicine 2024; 107:105276. [PMID: 39197222 PMCID: PMC11399610 DOI: 10.1016/j.ebiom.2024.105276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Deployment and access to state-of-the-art precision medicine technologies remains a fundamental challenge in providing equitable global cancer care in low-resource settings. The expansion of digital pathology in recent years and its potential interface with diagnostic artificial intelligence algorithms provides an opportunity to democratize access to personalized medicine. Current digital pathology workstations, however, cost thousands to hundreds of thousands of dollars. As cancer incidence rises in many low- and middle-income countries, the validation and implementation of low-cost automated diagnostic tools will be crucial to helping healthcare providers manage the growing burden of cancer. METHODS Here we describe a low-cost ($230) workstation for digital slide capture and computational analysis composed of open-source components. We analyze the predictive performance of deep learning models when they are used to evaluate pathology images captured using this open-source workstation versus images captured using common, significantly more expensive hardware. Validation studies assessed model performance on three distinct datasets and predictive models: head and neck squamous cell carcinoma (HPV positive versus HPV negative), lung cancer (adenocarcinoma versus squamous cell carcinoma), and breast cancer (invasive ductal carcinoma versus invasive lobular carcinoma). FINDINGS When compared to traditional pathology image capture methods, low-cost digital slide capture and analysis with the open-source workstation, including the low-cost microscope device, was associated with model performance of comparable accuracy for breast, lung, and HNSCC classification. At the patient level of analysis, AUROC was 0.84 for HNSCC HPV status prediction, 1.0 for lung cancer subtype prediction, and 0.80 for breast cancer classification. INTERPRETATION Our ability to maintain model performance despite decreased image quality and low-power computational hardware demonstrates that it is feasible to massively reduce costs associated with deploying deep learning models for digital pathology applications. Improving access to cutting-edge diagnostic tools may provide an avenue for reducing disparities in cancer care between high- and low-income regions. FUNDING Funding for this project including personnel support was provided via grants from NIH/NCIR25-CA240134, NIH/NCIU01-CA243075, NIH/NIDCRR56-DE030958, NIH/NCIR01-CA276652, NIH/NCIK08-CA283261, NIH/NCI-SOAR25CA240134, SU2C (Stand Up to Cancer) Fanconi Anemia Research Fund - Farrah Fawcett Foundation Head and Neck Cancer Research Team Grant, and the European UnionHorizon Program (I3LUNG).
Collapse
Affiliation(s)
- Divya Choudhury
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | | | - Emma Dyer
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Sara Kochanny
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Siddhi Ramesh
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Frederick M Howard
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | | | | | - Jefree Schulte
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, USA
| | - Marina C Garassino
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Jakob N Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany; German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA.
| |
Collapse
|
142
|
Ahmad F, Muhmood T. Clinical translation of nanomedicine with integrated digital medicine and machine learning interventions. Colloids Surf B Biointerfaces 2024; 241:114041. [PMID: 38897022 DOI: 10.1016/j.colsurfb.2024.114041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Nanomaterials based therapeutics transform the ways of disease prevention, diagnosis and treatment with increasing sophistications in nanotechnology at a breakneck pace, but very few could reach to the clinic due to inconsistencies in preclinical studies followed by regulatory hinderances. To tackle this, integrating the nanomedicine discovery with digital medicine provide technologies as tools of specific biological activity measurement. Hence, overcome the redundancies in nanomedicine discovery by the on-site data acquisition and analytics through integrating intelligent sensors and artificial intelligence (AI) or machine learning (ML). Integrated AI/ML wearable sensors directly gather clinically relevant biochemical information from the subject's body and process data for physicians to make right clinical decision(s) in a time and cost-effective way. This review summarizes insights and recommend the infusion of actionable big data computation enabled sensors in burgeoning field of nanomedicine at academia, research institutes, and pharmaceutical industries, with a potential of clinical translation. Furthermore, many blind spots are present in modern clinically relevant computation, one of which could prevent ML-guided low-cost new nanomedicine development from being successfully translated into the clinic was also discussed.
Collapse
Affiliation(s)
- Farooq Ahmad
- State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources, College of Chemistry, Xinjiang University, Urumqi 830017, China.
| | - Tahir Muhmood
- International Iberian Nanotechnology Laboratory (INL), Avenida Mestre José Veiga, Braga 4715-330, Portugal.
| |
Collapse
|
143
|
Mansoor MA, Ibrahim AF, Kidd N. The Impact of Artificial Intelligence on Internal Medicine Physicians: A Survey of Procedural and Non-procedural Specialties. Cureus 2024; 16:e69121. [PMID: 39398704 PMCID: PMC11466679 DOI: 10.7759/cureus.69121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Artificial intelligence (AI) is increasingly being integrated into various aspects of healthcare, including internal medicine. However, the impact of AI on physicians across different internal medicine specialties remains unclear. This study assesses AI's adoption, utilization, and perceived impact among procedural and non-procedural internal medicine physicians. METHODS A comprehensive survey questionnaire was designed to cover current AI use, perceived impact on diagnostic accuracy, treatment decisions, patient outcomes, challenges, ethical concerns, and future expectations. The survey was distributed to a diverse sample of internal medicine physicians across various specialties, including procedural (e.g., interventional cardiology, gastroenterology) and non-procedural (e.g., endocrinology, rheumatology) fields. Responses were analyzed using descriptive statistics, chi-square tests, t-tests, and logistic regression. RESULTS The survey received responses from 22 internal medicine physicians, with 64% (n=14) representing procedural specialties and 36% (n=8) representing non-procedural specialties. Sixty-eight percent (n=15) of respondents reported using AI tools in their practice, with higher adoption rates among procedural specialties (n=11, 79%) compared to non-procedural specialties (n=4, 50%). Surveyed physicians reported that AI improved diagnostic accuracy (n=12, 80%), treatment decisions (n=10, 67%), and patient outcomes (n=13, 87%). However, 55% (n=12) of respondents expressed concerns about the interpretability and transparency of AI algorithms. Non-procedural specialists were more likely to perceive AI as a threat to their job security (n=3, 38%) than procedural specialists (n=3, 21%). The most common challenges to AI adoption were lack of training (n=16, 73%), cost (n=13, 59%), and data privacy concerns (n=11, 50%). CONCLUSION This study assesses the perceived impact of AI on internal medicine physicians, highlighting the differences between procedural and non-procedural specialties. The findings underscore the need for specialty-specific considerations in developing and implementing AI tools. While AI can potentially improve diagnostic accuracy, treatment decisions, and patient outcomes, addressing challenges such as lack of training, cost, and data privacy concerns is crucial for widespread adoption. Moreover, the study emphasizes the importance of ensuring the interpretability and transparency of AI algorithms to foster trust among physicians. As AI continues to evolve, it is essential to engage internal medicine physicians across specialties in the development process to create AI tools that effectively complement their expertise and improve patient care. Further research should focus on developing best practices for AI integration in internal medicine and evaluating the long-term impact on patient outcomes and healthcare systems.
Collapse
Affiliation(s)
- Masab A Mansoor
- Internal Medicine, Edward Via College of Osteopathic Medicine, Monroe, USA
| | - Andrew F Ibrahim
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, USA
| | - Nicholas Kidd
- Family Medicine, University of Virginia, Charlottesville, USA
| |
Collapse
|
144
|
Kamath CC, Wissler Gerdes EO, Barry BA, Minteer SA, Comfere NI, Peters MS, Wieland CN, Bews KA, Habermann EB, Ridgeway JL. Staff Experiences Transitioning to Digital Dermatopathology in a Tertiary Academic Medical Center: Lessons Learned From Implementation Science. MAYO CLINIC PROCEEDINGS. DIGITAL HEALTH 2024; 2:289-298. [PMID: 40206115 PMCID: PMC11975994 DOI: 10.1016/j.mcpdig.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Digital pathology (DP) transforms practice by replacing traditional glass slide review with digital whole slide images and workflows. Although digitization may improve accuracy and efficiency, transitioning to digital practice requires staff to learn new skills and adopt new ways of working and collaborating. In this study, we aimed to evaluate the experiences and perceptions of individuals involved in the day-to-day work of implementing DP in a tertiary academic medical center using Normalization Process Theory, a social theory that explains the processes by which innovations are operationalized and sustained in practice. Between September 2021 and June 2022, dermatopathologists, referring clinicians, and support staff at Mayo Clinic (Minnesota, Florida, and Arizona) participated in interviews (n=22) and completed surveys (n=34) concerning the transition. Normalization Process Theory informed the selection of validated survey items (Normalization Measure Development Questionnaire) and guided qualitative analysis. Participants reported high agreement with statements related to shared understanding and potential value of DP for workflow integration and working relationships. Qualitative themes reflecting the way organization and social context enable these processes were mapped onto implementation stages and related key activities. We found that earlier processes of implementation (understanding and working out participation) were better supported than later stages (doing it and reflecting on it). Our analysis helps identify targets for further intervention to hasten and help sustain implementation, including additional support in software and technological integration, workflows and work redesign, and regular monitoring and feedback systems. The use of implementation theory, such as Normalization Process Theory, may provide useful pointers to enable other similar digital system transition efforts.
Collapse
Affiliation(s)
- Celia C. Kamath
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Division of Health Care Delivery Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | | | - Barbara A. Barry
- Division of Health Care Delivery Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Sarah A. Minteer
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN
| | - Nneka I. Comfere
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Department of Dermatopathology, Mayo Clinic, Rochester, MN
| | - Margot S. Peters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Department of Dermatopathology, Mayo Clinic, Rochester, MN
| | - Carilyn N. Wieland
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
- Department of Dermatopathology, Mayo Clinic, Rochester, MN
| | - Katherine A. Bews
- Division of Health Care Delivery Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Elizabeth B. Habermann
- Division of Health Care Delivery Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Jennifer L. Ridgeway
- Division of Health Care Delivery Research, Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| |
Collapse
|
145
|
Chen Y, Xu Z, Ren S, Huang ZL, Wang Z. Image stitching algorithm for super-resolution localization microscopy combined with fluorescence noise prior. BIOMEDICAL OPTICS EXPRESS 2024; 15:5411-5428. [PMID: 39296408 PMCID: PMC11407243 DOI: 10.1364/boe.534658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 09/21/2024]
Abstract
Super-resolution panoramic pathological imaging provides a powerful tool for biologists to observe the ultrastructure of samples. Localization data can maintain the essential ultrastructural information of biological samples with a small storage space, and also provides a new opportunity for stitching super-resolution images. However, the existing image stitching methods based on localization data cannot accurately calculate the registration offset of sample regions with no or few structural points and thus lead to registration errors. Here, we proposed a stitching framework called PNanoStitcher. The framework fully utilizes the distribution characteristics of the background fluorescence noise in the stitching region and solves the stitching failure in sample regions with no or few structural points. We verified our method using both simulated and experimental datasets, and compared it with existing stitching methods. PNanoStitcher achieved superior stitching results on biological samples with no structural and few structural regions. The study provides an important driving force for the development of super-resolution digital pathology.
Collapse
Affiliation(s)
- Yanzhu Chen
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Zhiwang Xu
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Shijie Ren
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Zhen-Li Huang
- School of Biomedical Engineering, Hainan University, Sanya 572025, China
| | - Zhengxia Wang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| |
Collapse
|
146
|
Nardone V, Marmorino F, Germani MM, Cichowska-Cwalińska N, Menditti VS, Gallo P, Studiale V, Taravella A, Landi M, Reginelli A, Cappabianca S, Girnyi S, Cwalinski T, Boccardi V, Goyal A, Skokowski J, Oviedo RJ, Abou-Mrad A, Marano L. The Role of Artificial Intelligence on Tumor Boards: Perspectives from Surgeons, Medical Oncologists and Radiation Oncologists. Curr Oncol 2024; 31:4984-5007. [PMID: 39329997 PMCID: PMC11431448 DOI: 10.3390/curroncol31090369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The integration of multidisciplinary tumor boards (MTBs) is fundamental in delivering state-of-the-art cancer treatment, facilitating collaborative diagnosis and management by a diverse team of specialists. Despite the clear benefits in personalized patient care and improved outcomes, the increasing burden on MTBs due to rising cancer incidence and financial constraints necessitates innovative solutions. The advent of artificial intelligence (AI) in the medical field offers a promising avenue to support clinical decision-making. This review explores the perspectives of clinicians dedicated to the care of cancer patients-surgeons, medical oncologists, and radiation oncologists-on the application of AI within MTBs. Additionally, it examines the role of AI across various clinical specialties involved in cancer diagnosis and treatment. By analyzing both the potential and the challenges, this study underscores how AI can enhance multidisciplinary discussions and optimize treatment plans. The findings highlight the transformative role that AI may play in refining oncology care and sustaining the efficacy of MTBs amidst growing clinical demands.
Collapse
Affiliation(s)
- Valerio Nardone
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Federica Marmorino
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Marco Maria Germani
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | | | - Vittorio Salvatore Menditti
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Paolo Gallo
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Vittorio Studiale
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Ada Taravella
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Matteo Landi
- Unit of Medical Oncology 2, Azienda Ospedaliera Universitaria Pisana, 56126 Pisa, Italy; (F.M.); (M.M.G.); (V.S.); (A.T.); (M.L.)
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Alfonso Reginelli
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Salvatore Cappabianca
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, 80131 Naples, Italy; (V.N.); (V.S.M.); (P.G.); (A.R.); (S.C.)
| | - Sergii Girnyi
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
| | - Tomasz Cwalinski
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
| | - Virginia Boccardi
- Division of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, 06123 Perugia, Italy;
| | - Aman Goyal
- Adesh Institute of Medical Sciences and Research, Bathinda 151109, Punjab, India;
| | - Jaroslaw Skokowski
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
- Department of Medicine, Academy of Applied Medical and Social Sciences-AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland
| | - Rodolfo J. Oviedo
- Nacogdoches Medical Center, Nacogdoches, TX 75965, USA
- Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX 77021, USA
- College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA
| | - Adel Abou-Mrad
- Centre Hospitalier Universitaire d’Orléans, 45100 Orléans, France;
| | - Luigi Marano
- Department of General Surgery and Surgical Oncology, “Saint Wojciech” Hospital, “Nicolaus Copernicus” Health Center, 80-462 Gdańsk, Poland; (S.G.); (T.C.); (J.S.); (L.M.)
- Department of Medicine, Academy of Applied Medical and Social Sciences-AMiSNS: Akademia Medycznych I Spolecznych Nauk Stosowanych, 82-300 Elbląg, Poland
| |
Collapse
|
147
|
Kidder BL. Advanced image generation for cancer using diffusion models. Biol Methods Protoc 2024; 9:bpae062. [PMID: 39258159 PMCID: PMC11387006 DOI: 10.1093/biomethods/bpae062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/25/2024] [Accepted: 08/21/2024] [Indexed: 09/12/2024] Open
Abstract
Deep neural networks have significantly advanced the field of medical image analysis, yet their full potential is often limited by relatively small dataset sizes. Generative modeling, particularly through diffusion models, has unlocked remarkable capabilities in synthesizing photorealistic images, thereby broadening the scope of their application in medical imaging. This study specifically investigates the use of diffusion models to generate high-quality brain MRI scans, including those depicting low-grade gliomas, as well as contrast-enhanced spectral mammography (CESM) and chest and lung X-ray images. By leveraging the DreamBooth platform, we have successfully trained stable diffusion models utilizing text prompts alongside class and instance images to generate diverse medical images. This approach not only preserves patient anonymity but also substantially mitigates the risk of patient re-identification during data exchange for research purposes. To evaluate the quality of our synthesized images, we used the Fréchet inception distance metric, demonstrating high fidelity between the synthesized and real images. Our application of diffusion models effectively captures oncology-specific attributes across different imaging modalities, establishing a robust framework that integrates artificial intelligence in the generation of oncological medical imagery.
Collapse
Affiliation(s)
- Benjamin L Kidder
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, 48201, United States
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, United States
| |
Collapse
|
148
|
Mubarak M, Rashid R, Sapna F, Shakeel S. Expanding role and scope of artificial intelligence in the field of gastrointestinal pathology. Artif Intell Gastroenterol 2024; 5:91550. [DOI: 10.35712/aig.v5.i2.91550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/06/2024] [Accepted: 07/29/2024] [Indexed: 08/08/2024] Open
Abstract
Digital pathology (DP) and its subsidiaries including artificial intelligence (AI) are rapidly making inroads into the area of diagnostic anatomic pathology (AP) including gastrointestinal (GI) pathology. It is poised to revolutionize the field of diagnostic AP. Historically, AP has been slow to adopt digital technology, but this is changing rapidly, with many centers worldwide transitioning to DP. Coupled with advanced techniques of AI such as deep learning and machine learning, DP is likely to transform histopathology from a subjective field to an objective, efficient, and transparent discipline. AI is increasingly integrated into GI pathology, offering numerous advancements and improvements in overall diagnostic accuracy, efficiency, and patient care. Specifically, AI in GI pathology enhances diagnostic accuracy, streamlines workflows, provides predictive insights, integrates multimodal data, supports research, and aids in education and training, ultimately improving patient care and outcomes. This review summarized the latest developments in the role and scope of AI in AP with a focus on GI pathology. The main aim was to provide updates and create awareness among the pathology community.
Collapse
Affiliation(s)
- Muhammed Mubarak
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Rahma Rashid
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| | - Fnu Sapna
- Department of Pathology, Montefiore Medical Center, The University Hospital for Albert Einstein School of Medicine, Bronx, NY 10461, United States
| | - Shaheera Shakeel
- Department of Histopathology, Sindh Institute of Urology and Transplantation, Karachi 74200, Sindh, Pakistan
| |
Collapse
|
149
|
Botros M, de Boer OJ, Cardenas B, Bekkers EJ, Jansen M, van der Wel MJ, Sánchez CI, Meijer SL. Deep Learning for Histopathological Assessment of Esophageal Adenocarcinoma Precursor Lesions. Mod Pathol 2024; 37:100531. [PMID: 38830407 DOI: 10.1016/j.modpat.2024.100531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/06/2024] [Accepted: 05/28/2024] [Indexed: 06/05/2024]
Abstract
Histopathological assessment of esophageal biopsies is a key part in the management of patients with Barrett esophagus (BE) but prone to observer variability and reliable diagnostic methods are needed. Artificial intelligence (AI) is emerging as a powerful tool for aided diagnosis but often relies on abstract test and validation sets while real-world behavior is unknown. In this study, we developed a 2-stage AI system for histopathological assessment of BE-related dysplasia using deep learning to enhance the efficiency and accuracy of the pathology workflow. The AI system was developed and trained on 290 whole-slide images (WSIs) that were annotated at glandular and tissue levels. The system was designed to identify individual glands, grade dysplasia, and assign a WSI-level diagnosis. The proposed method was evaluated by comparing the performance of our AI system with that of a large international and heterogeneous group of 55 gastrointestinal pathologists assessing 55 digitized biopsies spanning the complete spectrum of BE-related dysplasia. The AI system correctly graded 76.4% of the WSIs, surpassing the performance of 53 out of the 55 participating pathologists. Furthermore, the receiver-operating characteristic analysis showed that the system's ability to predict the absence (nondysplastic BE) versus the presence of any dysplasia was with an area under the curve of 0.94 and a sensitivity of 0.92 at a specificity of 0.94. These findings demonstrate that this AI system has the potential to assist pathologists in assessment of BE-related dysplasia. The system's outputs could provide a reliable and consistent secondary diagnosis in challenging cases or be used for triaging low-risk nondysplastic biopsies, thereby reducing the workload of pathologists and increasing throughput.
Collapse
Affiliation(s)
- Michel Botros
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Quantitative Healthcare Analysis Group, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands; Amsterdam Machine Learning Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Onno J de Boer
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Bryan Cardenas
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Amsterdam Machine Learning Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik J Bekkers
- Amsterdam Machine Learning Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Marnix Jansen
- Research Department of Pathology, Cancer Institute, University College London, London, United Kingdom
| | - Myrtle J van der Wel
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Clara I Sánchez
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, Amsterdam, The Netherlands; Quantitative Healthcare Analysis Group, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Sybren L Meijer
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, The Netherlands.
| |
Collapse
|
150
|
Cai C, Zhou Y, Jiao Y, Li L, Xu J. Prognostic Analysis Combining Histopathological Features and Clinical Information to Predict Colorectal Cancer Survival from Whole-Slide Images. Dig Dis Sci 2024; 69:2985-2995. [PMID: 38837111 DOI: 10.1007/s10620-024-08501-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant tumor within the digestive tract with both a high incidence rate and mortality. Early detection and intervention could improve patient clinical outcomes and survival. METHODS This study computationally investigates a set of prognostic tissue and cell features from diagnostic tissue slides. With the combination of clinical prognostic variables, the pathological image features could predict the prognosis in CRC patients. Our CRC prognosis prediction pipeline sequentially consisted of three modules: (1) A MultiTissue Net to delineate outlines of different tissue types within the WSI of CRC for further ROI selection by pathologists. (2) Development of three-level quantitative image metrics related to tissue compositions, cell shape, and hidden features from a deep network. (3) Fusion of multi-level features to build a prognostic CRC model for predicting survival for CRC. RESULTS Experimental results suggest that each group of features has a particular relationship with the prognosis of patients in the independent test set. In the fusion features combination experiment, the accuracy rate of predicting patients' prognosis and survival status is 81.52%, and the AUC value is 0.77. CONCLUSION This paper constructs a model that can predict the postoperative survival of patients by using image features and clinical information. Some features were found to be associated with the prognosis and survival of patients.
Collapse
Affiliation(s)
- Chengfei Cai
- School of Automation, Nanjing University of Information Science and Technology, Nanjing, 210044, China.
- College of Information Engineering, Taizhou University, Taizhou, 225300, China.
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, 210044, China.
| | - Yangshu Zhou
- Department of Pathology, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280, China
| | - Yiping Jiao
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| | - Liang Li
- Department of Pathology, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Jun Xu
- Institute for AI in Medicine, School of Artificial Intelligence, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| |
Collapse
|