1
|
Lee DN, Li Y, Olsson LT, Hamilton AM, Calhoun BC, Hoadley KA, Marron JS, Troester MA. Image analysis-based identification of high risk ER-positive, HER2-negative breast cancers. Breast Cancer Res 2024; 26:177. [PMID: 39633505 PMCID: PMC11616316 DOI: 10.1186/s13058-024-01915-5] [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: 02/16/2024] [Accepted: 09/12/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Breast cancer subtypes Luminal A and Luminal B are classified by the expression of PAM50 genes and may benefit from different treatment strategies. Machine learning models based on H&E images may contain features associated with subtype, allowing early identification of tumors with higher risk of recurrence. METHODS H&E images (n = 630 ER+/HER2-breast cancers) were pixel-level segmented into epithelium and stroma. Convolutional neural network and multiple instance learning were used to extract image features from original and segmented images. Patient-level classification models were trained to discriminate Luminal A versus B image features in tenfold cross-validation, with or without grade adjustment. The best-performing visual classifier was incorporated into envisioned diagnostic protocols as an alternative to genomic testing (PAM50). The protocols were then compared in time-to-recurrence models. RESULTS Among ER+/HER2-tumors, the image-based protocol differentiated recurrence times with a hazard ratio (HR) of 2.81 (95% CI: 1.73-4.56), which was similar to the HR for PAM50 (2.66, 95% CI: 1.65-4.28). Grade adjustment did not improve subtype prediction accuracy, but did help balance sensitivity and specificity. Among high grade participants, sensitivity and specificity (0.734 and 0.474, respectively) became more similar (0.732 and 0.624, respectively) in grade-adjusted models. The original and epithelium-specific images had similar performance and highest accuracy, followed by stroma or binarized images showing only the epithelial-stromal interface. CONCLUSIONS Given low rates of genomic testing uptake nationally, image-based methods may help identify ER+/HER2-patients who could benefit from testing.
Collapse
Affiliation(s)
- Dong Neuck Lee
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Yao Li
- Department of Statistics and Operations Research, University of North Carolina, Chapel Hill, NC, USA
| | - Linnea T Olsson
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Alina M Hamilton
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Benjamin C Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | | | - J S Marron
- Department of Statistics and Operations Research, University of North Carolina, Chapel Hill, NC, USA.
| | - Melissa A Troester
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Tafavvoghi M, Bongo LA, Shvetsov N, Busund LTR, Møllersen K. Publicly available datasets of breast histopathology H&E whole-slide images: A scoping review. J Pathol Inform 2024; 15:100363. [PMID: 38405160 PMCID: PMC10884505 DOI: 10.1016/j.jpi.2024.100363] [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: 09/14/2023] [Revised: 11/24/2023] [Accepted: 01/23/2024] [Indexed: 02/27/2024] Open
Abstract
Advancements in digital pathology and computing resources have made a significant impact in the field of computational pathology for breast cancer diagnosis and treatment. However, access to high-quality labeled histopathological images of breast cancer is a big challenge that limits the development of accurate and robust deep learning models. In this scoping review, we identified the publicly available datasets of breast H&E-stained whole-slide images (WSIs) that can be used to develop deep learning algorithms. We systematically searched 9 scientific literature databases and 9 research data repositories and found 17 publicly available datasets containing 10 385 H&E WSIs of breast cancer. Moreover, we reported image metadata and characteristics for each dataset to assist researchers in selecting proper datasets for specific tasks in breast cancer computational pathology. In addition, we compiled 2 lists of breast H&E patches and private datasets as supplementary resources for researchers. Notably, only 28% of the included articles utilized multiple datasets, and only 14% used an external validation set, suggesting that the performance of other developed models may be susceptible to overestimation. The TCGA-BRCA was used in 52% of the selected studies. This dataset has a considerable selection bias that can impact the robustness and generalizability of the trained algorithms. There is also a lack of consistent metadata reporting of breast WSI datasets that can be an issue in developing accurate deep learning models, indicating the necessity of establishing explicit guidelines for documenting breast WSI dataset characteristics and metadata.
Collapse
Affiliation(s)
- Masoud Tafavvoghi
- Department of Community Medicine, Uit The Arctic University of Norway, Tromsø, Norway
| | - Lars Ailo Bongo
- Department of Computer Science, Uit The Arctic University of Norway, Tromsø, Norway
| | - Nikita Shvetsov
- Department of Computer Science, Uit The Arctic University of Norway, Tromsø, Norway
| | | | - Kajsa Møllersen
- Department of Community Medicine, Uit The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
3
|
Bergstrom EN, Abbasi A, Díaz-Gay M, Galland L, Ladoire S, Lippman SM, Alexandrov LB. Deep Learning Artificial Intelligence Predicts Homologous Recombination Deficiency and Platinum Response From Histologic Slides. J Clin Oncol 2024; 42:3550-3560. [PMID: 39083703 PMCID: PMC11469627 DOI: 10.1200/jco.23.02641] [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: 12/08/2023] [Revised: 04/23/2024] [Accepted: 05/28/2024] [Indexed: 08/02/2024] Open
Abstract
PURPOSE Cancers with homologous recombination deficiency (HRD) can benefit from platinum salts and poly(ADP-ribose) polymerase inhibitors. Standard diagnostic tests for detecting HRD require molecular profiling, which is not universally available. METHODS We trained DeepHRD, a deep learning platform for predicting HRD from hematoxylin and eosin (H&E)-stained histopathological slides, using primary breast (n = 1,008) and ovarian (n = 459) cancers from The Cancer Genome Atlas (TCGA). DeepHRD was compared with four standard HRD molecular tests using breast (n = 349) and ovarian (n = 141) cancers from multiple independent data sets, including platinum-treated clinical cohorts with RECIST progression-free survival (PFS), complete response (CR), and overall survival (OS) endpoints. RESULTS DeepHRD predicted HRD from held-out H&E-stained breast cancer slides in TCGA with an AUC of 0.81 (95% CI, 0.77 to 0.85). This performance was confirmed in two independent primary breast cancer cohorts (AUC, 0.76 [95% CI, 0.71 to 0.82]). In an external platinum-treated metastatic breast cancer cohort, samples predicted as HRD had higher complete CR (AUC, 0.76 [95% CI, 0.54 to 0.93]) with 3.7-fold increase in median PFS (14.4 v 3.9 months; P = .0019) and hazard ratio (HR) of 0.45 (P = .0047). There were no significant differences in nonplatinum treatment outcome by predicted HRD status in three breast cancer cohorts, including CR (AUC, 0.39) and PFS (HR, 0.98, P = .95) in taxane-treated metastatic breast cancer. Through transfer learning to high-grade serous ovarian cancer, DeepHRD-predicted HRD samples had better OS after first-line (HR, 0.46; P = .030) and neoadjuvant (HR, 0.49; P = .015) platinum therapy in two cohorts. CONCLUSION DeepHRD can predict HRD in breast and ovarian cancers directly from routine H&E slides across multiple external cohorts, slide scanners, and tissue fixation variables. When compared with molecular testing, DeepHRD classified 1.8- to 3.1-fold more patients with HRD, which exhibited better OS in high-grade serous ovarian cancer and platinum-specific PFS in metastatic breast cancer.
Collapse
Affiliation(s)
- Erik N. Bergstrom
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Ammal Abbasi
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Marcos Díaz-Gay
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
| | - Loïck Galland
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
- Platform of Transfer in Biological Oncology, Centre Georges-François Leclerc, Dijon, France
- University of Burgundy-Franche Comté, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France
- Platform of Transfer in Biological Oncology, Centre Georges-François Leclerc, Dijon, France
- University of Burgundy-Franche Comté, France
- Centre de Recherche INSERM LNC-UMR1231, Dijon, France
| | | | - Ludmil B. Alexandrov
- Moores Cancer Center, UC San Diego, La Jolla, CA
- Department of Cellular and Molecular Medicine, UC San Diego, La Jolla, CA
- Department of Bioengineering, UC San Diego, La Jolla, CA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA
| |
Collapse
|
4
|
Wang X, Zhao J, Marostica E, Yuan W, Jin J, Zhang J, Li R, Tang H, Wang K, Li Y, Wang F, Peng Y, Zhu J, Zhang J, Jackson CR, Zhang J, Dillon D, Lin NU, Sholl L, Denize T, Meredith D, Ligon KL, Signoretti S, Ogino S, Golden JA, Nasrallah MP, Han X, Yang S, Yu KH. A pathology foundation model for cancer diagnosis and prognosis prediction. Nature 2024; 634:970-978. [PMID: 39232164 DOI: 10.1038/s41586-024-07894-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 08/01/2024] [Indexed: 09/06/2024]
Abstract
Histopathology image evaluation is indispensable for cancer diagnoses and subtype classification. Standard artificial intelligence methods for histopathology image analyses have focused on optimizing specialized models for each diagnostic task1,2. Although such methods have achieved some success, they often have limited generalizability to images generated by different digitization protocols or samples collected from different populations3. Here, to address this challenge, we devised the Clinical Histopathology Imaging Evaluation Foundation (CHIEF) model, a general-purpose weakly supervised machine learning framework to extract pathology imaging features for systematic cancer evaluation. CHIEF leverages two complementary pretraining methods to extract diverse pathology representations: unsupervised pretraining for tile-level feature identification and weakly supervised pretraining for whole-slide pattern recognition. We developed CHIEF using 60,530 whole-slide images spanning 19 anatomical sites. Through pretraining on 44 terabytes of high-resolution pathology imaging datasets, CHIEF extracted microscopic representations useful for cancer cell detection, tumour origin identification, molecular profile characterization and prognostic prediction. We successfully validated CHIEF using 19,491 whole-slide images from 32 independent slide sets collected from 24 hospitals and cohorts internationally. Overall, CHIEF outperformed the state-of-the-art deep learning methods by up to 36.1%, showing its ability to address domain shifts observed in samples from diverse populations and processed by different slide preparation methods. CHIEF provides a generalizable foundation for efficient digital pathology evaluation for patients with cancer.
Collapse
Affiliation(s)
- Xiyue Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Junhan Zhao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA, USA
| | - Wei Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Jietian Jin
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiayu Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongping Tang
- Department of Pathology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Kanran Wang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yu Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Fang Wang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yulong Peng
- Department of Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junyou Zhu
- Department of Burn, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Christopher R Jackson
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hummelstown, PA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Meredith
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuji Ogino
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sen Yang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
5
|
Hölscher DL, Bülow RD. Decoding pathology: the role of computational pathology in research and diagnostics. Pflugers Arch 2024:10.1007/s00424-024-03002-2. [PMID: 39095655 DOI: 10.1007/s00424-024-03002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/25/2024] [Accepted: 07/25/2024] [Indexed: 08/04/2024]
Abstract
Traditional histopathology, characterized by manual quantifications and assessments, faces challenges such as low-throughput and inter-observer variability that hinder the introduction of precision medicine in pathology diagnostics and research. The advent of digital pathology allowed the introduction of computational pathology, a discipline that leverages computational methods, especially based on deep learning (DL) techniques, to analyze histopathology specimens. A growing body of research shows impressive performances of DL-based models in pathology for a multitude of tasks, such as mutation prediction, large-scale pathomics analyses, or prognosis prediction. New approaches integrate multimodal data sources and increasingly rely on multi-purpose foundation models. This review provides an introductory overview of advancements in computational pathology and discusses their implications for the future of histopathology in research and diagnostics.
Collapse
Affiliation(s)
- David L Hölscher
- Department for Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
- Institute for Pathology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Roman D Bülow
- Institute for Pathology, RWTH Aachen University Hospital, Pauwelsstraße 30, 52074, Aachen, Germany.
| |
Collapse
|
6
|
Zhuo X, Deng H, Qiu M, Qiu X. Pathomic model based on histopathological features and machine learning to predict IDO1 status and its association with breast cancer prognosis. Breast Cancer Res Treat 2024; 207:151-165. [PMID: 38780888 PMCID: PMC11230954 DOI: 10.1007/s10549-024-07350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE To establish a pathomic model using histopathological image features for predicting indoleamine 2,3-dioxygenase 1 (IDO1) status and its relationship with overall survival (OS) in breast cancer. METHODS A pathomic model was constructed using machine learning and histopathological images obtained from The Cancer Genome Atlas database to predict IDO1 expression. The model performance was evaluated based on the area under the curve, calibration curve, and decision curve analysis (DCA). Prediction scores (PSes) were generated from the model and applied to divide the patients into two groups. Survival outcomes, gene set enrichment, immune microenvironment, and tumor mutations were assessed between the two groups. RESULTS Survival analysis followed by multivariate correction revealed that high IDO1 is a protective factor for OS. Further, the model was calibrated, and it exhibited good discrimination. Additionally, the DCA showed that the proposed model provided a good clinical net benefit. The Kaplan-Meier analysis revealed a positive correlation between high PS and improved OS. Univariate and multivariate Cox regression analyses demonstrated that PS is an independent protective factor for OS. Moreover, differentially expressed genes were enriched in various essential biological processes, including extracellular matrix receptor interaction, angiogenesis, transforming growth factor β signaling, epithelial mesenchymal transition, cell junction, tryptophan metabolism, and heme metabolic processes. PS was positively correlated with M1 macrophages, CD8 + T cells, T follicular helper cells, and tumor mutational burden. CONCLUSION These results indicate the potential ability of the proposed pathomic model to predict IDO1 status and the OS of breast cancer patients.
Collapse
Affiliation(s)
- Xiaohua Zhuo
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, China
| | - Hailong Deng
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, China
| | - Mingzhu Qiu
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, China
| | - Xiaoming Qiu
- Department of Pathology, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian, China.
| |
Collapse
|
7
|
Unger M, Kather JN. A systematic analysis of deep learning in genomics and histopathology for precision oncology. BMC Med Genomics 2024; 17:48. [PMID: 38317154 PMCID: PMC10845449 DOI: 10.1186/s12920-024-01796-9] [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: 08/03/2023] [Accepted: 01/02/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Digitized histopathological tissue slides and genomics profiling data are available for many patients with solid tumors. In the last 5 years, Deep Learning (DL) has been broadly used to extract clinically actionable information and biological knowledge from pathology slides and genomic data in cancer. In addition, a number of recent studies have introduced multimodal DL models designed to simultaneously process both images from pathology slides and genomic data as inputs. By comparing patterns from one data modality with those in another, multimodal DL models are capable of achieving higher performance compared to their unimodal counterparts. However, the application of these methodologies across various tumor entities and clinical scenarios lacks consistency. METHODS Here, we present a systematic survey of the academic literature from 2010 to November 2023, aiming to quantify the application of DL for pathology, genomics, and the combined use of both data types. After filtering 3048 publications, our search identified 534 relevant articles which then were evaluated by basic (diagnosis, grading, subtyping) and advanced (mutation, drug response and survival prediction) application types, publication year and addressed cancer tissue. RESULTS Our analysis reveals a predominant application of DL in pathology compared to genomics. However, there is a notable surge in DL incorporation within both domains. Furthermore, while DL applied to pathology primarily targets the identification of histology-specific patterns in individual tissues, DL in genomics is more commonly used in a pan-cancer context. Multimodal DL, on the contrary, remains a niche topic, evidenced by a limited number of publications, primarily focusing on prognosis predictions. CONCLUSION In summary, our quantitative analysis indicates that DL not only has a well-established role in histopathology but is also being successfully integrated into both genomic and multimodal applications. In addition, there is considerable potential in multimodal DL for harnessing further advanced tasks, such as predicting drug response. Nevertheless, this review also underlines the need for further research to bridge the existing gaps in these fields.
Collapse
Affiliation(s)
- Michaela Unger
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
8
|
Mohammed Zaidh S, Aher KB, Bhavar GB, Irfan N, Ahmed HN, Ismail Y. Genes adaptability and NOL6 protein inhibition studies of fabricated flavan-3-ols lead skeleton intended to treat breast carcinoma. Int J Biol Macromol 2024; 258:127661. [PMID: 37898257 DOI: 10.1016/j.ijbiomac.2023.127661] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/10/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Breast cancer invasive 2.3 million women worldly and second prominent factor of cancer-related mortality. Finding a new site-specific and safe small molecule is a current need in this field. With the aid of deep learning Algorithms, we analyzed the published big database from cancer CBioportal to find the best target protein. Further, Multi-omics analysis such as enrichment analysis, scores of molecular, RNA biological function at a cellular level, and protein domain were obtained and matched to find the better hit molecules. The gene analysis output shows nucleolar protein 6 plays a significant responsibility in breast carcinoma and 354 natural and synthetic lead molecules are docked inside the active site. Docking result gave the output hit molecule falavan-3-ols with a binding score of -5.325 (Kcal/mol) and interaction analysis illustrates, 13 active amino acids favoring the binding interaction with functional groups of the hit molecule compared to the standard molecule Abemacilib (-2.857 (Kcal/mol)). Best docked complex of flavan-3-ols and NOL6 protein subjected to dynamic simulation 100 ns to study the stability. The results proved that π-π stacked, carbon‑hydrogen and electrostatic interactions are stable throughout the 100 ns simulation. The overall results conclude the hit molecule flavan-3-ol will be a safe and potent lead molecule to generate and treat breast carcinoma patients.
Collapse
Affiliation(s)
- S Mohammed Zaidh
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai 600048, India
| | - Kiran Balasaheb Aher
- Department of Pharmaceutical Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| | - Girija Balasaheb Bhavar
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra 424001, India
| | - N Irfan
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai 600048, India.
| | - Haja Nazeer Ahmed
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai 600048, India
| | - Y Ismail
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai 600048, India
| |
Collapse
|
9
|
Wu X, Li W, Tu H. Big data and artificial intelligence in cancer research. Trends Cancer 2024; 10:147-160. [PMID: 37977902 DOI: 10.1016/j.trecan.2023.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
The field of oncology has witnessed an extraordinary surge in the application of big data and artificial intelligence (AI). AI development has made multiscale and multimodal data fusion and analysis possible. A new era of extracting information from complex big data is rapidly evolving. However, challenges related to efficient data curation, in-depth analysis, and utilization remain. We provide a comprehensive overview of the current state of the art in big data and computational analysis, highlighting key applications, challenges, and future opportunities in cancer research. By sketching the current landscape, we seek to foster a deeper understanding and facilitate the advancement of big data utilization in oncology, call for interdisciplinary collaborations, ultimately contributing to improved patient outcomes and a profound understanding of cancer.
Collapse
Affiliation(s)
- Xifeng Wu
- Department of Big Data in Health Science, School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; National Institute for Data Science in Health and Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Wenyuan Li
- Department of Big Data in Health Science, School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Huakang Tu
- Department of Big Data in Health Science, School of Public Health, Center of Clinical Big Data and Analytics of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
10
|
Yang Q, Deng S, Preibsch H, Schade T, Koch A, Berezhnoy G, Zizmare L, Fischer A, Gückel B, Staebler A, Hartkopf AD, Pichler BJ, la Fougère C, Hahn M, Bonzheim I, Nikolaou K, Trautwein C. Image-guided metabolomics and transcriptomics reveal tumour heterogeneity in luminal A and B human breast cancer beyond glucose tracer uptake. Clin Transl Med 2024; 14:e1550. [PMID: 38332687 PMCID: PMC10853679 DOI: 10.1002/ctm2.1550] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Breast cancer is a metabolically heterogeneous disease, and although the concept of heterogeneous cancer metabolism is known, its precise role in human breast cancer is yet to be fully elucidated. METHODS We investigated in an explorative approach a cohort of 42 primary mamma carcinoma patients with positron emission tomography/magnetic resonance imaging (PET/MR) prior to surgery, followed by histopathology and molecular diagnosis. From a subset of patients, which showed high metabolic heterogeneity based on tracer uptake and pathology classification, tumour centre and periphery specimen tissue samples were further investigated by a targeted breast cancer gene expression panel and quantitative metabolomics by nuclear magnetic resonance (NMR) spectroscopy. All data were analysed in a combinatory approach. RESULTS [18 F]FDG (2-deoxy-2-[fluorine-18]fluoro-d-glucose) tracer uptake confirmed dominance of glucose metabolism in the breast tumour centre, with lower levels in the periphery. Additionally, we observed differences in lipid and proliferation related genes between luminal A and B subtypes in the centre and periphery. Tumour periphery showed elevated acetate levels and enrichment in lipid metabolic pathways genes especially in luminal B. Furthermore, serine was increased in the periphery and higher expression of thymidylate synthase (TYMS) indicated one-carbon metabolism increased in tumour periphery. The overall metabolic activity based on [18 F]FDG uptake of luminal B subtype was higher than that of luminal A and the difference between the periphery and centre increased with tumour grade. CONCLUSION Our analysis indicates variations in metabolism among different breast cancer subtypes and sampling locations which details the heterogeneity of the breast tumours. Correlation analysis of [18 F]FDG tracer uptake, transcriptome and tumour metabolites like acetate and serine facilitate the search for new candidates for metabolic tracers and permit distinguishing luminal A and B. This knowledge may help to differentiate subtypes preclinically or to provide patients guide for neoadjuvant therapy and optimised surgical protocols based on individual tumour metabolism.
Collapse
Affiliation(s)
- Qianlu Yang
- Department of Preclinical Imaging and RadiopharmacyWerner Siemens Imaging CenterUniversity Hospital TuebingenTuebingenGermany
| | - Sisi Deng
- Department of Preclinical Imaging and RadiopharmacyWerner Siemens Imaging CenterUniversity Hospital TuebingenTuebingenGermany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
| | - Heike Preibsch
- Department of Diagnostic and Interventional RadiologyUniversity Hospital TuebingenTuebingenGermany
| | - Tim‐Colin Schade
- Department of Pathology and NeuropathologyUniversity Hospital TuebingenTuebingenGermany
| | - André Koch
- Department of Women's HealthUniversity Hospital TuebingenTuebingenGermany
| | - Georgy Berezhnoy
- Department of Preclinical Imaging and RadiopharmacyWerner Siemens Imaging CenterUniversity Hospital TuebingenTuebingenGermany
| | - Laimdota Zizmare
- Department of Preclinical Imaging and RadiopharmacyWerner Siemens Imaging CenterUniversity Hospital TuebingenTuebingenGermany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
| | - Anna Fischer
- Department of Pathology and NeuropathologyUniversity Hospital TuebingenTuebingenGermany
| | - Brigitte Gückel
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
- Department of Diagnostic and Interventional RadiologyUniversity Hospital TuebingenTuebingenGermany
| | - Annette Staebler
- Department of Pathology and NeuropathologyUniversity Hospital TuebingenTuebingenGermany
| | | | - Bernd J. Pichler
- Department of Preclinical Imaging and RadiopharmacyWerner Siemens Imaging CenterUniversity Hospital TuebingenTuebingenGermany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
- German Cancer Research CenterGerman Cancer Consortium DKTKPartner Site TuebingenTuebingenGermany
| | - Christian la Fougère
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
- German Cancer Research CenterGerman Cancer Consortium DKTKPartner Site TuebingenTuebingenGermany
- Department of Nuclear Medicine and Clinical Molecular ImagingUniversity Hospital TuebingenTuebingenGermany
| | - Markus Hahn
- Department of Women's HealthUniversity Hospital TuebingenTuebingenGermany
| | - Irina Bonzheim
- Department of Pathology and NeuropathologyUniversity Hospital TuebingenTuebingenGermany
| | - Konstantin Nikolaou
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
- Department of Diagnostic and Interventional RadiologyUniversity Hospital TuebingenTuebingenGermany
- German Cancer Research CenterGerman Cancer Consortium DKTKPartner Site TuebingenTuebingenGermany
| | - Christoph Trautwein
- Department of Preclinical Imaging and RadiopharmacyWerner Siemens Imaging CenterUniversity Hospital TuebingenTuebingenGermany
- Cluster of Excellence iFIT (EXC 2180) “Image Guided and Functionally Instructed Tumor Therapies”University of TuebingenTuebingenGermany
| |
Collapse
|
11
|
Kohane IS, Churchill S, Tan ALM, Vella M, Perry CL. The digital-physical divide for pathology research. Lancet Digit Health 2023; 5:e859-e861. [PMID: 38000870 DOI: 10.1016/s2589-7500(23)00184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/01/2023] [Accepted: 09/13/2023] [Indexed: 11/26/2023]
Affiliation(s)
- Isaac S Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA.
| | - Susanne Churchill
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Amelia Li Min Tan
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Margaret Vella
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Cassandra L Perry
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Nasrallah MP, Zhao J, Tsai CC, Meredith D, Marostica E, Ligon KL, Golden JA, Yu KH. Machine learning for cryosection pathology predicts the 2021 WHO classification of glioma. MED 2023; 4:526-540.e4. [PMID: 37421953 PMCID: PMC10527821 DOI: 10.1016/j.medj.2023.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/17/2023] [Accepted: 06/06/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Timely and accurate intraoperative cryosection evaluations remain the gold standard for guiding surgical treatments for gliomas. However, the tissue-freezing process often generates artifacts that make histologic interpretation difficult. In addition, the 2021 WHO Classification of Tumors of the Central Nervous System incorporates molecular profiles in the diagnostic categories, so standard visual evaluation of cryosections alone cannot completely inform diagnoses based on the new classification system. METHODS To address these challenges, we develop the context-aware Cryosection Histopathology Assessment and Review Machine (CHARM) using samples from 1,524 glioma patients from three different patient populations to systematically analyze cryosection slides. FINDINGS Our CHARM models successfully identified malignant cells (AUROC = 0.98 ± 0.01 in the independent validation cohort), distinguished isocitrate dehydrogenase (IDH)-mutant tumors from wild type (AUROC = 0.79-0.82), classified three major types of molecularly defined gliomas (AUROC = 0.88-0.93), and identified the most prevalent subtypes of IDH-mutant tumors (AUROC = 0.89-0.97). CHARM further predicts clinically important genetic alterations in low-grade glioma, including ATRX, TP53, and CIC mutations, CDKN2A/B homozygous deletion, and 1p/19q codeletion via cryosection images. CONCLUSIONS Our approaches accommodate the evolving diagnostic criteria informed by molecular studies, provide real-time clinical decision support, and will democratize accurate cryosection diagnoses. FUNDING Supported in part by the National Institute of General Medical Sciences grant R35GM142879, the Google Research Scholar Award, the Blavatnik Center for Computational Biomedicine Award, the Partners' Innovation Discovery Grant, and the Schlager Family Award for Early Stage Digital Health Innovations.
Collapse
Affiliation(s)
- MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhan Zhao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Cheng Che Tsai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - David Meredith
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA 02139, USA
| | - Keith L Ligon
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Ochoa S, Hernández-Lemus E. Molecular mechanisms of multi-omic regulation in breast cancer. Front Oncol 2023; 13:1148861. [PMID: 37564937 PMCID: PMC10411627 DOI: 10.3389/fonc.2023.1148861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023] Open
Abstract
Breast cancer is a complex disease that is influenced by the concurrent influence of multiple genetic and environmental factors. Recent advances in genomics and other high throughput biomolecular techniques (-omics) have provided numerous insights into the molecular mechanisms underlying breast cancer development and progression. A number of these mechanisms involve multiple layers of regulation. In this review, we summarize the current knowledge on the role of multiple omics in the regulation of breast cancer, including the effects of DNA methylation, non-coding RNA, and other epigenomic changes. We comment on how integrating such diverse mechanisms is envisioned as key to a more comprehensive understanding of breast carcinogenesis and cancer biology with relevance to prognostics, diagnostics and therapeutics. We also discuss the potential clinical implications of these findings and highlight areas for future research. Overall, our understanding of the molecular mechanisms of multi-omic regulation in breast cancer is rapidly increasing and has the potential to inform the development of novel therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Soledad Ochoa
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Center for Complexity Sciences, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
14
|
Arriaga-Canon C, Contreras-Espinosa L, Aguilar-Villanueva S, Bargalló-Rocha E, García-Gordillo JA, Cabrera-Galeana P, Castro-Hernández C, Jiménez-Trejo F, Herrera LA. The Clinical Utility of lncRNAs and Their Application as Molecular Biomarkers in Breast Cancer. Int J Mol Sci 2023; 24:ijms24087426. [PMID: 37108589 PMCID: PMC10138835 DOI: 10.3390/ijms24087426] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/04/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Given their tumor-specific and stage-specific gene expression, long non-coding RNAs (lncRNAs) have demonstrated to be potential molecular biomarkers for diagnosis, prognosis, and treatment response. Particularly, the lncRNAs DSCAM-AS1 and GATA3-AS1 serve as examples of this because of their high subtype-specific expression profile in luminal B-like breast cancer. This makes them candidates to use as molecular biomarkers in clinical practice. However, lncRNA studies in breast cancer are limited in sample size and are restricted to the determination of their biological function, which represents an obstacle for its inclusion as molecular biomarkers of clinical utility. Nevertheless, due to their expression specificity among diseases, such as cancer, and their stability in body fluids, lncRNAs are promising molecular biomarkers that could improve the reliability, sensitivity, and specificity of molecular techniques used in clinical diagnosis. The development of lncRNA-based diagnostics and lncRNA-based therapeutics will be useful in routine medical practice to improve patient clinical management and quality of life.
Collapse
Affiliation(s)
- Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico
| | - Laura Contreras-Espinosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico
- Posgrado en Ciencias Biológicas, Unidad de Posgrado, Edificio D, 1° Piso, Circuito de Posgrados, Ciudad Universitaria, Coyoacán, Mexico City C.P. 04510, Mexico
| | - Sergio Aguilar-Villanueva
- Departamento de Tumores Mamarios, Instituto Nacional de Cancerología, Tlalpan, Mexico City C.P. 14080, Mexico
| | - Enrique Bargalló-Rocha
- Departamento de Tumores Mamarios, Instituto Nacional de Cancerología, Tlalpan, Mexico City C.P. 14080, Mexico
| | - José Antonio García-Gordillo
- Departamento de Oncología Médica de Mama, Instituto Nacional de Cancerología, Tlalpan, Mexico City C.P. 14080, Mexico
| | - Paula Cabrera-Galeana
- Departamento de Oncología Médica de Mama, Instituto Nacional de Cancerología, Tlalpan, Mexico City C.P. 14080, Mexico
| | - Clementina Castro-Hernández
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico
| | | | - L A Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, UNAM, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan, Mexico City C.P. 14080, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey C.P. 64710, Mexico
| |
Collapse
|
15
|
Tsai PC, Lee TH, Kuo KC, Su FY, Lee TLM, Marostica E, Ugai T, Zhao M, Lau MC, Väyrynen JP, Giannakis M, Takashima Y, Kahaki SM, Wu K, Song M, Meyerhardt JA, Chan AT, Chiang JH, Nowak J, Ogino S, Yu KH. Histopathology images predict multi-omics aberrations and prognoses in colorectal cancer patients. Nat Commun 2023; 14:2102. [PMID: 37055393 PMCID: PMC10102208 DOI: 10.1038/s41467-023-37179-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 03/03/2023] [Indexed: 04/15/2023] Open
Abstract
Histopathologic assessment is indispensable for diagnosing colorectal cancer (CRC). However, manual evaluation of the diseased tissues under the microscope cannot reliably inform patient prognosis or genomic variations crucial for treatment selections. To address these challenges, we develop the Multi-omics Multi-cohort Assessment (MOMA) platform, an explainable machine learning approach, to systematically identify and interpret the relationship between patients' histologic patterns, multi-omics, and clinical profiles in three large patient cohorts (n = 1888). MOMA successfully predicts the overall survival, disease-free survival (log-rank test P-value<0.05), and copy number alterations of CRC patients. In addition, our approaches identify interpretable pathology patterns predictive of gene expression profiles, microsatellite instability status, and clinically actionable genetic alterations. We show that MOMA models are generalizable to multiple patient populations with different demographic compositions and pathology images collected from distinctive digitization methods. Our machine learning approaches provide clinically actionable predictions that could inform treatments for colorectal cancer patients.
Collapse
Affiliation(s)
- Pei-Chen Tsai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Tsung-Hua Lee
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Kun-Chi Kuo
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Fang-Yi Su
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Tsung-Lu Michael Lee
- Department of Computer Science and Information Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan ROC
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA, USA
| | - Tomotaka Ugai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Melissa Zhao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Mai Chan Lau
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Juha P Väyrynen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Marios Giannakis
- Department of Medicine, Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Andrew T Chan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jung-Hsien Chiang
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC.
| | - Jonathan Nowak
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
16
|
Zhong Y, Ren J, Li R, Xuan Y, Yao W, Yang Q, Gan Y, Yu S, Yuan J. Prediction of the Endocrine disruption profile of fluorinated biphenyls and analogues: An in silico study. CHEMOSPHERE 2023; 314:137701. [PMID: 36587920 DOI: 10.1016/j.chemosphere.2022.137701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 06/17/2023]
Abstract
Fluorinated biphenyls and their analogues (FBAs) are considered new persistent organic pollutants, but their endocrine-disrupting effects are still unknown. To fill this gap, the binding probability of 44 FBAs to different nuclear hormone receptors (NHRs) was predicted using Endocrine Disruptome. And molecular similarity and network toxicology analysis were used to strengthen the docking screening. The docking results showed that FBAs could have high binding potential for various NHRs, such as estrogen receptors β antagonism (ERβ an), liver X receptors α (LXRα), estrogen receptors α (ERα), and liver X receptors β (LXRβ). The similarity analysis found that the degree of overlap of the NHR repertoire was related to the Tanimoto coefficient of FBAs. Network toxicology verified a part of docking screening results and identified endocrine-disrupting pathways worthy of attention. This study found out potential endocrine-disrupting FBAs and their vulnerable, and developed a workflow that would leverage in silico approaches including molecular docking, similarity, and network toxicology for risk prioritization of potential endocrine-disrupting compounds.
Collapse
Affiliation(s)
- Yuyan Zhong
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Jing Ren
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Rui Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yuxin Xuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Wu Yao
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Qianye Yang
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China
| | - Yin Gan
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, Henan, 475004, PR China
| | - Shuling Yu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University, Kaifeng, Henan, 475004, PR China.
| | - Jintao Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, PR China.
| |
Collapse
|
17
|
Wang S, Wang S, Wang Z. A survey on multi-omics-based cancer diagnosis using machine learning with the potential application in gastrointestinal cancer. Front Med (Lausanne) 2023; 9:1109365. [PMID: 36703893 PMCID: PMC9871466 DOI: 10.3389/fmed.2022.1109365] [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: 11/27/2022] [Accepted: 12/28/2022] [Indexed: 01/12/2023] Open
Abstract
Gastrointestinal cancer is becoming increasingly common, which leads to over 3 million deaths every year. No typical symptoms appear in the early stage of gastrointestinal cancer, posing a significant challenge in the diagnosis and treatment of patients with gastrointestinal cancer. Many patients are in the middle and late stages of gastrointestinal cancer when they feel uncomfortable, unfortunately, most of them will die of gastrointestinal cancer. Recently, various artificial intelligence techniques like machine learning based on multi-omics have been presented for cancer diagnosis and treatment in the era of precision medicine. This paper provides a survey on multi-omics-based cancer diagnosis using machine learning with potential application in gastrointestinal cancer. Particularly, we make a comprehensive summary and analysis from the perspective of multi-omics datasets, task types, and multi-omics-based integration methods. Furthermore, this paper points out the remaining challenges of multi-omics-based cancer diagnosis using machine learning and discusses future topics.
Collapse
Affiliation(s)
- Suixue Wang
- School of Information and Communication Engineering, Hainan University, Haikou, China
| | - Shuling Wang
- Department of Neurology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou, China
| | - Zhengxia Wang
- School of Computer Science and Technology, Hainan University, Haikou, China
| |
Collapse
|
18
|
Chan RC, To CKC, Cheng KCT, Yoshikazu T, Yan LLA, Tse GM. Artificial intelligence in breast cancer histopathology. Histopathology 2023; 82:198-210. [PMID: 36482271 DOI: 10.1111/his.14820] [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: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 12/13/2022]
Abstract
This is a review on the use of artificial intelligence for digital breast pathology. A systematic search on PubMed was conducted, identifying 17,324 research papers related to breast cancer pathology. Following a semimanual screening, 664 papers were retrieved and pursued. The papers are grouped into six major tasks performed by pathologists-namely, molecular and hormonal analysis, grading, mitotic figure counting, ki-67 indexing, tumour-infiltrating lymphocyte assessment, and lymph node metastases identification. Under each task, open-source datasets for research to build artificial intelligence (AI) tools are also listed. Many AI tools showed promise and demonstrated feasibility in the automation of routine pathology investigations. We expect continued growth of AI in this field as new algorithms mature.
Collapse
Affiliation(s)
- Ronald Ck Chan
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chun Kit Curtis To
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ka Chuen Tom Cheng
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tada Yoshikazu
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Lai Ling Amy Yan
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
19
|
Couture HD. Deep Learning-Based Prediction of Molecular Tumor Biomarkers from H&E: A Practical Review. J Pers Med 2022; 12:2022. [PMID: 36556243 PMCID: PMC9784641 DOI: 10.3390/jpm12122022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/26/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Molecular and genomic properties are critical in selecting cancer treatments to target individual tumors, particularly for immunotherapy. However, the methods to assess such properties are expensive, time-consuming, and often not routinely performed. Applying machine learning to H&E images can provide a more cost-effective screening method. Dozens of studies over the last few years have demonstrated that a variety of molecular biomarkers can be predicted from H&E alone using the advancements of deep learning: molecular alterations, genomic subtypes, protein biomarkers, and even the presence of viruses. This article reviews the diverse applications across cancer types and the methodology to train and validate these models on whole slide images. From bottom-up to pathologist-driven to hybrid approaches, the leading trends include a variety of weakly supervised deep learning-based approaches, as well as mechanisms for training strongly supervised models in select situations. While results of these algorithms look promising, some challenges still persist, including small training sets, rigorous validation, and model explainability. Biomarker prediction models may yield a screening method to determine when to run molecular tests or an alternative when molecular tests are not possible. They also create new opportunities in quantifying intratumoral heterogeneity and predicting patient outcomes.
Collapse
|
20
|
Deep learning models for histologic grading of breast cancer and association with disease prognosis. NPJ Breast Cancer 2022; 8:113. [PMID: 36192400 PMCID: PMC9530224 DOI: 10.1038/s41523-022-00478-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 09/01/2022] [Indexed: 12/02/2022] Open
Abstract
Histologic grading of breast cancer involves review and scoring of three well-established morphologic features: mitotic count, nuclear pleomorphism, and tubule formation. Taken together, these features form the basis of the Nottingham Grading System which is used to inform breast cancer characterization and prognosis. In this study, we develop deep learning models to perform histologic scoring of all three components using digitized hematoxylin and eosin-stained slides containing invasive breast carcinoma. We first evaluate model performance using pathologist-based reference standards for each component. To complement this typical approach to evaluation, we further evaluate the deep learning models via prognostic analyses. The individual component models perform at or above published benchmarks for algorithm-based grading approaches, achieving high concordance rates with pathologist grading. Further, prognostic performance using deep learning-based grading is on par with that of pathologists performing review of matched slides. By providing scores for each component feature, the deep-learning based approach also provides the potential to identify the grading components contributing most to prognostic value. This may enable optimized prognostic models, opportunities to improve access to consistent grading, and approaches to better understand the links between histologic features and clinical outcomes in breast cancer.
Collapse
|
21
|
Investigating How Reproducibility and Geometrical Representation in UMAP Dimensionality Reduction Impact the Stratification of Breast Cancer Tumors. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12094247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Advances in next-generation sequencing have provided high-dimensional RNA-seq datasets, allowing the stratification of some tumor patients based on their transcriptomic profiles. Machine learning methods have been used to reduce and cluster high-dimensional data. Recently, uniform manifold approximation and projection (UMAP) was applied to project genomic datasets in low-dimensional Euclidean latent space. Here, we evaluated how different representations of the UMAP embedding can impact the analysis of breast cancer (BC) stratification. We projected BC RNA-seq data on Euclidean, spherical, and hyperbolic spaces, and stratified BC patients via clustering algorithms. We also proposed a pipeline to yield more reproducible clustering outputs. The results show how the selection of the latent space can affect downstream stratification results and suggest that the exploration of different geometrical representations is recommended to explore data structure and samples’ relationships.
Collapse
|
22
|
Brown LJ, Achinger-Kawecka J, Portman N, Clark S, Stirzaker C, Lim E. Epigenetic Therapies and Biomarkers in Breast Cancer. Cancers (Basel) 2022; 14:474. [PMID: 35158742 PMCID: PMC8833457 DOI: 10.3390/cancers14030474] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/07/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Epigenetic therapies remain a promising, but still not widely used, approach in the management of patients with cancer. To date, the efficacy and use of epigenetic therapies has been demonstrated primarily in the management of haematological malignancies, with limited supportive data in solid malignancies. The most studied epigenetic therapies in breast cancer are those that target DNA methylation and histone modification; however, none have been approved for routine clinical use. The majority of pre-clinical and clinical studies have focused on triple negative breast cancer (TNBC) and hormone-receptor positive breast cancer. Even though the use of epigenetic therapies alone in the treatment of breast cancer has not shown significant clinical benefit, these therapies show most promise in use in combinations with other treatments. With improving technologies available to study the epigenetic landscape in cancer, novel epigenetic alterations are increasingly being identified as potential biomarkers of response to conventional and epigenetic therapies. In this review, we describe epigenetic targets and potential epigenetic biomarkers in breast cancer, with a focus on clinical trials of epigenetic therapies. We describe alterations to the epigenetic landscape in breast cancer and in treatment resistance, highlighting mechanisms and potential targets for epigenetic therapies. We provide an updated review on epigenetic therapies in the pre-clinical and clinical setting in breast cancer, with a focus on potential real-world applications. Finally, we report on the potential value of epigenetic biomarkers in diagnosis, prognosis and prediction of response to therapy, to guide and inform the clinical management of breast cancer patients.
Collapse
Affiliation(s)
- Lauren Julia Brown
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Joanna Achinger-Kawecka
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Neil Portman
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Susan Clark
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Clare Stirzaker
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| | - Elgene Lim
- School of Clinical Medicine, St. Vincent’s Campus, University of New South Wales (UNSW), Sydney, NSW 2010, Australia; (L.J.B.); (J.A.-K.); (N.P.); (S.C.); (C.S.)
- Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
| |
Collapse
|