1
|
Yeghaian M, Bodalal Z, van den Broek D, Haanen JBAG, Beets-Tan RGH, Trebeschi S, van Gerven MAJ. Multimodal integration of longitudinal noninvasive diagnostics for survival prediction in immunotherapy using deep learning. J Am Med Inform Assoc 2025:ocaf074. [PMID: 40418276 DOI: 10.1093/jamia/ocaf074] [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: 12/09/2024] [Revised: 01/17/2025] [Accepted: 04/29/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVES Immunotherapies have revolutionized the landscape of cancer treatments. However, our understanding of response patterns in advanced cancers treated with immunotherapy remains limited. By leveraging routinely collected noninvasive longitudinal and multimodal data with artificial intelligence, we could unlock the potential to transform immunotherapy for cancer patients, paving the way for personalized treatment approaches. MATERIALS AND METHODS In this study, we developed a novel artificial neural network architecture, multimodal transformer-based simple temporal attention (MMTSimTA) network, building upon a combination of recent successful developments. We integrated pre- and on-treatment blood measurements, prescribed medications, and CT-based volumes of organs from a large pan-cancer cohort of 694 patients treated with immunotherapy to predict mortality at 3, 6, 9, and 12 months. Different variants of our extended MMTSimTA network were implemented and compared to baseline methods, incorporating intermediate and late fusion-based integration methods. RESULTS The strongest prognostic performance was demonstrated using a variant of the MMTSimTA model with area under the curves of 0.84 ± 0.04, 0.83 ± 0.02, 0.82 ± 0.02, 0.81 ± 0.03 for 3-, 6-, 9-, and 12-month survival prediction, respectively. DISCUSSION Our findings show that integrating noninvasive longitudinal data using our novel architecture yields an improved multimodal prognostic performance, especially in short-term survival prediction. CONCLUSION Our study demonstrates that multimodal longitudinal integration of noninvasive data using deep learning may offer a promising approach for personalized prognostication in immunotherapy-treated cancer patients.
Collapse
Affiliation(s)
- Melda Yeghaian
- Department of Machine Learning and Neural Computing, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 GD, The Netherlands
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Zuhir Bodalal
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Daan van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
| | - John B A G Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- Division of Molecular Oncology and Immunology, Oncode Institute, Amsterdam 1066 CX, The Netherlands
- Department of Medical Oncology, Leiden University Medical Center, Leiden 2333 ZG, The Netherlands
- Melanoma Clinic, Centre Hospitalier Universitaire Vaudois, Lausanne 1005, Switzerland
| | - Regina G H Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
- Faculty of Health Science, University of Southern Denmark, Odense 5230, Denmark
| | - Stefano Trebeschi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam 1066 CX, The Netherlands
- GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht 6229 ER, The Netherlands
| | - Marcel A J van Gerven
- Department of Machine Learning and Neural Computing, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen 6525 GD, The Netherlands
| |
Collapse
|
2
|
Yi X, Yu X, Li C, Li J, Cao H, Lu Q, Li J, Hou J. Deep learning radiopathomics based on pretreatment MRI and whole slide images for predicting over survival in locally advanced nasopharyngeal carcinoma. Radiother Oncol 2025:110949. [PMID: 40409367 DOI: 10.1016/j.radonc.2025.110949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 04/27/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
PURPOSE To develop an integrative radiopathomic model based on deep learning to predict overall survival (OS) in locally advanced nasopharyngeal carcinoma (LANPC) patients. MATERIALS AND METHODS A cohort of 343 LANPC patients with pretreatment MRI and whole slide image (WSI) were randomly divided into training (n = 202), validation (n = 91), and external test (n = 50) sets. For WSIs, a self-attention mechanism was employed to assess the significance of different patches for the prognostic task, aggregating them into a WSI-level representation. For MRI, a multilayer perceptron was used to encode the extracted radiomic features, resulting in an MRI-level representation. These were combined in a multimodal fusion model to produce prognostic predictions. Model performances were evaluated using the concordance index (C-index), and Kaplan-Meier curves were employed for risk stratification. To enhance model interpretability, attention-based and Integrated Gradients techniques were applied to explain how WSIs and MRI features contribute to prognosis predictions. RESULTS The radiopathomics model achieved high predictive accuracy in predicting the OS, with a C-index of 0.755 (95 % CI: 0.673-0.838) and 0.744 (95 % CI: 0.623-0.808) in the training and validation sets, respectively, outperforming single-modality models (radiomic signature: 0.636, 95 % CI: 0.584-0.688; deep pathomic signature: 0.736, 95 % CI: 0.684-0.810). In the external test, similar findings were observed for the predictive performance of the radiopathomics, radiomic signature, and deep pathomic signature, with their C-indices being 0.735, 0.626, and 0.660 respectively. The radiopathomics model effectively stratified patients into high- and low-risk groups (P < 0.001). Additionally, attention heatmaps revealed that high-attention regions corresponded with tumor areas in both risk groups. CONCLUSIO n: The radiopathomics model holds promise for predicting clinical outcomes in LANPC patients, offering a potential tool for improving clinical decision-making.
Collapse
Affiliation(s)
- Xiaochun Yi
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013 Hunan, PR China
| | - Xiaoping Yu
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013 Hunan, PR China
| | - Congrui Li
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013 Hunan, PR China
| | - Junjian Li
- Hunan Provincial Key Laboratory on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, PR China
| | - Hui Cao
- Department of Health Service Center, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013 Hunan, PR China
| | - Qiang Lu
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013 Hunan, PR China
| | - Junjun Li
- Department of Pathology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, Hunan 410013, PR China
| | - Jing Hou
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013 Hunan, PR China.
| |
Collapse
|
3
|
Nikolaou N, Salazar D, RaviPrakash H, Gonçalves M, Mulla R, Burlutskiy N, Markuzon N, Jacob E. A machine learning approach for multimodal data fusion for survival prediction in cancer patients. NPJ Precis Oncol 2025; 9:128. [PMID: 40325104 PMCID: PMC12053085 DOI: 10.1038/s41698-025-00917-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/19/2025] [Indexed: 05/07/2025] Open
Abstract
Technological advancements of the past decade have transformed cancer research, improving patient survival predictions through genotyping and multimodal data analysis. However, there is no comprehensive machine-learning pipeline for comparing methods to enhance these predictions. To address this, a versatile pipeline using The Cancer Genome Atlas (TCGA) data was developed, incorporating various data modalities such as transcripts, proteins, metabolites, and clinical factors. This approach manages challenges like high dimensionality, small sample sizes, and data heterogeneity. By applying different feature extraction and fusion strategies, notably late fusion models, the effectiveness of integrating diverse data types was demonstrated. Late fusion models consistently outperformed single-modality approaches in TCGA lung, breast, and pan-cancer datasets, offering higher accuracy and robustness. This research highlights the potential of comprehensive multimodal data integration in precision oncology to improve survival predictions for cancer patients. The study provides a reusable pipeline for the research community, suggesting future work on larger cohorts.
Collapse
Affiliation(s)
- Nikolaos Nikolaou
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, UK
- Department of Physics & Astronomy, University College London, London, UK
| | - Domingo Salazar
- Oncology Data Science, Oncology R&D, AstraZeneca, Cambridge, UK
| | | | | | - Rob Mulla
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA
| | | | - Natasha Markuzon
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA.
| | - Etai Jacob
- Oncology Data Science, Oncology R&D, AstraZeneca, Waltham, MA, USA.
| |
Collapse
|
4
|
Yang H, Wang J, Wang W, Shi S, Liu L, Yao Y, Tian G, Wang P, Yang J. MMsurv: a multimodal multi-instance multi-cancer survival prediction model integrating pathological images, clinical information, and sequencing data. Brief Bioinform 2025; 26:bbaf209. [PMID: 40366860 PMCID: PMC12077396 DOI: 10.1093/bib/bbaf209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/03/2024] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Accurate prediction of patient survival rates in cancer treatment is essential for effective therapeutic planning. Unfortunately, current models often underutilize the extensive multimodal data available, affecting confidence in predictions. This study presents MMSurv, an interpretable multimodal deep learning model to predict survival in different types of cancer. MMSurv integrates clinical information, sequencing data, and hematoxylin and eosin-stained whole-slide images (WSIs) to forecast patient survival. Specifically, we segment tumor regions from WSIs into image tiles and employ neural networks to encode each tile into one-dimensional feature vectors. We then optimize clinical features by applying word embedding techniques, inspired by natural language processing, to the clinical data. To better utilize the complementarity of multimodal data, this study proposes a novel fusion method, multimodal fusion method based on compact bilinear pooling and transformer, which integrates bilinear pooling with Transformer architecture. The fused features are then processed through a dual-layer multi-instance learning model to remove prognosis-irrelevant image patches and predict each patient's survival risk. Furthermore, we employ cell segmentation to investigate the cellular composition within the tiles that received high attention from the model, thereby enhancing its interpretive capacity. We evaluate our approach on six cancer types from The Cancer Genome Atlas. The results demonstrate that utilizing multimodal data leads to higher predictive accuracy compared to using single-modal image data, with an average C-index increase from 0.6750 to 0.7283. Additionally, we compare our proposed baseline model with state-of-the-art methods using the C-index and five-fold cross-validation approach, revealing a significant average improvement of nearly 10% in our model's performance.
Collapse
Affiliation(s)
- Hailong Yang
- School of Electrical and Information Engineering, Anhui University of Technology, No. 1530 Maxiang Road, Huashan District, Ma’anshan, Anhui 243032, China
- Department of Sciences, Geneis Beijing Co., Ltd., No. 31 Xinbei Road, Laiguangying, Chaoyang District, Beijing 100102, China
| | - Jia Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing 100142, P.R. China
| | - Wenyan Wang
- School of Electrical and Information Engineering, Anhui University of Technology, No. 1530 Maxiang Road, Huashan District, Ma’anshan, Anhui 243032, China
| | - Shufang Shi
- Department of Sciences, Geneis Beijing Co., Ltd., No. 31 Xinbei Road, Laiguangying, Chaoyang District, Beijing 100102, China
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Lijing Liu
- Qinghe Clinic, Beijing North Medical District, Chinese PLA General Hospital, No. 100 Youyi Road, Haidian District, Beijing 100080, China
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, No. 99 Longkunnan Road, Longhua District, Haikou, Hainan 571158, China
| | - Geng Tian
- Department of Sciences, Geneis Beijing Co., Ltd., No. 31 Xinbei Road, Laiguangying, Chaoyang District, Beijing 100102, China
| | - Peizhen Wang
- School of Electrical and Information Engineering, Anhui University of Technology, No. 1530 Maxiang Road, Huashan District, Ma’anshan, Anhui 243032, China
| | - Jialiang Yang
- Department of Sciences, Geneis Beijing Co., Ltd., No. 31 Xinbei Road, Laiguangying, Chaoyang District, Beijing 100102, China
| |
Collapse
|
5
|
Shi Y, Wang M, Liu H, Zhao F, Li A, Chen X. MIF: Multi-Shot Interactive Fusion Model for Cancer Survival Prediction Using Pathological Image and Genomic Data. IEEE J Biomed Health Inform 2025; 29:3247-3258. [PMID: 38324434 DOI: 10.1109/jbhi.2024.3363161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Accurate cancer survival prediction is crucial for oncologists to determine therapeutic plan, which directly influences the treatment efficacy and survival outcome of patient. Recently, multimodal fusion-based prognostic methods have demonstrated effectiveness for survival prediction by fusing diverse cancer-related data from different medical modalities, e.g., pathological images and genomic data. However, these works still face significant challenges. First, most approaches attempt multimodal fusion by simple one-shot fusion strategy, which is insufficient to explore complex interactions underlying in highly disparate multimodal data. Second, current methods for investigating multimodal interactions face the capability-efficiency dilemma, which is the difficult balance between powerful modeling capability and applicable computational efficiency, thus impeding effective multimodal fusion. In this study, to encounter these challenges, we propose an innovative multi-shot interactive fusion method named MIF for precise survival prediction by utilizing pathological and genomic data. Particularly, a novel multi-shot fusion framework is introduced to promote multimodal fusion by decomposing it into successive fusing stages, thus delicately integrating modalities in a progressive way. Moreover, to address the capacity-efficiency dilemma, various affinity-based interactive modules are introduced to synergize the multi-shot framework. Specifically, by harnessing comprehensive affinity information as guidance for mining interactions, the proposed interactive modules can efficiently generate low-dimensional discriminative multimodal representations. Extensive experiments on different cancer datasets unravel that our method not only successfully achieves state-of-the-art performance by performing effective multimodal fusion, but also possesses high computational efficiency compared to existing survival prediction methods.
Collapse
|
6
|
Harkos C, Hadjigeorgiou AG, Voutouri C, Kumar AS, Stylianopoulos T, Jain RK. Using mathematical modelling and AI to improve delivery and efficacy of therapies in cancer. Nat Rev Cancer 2025; 25:324-340. [PMID: 39972158 DOI: 10.1038/s41568-025-00796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
Mathematical modelling has proven to be a valuable tool in predicting the delivery and efficacy of molecular, antibody-based, nano and cellular therapy in solid tumours. Mathematical models based on our understanding of the biological processes at subcellular, cellular and tissue level are known as mechanistic models that, in turn, are divided into continuous and discrete models. Continuous models are further divided into lumped parameter models - for describing the temporal distribution of medicine in tumours and normal organs - and distributed parameter models - for studying the spatiotemporal distribution of therapy in tumours. Discrete models capture interactions at the cellular and subcellular levels. Collectively, these models are useful for optimizing the delivery and efficacy of molecular, nanoscale and cellular therapy in tumours by incorporating the biological characteristics of tumours, the physicochemical properties of drugs, the interactions among drugs, cancer cells and various components of the tumour microenvironment, and for enabling patient-specific predictions when combined with medical imaging. Artificial intelligence-based methods, such as machine learning, have ushered in a new era in oncology. These data-driven approaches complement mechanistic models and have immense potential for improving cancer detection, treatment and drug discovery. Here we review these diverse approaches and suggest ways to combine mechanistic and artificial intelligence-based models to further improve patient treatment outcomes.
Collapse
Affiliation(s)
- Constantinos Harkos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Wu K, Jiang Z, Tang K, Shi J, Xie F, Wang W, Wu H, Zheng Y. Pan-Cancer Histopathology WSI Pre-Training With Position-Aware Masked Autoencoder. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:1610-1623. [PMID: 40030469 DOI: 10.1109/tmi.2024.3513358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Large-scale pre-training models have promoted the development of histopathology image analysis. However, existing self-supervised methods for histopathology images primarily focus on learning patch features, while there is a notable gap in the availability of pre-training models specifically designed for WSI-level feature learning. In this paper, we propose a novel self-supervised learning framework for pan-cancer WSI-level representation pre-training with the designed position-aware masked autoencoder (PAMA). Meanwhile, we propose the position-aware cross-attention (PACA) module with a kernel reorientation (KRO) strategy and an anchor dropout (AD) mechanism. The KRO strategy can capture the complete semantic structure and eliminate ambiguity in WSIs, and the AD contributes to enhancing the robustness and generalization of the model. We evaluated our method on 7 large-scale datasets from multiple organs for pan-cancer classification tasks. The results have demonstrated the effectiveness and generalization of PAMA in discriminative WSI representation learning and pan-cancer WSI pre-training. The proposed method was also compared with 8 WSI analysis methods. The experimental results have indicated that our proposed PAMA is superior to the state-of-the-art methods. The code and checkpoints are available at https://github.com/WkEEn/PAMA.
Collapse
|
8
|
Llinas-Bertran A, Butjosa-Espín M, Barberi V, Seoane JA. Multimodal data integration in early-stage breast cancer. Breast 2025; 80:103892. [PMID: 39922065 PMCID: PMC11973824 DOI: 10.1016/j.breast.2025.103892] [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: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The use of biomarkers in breast cancer has significantly improved patient outcomes through targeted therapies, such as hormone therapy anti-Her2 therapy and CDK4/6 or PARP inhibitors. However, existing knowledge does not fully encompass the diverse nature of breast cancer, particularly in triple-negative tumors. The integration of multi-omics and multimodal data has the potential to provide new insights into biological processes, to improve breast cancer patient stratification, enhance prognosis and response prediction, and identify new biomarkers. This review presents a comprehensive overview of the state-of-the-art multimodal (including molecular and image) data integration algorithms developed and with applicability to breast cancer stratification, prognosis, or biomarker identification. We examined the primary challenges and opportunities of these multimodal data integration algorithms, including their advantages, limitations, and critical considerations for future research. We aimed to describe models that are not only academically and preclinically relevant, but also applicable to clinical settings.
Collapse
Affiliation(s)
- Arnau Llinas-Bertran
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Butjosa-Espín
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Vittoria Barberi
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
9
|
Lee KH, Lee J, Choi GH, Yun J, Kang J, Choi J, Kim KM, Kim N. Deep Learning-Based Prediction of Post-treatment Survival in Hepatocellular Carcinoma Patients Using Pre-treatment CT Images and Clinical Data. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:1212-1223. [PMID: 39147884 PMCID: PMC11950573 DOI: 10.1007/s10278-024-01227-2] [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: 12/23/2023] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
The objective of this study was to develop and evaluate a model for predicting post-treatment survival in hepatocellular carcinoma (HCC) patients using their CT images and clinical information, including various treatment information. We collected pre-treatment contrast-enhanced CT images and clinical information including patient-related factors, initial treatment options, and survival status from 692 patients. The patient cohort was divided into a training cohort (n = 507), a testing cohort (n = 146), and an external CT cohort (n = 39), which included patients who underwent CT scans at other institutions. After model training using fivefold cross-validation, model validation was performed on both the testing cohort and the external CT cohort. Our cascaded model employed a 3D convolutional neural network (CNN) to extract features from CT images and derive final survival probabilities. These probabilities were obtained by concatenating previously predicted probabilities for each interval with the patient-related factors and treatment options. We utilized two consecutive fully connected layers for this process, resulting in a number of final outputs corresponding to the number of time intervals, with values representing conditional survival probabilities for each interval. Performance was assessed using the concordance index (C-index), the mean cumulative/dynamic area under the receiver operating characteristics curve (mC/D AUC), and the mean Brier score (mBS), calculated every 3 months. Through an ablation study, we found that using DenseNet-121 as the backbone network and setting the prediction interval to 6 months optimized the model's performance. The integration of multimodal data resulted in superior predictive capabilities compared to models using only CT images or clinical information (C index 0.824 [95% CI 0.822-0.826], mC/D AUC 0.893 [95% CI 0.891-0.895], and mBS 0.121 [95% CI 0.120-0.123] for internal test cohort; C index 0.750 [95% CI 0.747-0.753], mC/D AUC 0.819 [95% CI 0.816-0.823], and mBS 0.159 [95% CI 0.158-0.161] for external CT cohort, respectively). Our CNN-based discrete-time survival prediction model with CT images and clinical information demonstrated promising results in predicting post-treatment survival of patients with HCC.
Collapse
Affiliation(s)
- Kyung Hwa Lee
- Department of Radiation Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jungwook Lee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Gwang Hyeon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea
| | - Jihye Yun
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiseon Kang
- Department of Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jonggi Choi
- Department of Gastroenterology, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea
| | - Kang Mo Kim
- Department of Gastroenterology, Asan Liver Center, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| | - Namkug Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-Ro 43-Gil, Songpa-Gu, Seoul, 05505, Republic of Korea.
| |
Collapse
|
10
|
He B, Wang L, Zhou W, Liu H, Wang Y, Lv K, He K. A fusion model to predict the survival of colorectal cancer based on histopathological image and gene mutation. Sci Rep 2025; 15:9677. [PMID: 40113813 PMCID: PMC11926114 DOI: 10.1038/s41598-025-91420-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025] Open
Abstract
Colorectal cancer (CRC) is a prevalent gastrointestinal tumor worldwide with high morbidity and mortality. Predicting the survival of CRC patients not only enhances understanding of their life expectancies but also aids clinicians in making informed decisions regarding suitable adjuvant treatments. Although there are many clinical, genomic, and transcriptomic studies on this hot topic, only a few studies have explored the direction of integrating advanced deep learning algorithms and histopathological images. In addition, it is still unclear if combining histopathological images and molecular data can better predict patients' survival. To fill in this gap, we proposed in this study a novel multimodal deep learning computational framework using Multimodal Compact Bilinear Pooling (MCBP) to predict the 5-year survival of CRC patients from histopathological images, clinical information, and molecular data. We applied our framework to the cancer genome atlas (TCGA) CRC data, consisting of 84 samples with histopathological images, clinical information, mRNA sequencing data, and gene mutation data all available. Under the 5-fold cross-validation, the model using only histopathological images achieved an area under the curve (AUC) of 0.743. Whereas, the model combining image and clinical information and the model combining image and gene mutation information achieved AUCs of 0.771 and 0.773 respectively, better than that of the image solely. Our study demonstrates that histopathological images can reasonably predict the 5-year survival of CRC patients, and that the appropriate integration of these images with clinical or molecular data can further enhance predictive performance.
Collapse
Affiliation(s)
- Binsheng He
- First Clinical College, Changsha Medical University, Changsha, 410219, P. R. China
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410219, P. R. China
| | - Lixia Wang
- School of Mathematical Sciences, Ocean University of China, Qingdao, 266000, P. R. China
| | - Wenjing Zhou
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266033, P. R. China
| | - Haiyan Liu
- First Clinical College, Changsha Medical University, Changsha, 410219, P. R. China
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410219, P. R. China
| | - Yingxuan Wang
- School of Mathematical Sciences, Ocean University of China, Qingdao, 266000, P. R. China
| | - Kebo Lv
- School of Mathematical Sciences, Ocean University of China, Qingdao, 266000, P. R. China.
| | - Kunhui He
- First Clinical College, Changsha Medical University, Changsha, 410219, P. R. China.
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410219, P. R. China.
| |
Collapse
|
11
|
Creux C, Zehraoui F, Radvanyi F, Tahi F. MMnc: multi-modal interpretable representation for non-coding RNA classification and class annotation. BIOINFORMATICS (OXFORD, ENGLAND) 2025; 41:btaf051. [PMID: 39891346 PMCID: PMC11890286 DOI: 10.1093/bioinformatics/btaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/16/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
MOTIVATION As the biological roles and disease implications of non-coding RNAs continue to emerge, the need to thoroughly characterize previously unexplored non-coding RNAs becomes increasingly urgent. These molecules hold potential as biomarkers and therapeutic targets. However, the vast and complex nature of non-coding RNAs data presents a challenge. We introduce MMnc, an interpretable deep-learning approach designed to classify non-coding RNAs into functional groups. MMnc leverages multiple data sources-such as the sequence, secondary structure, and expression-using attention-based multi-modal data integration. This ensures the learning of meaningful representations while accounting for missing sources in some samples. RESULTS Our findings demonstrate that MMnc achieves high classification accuracy across diverse non-coding RNA classes. The method's modular architecture allows for the consideration of multiple types of modalities, whereas other tools only consider one or two at most. MMnc is resilient to missing data, ensuring that all available information is effectively utilized. Importantly, the generated attention scores offer interpretable insights into the underlying patterns of the different non-coding RNA classes, potentially driving future non-coding RNA research and applications. AVAILABILITY AND IMPLEMENTATION Data and source code can be found at EvryRNA.ibisc.univ-evry.fr/EvryRNA/MMnc.
Collapse
Affiliation(s)
- Constance Creux
- Université Paris-Saclay, Univ Evry, IBISC, Evry-Courcouronnes 91020, France
- Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris 75248, France
| | - Farida Zehraoui
- Université Paris-Saclay, Univ Evry, IBISC, Evry-Courcouronnes 91020, France
| | - François Radvanyi
- Molecular Oncology, PSL Research University, CNRS, UMR 144, Institut Curie, Paris 75248, France
| | - Fariza Tahi
- Université Paris-Saclay, Univ Evry, IBISC, Evry-Courcouronnes 91020, France
| |
Collapse
|
12
|
Carrillo-Perez F, Pizurica M, Zheng Y, Nandi TN, Madduri R, Shen J, Gevaert O. Generation of synthetic whole-slide image tiles of tumours from RNA-sequencing data via cascaded diffusion models. Nat Biomed Eng 2025; 9:320-332. [PMID: 38514775 DOI: 10.1038/s41551-024-01193-8] [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: 12/27/2022] [Accepted: 02/29/2024] [Indexed: 03/23/2024]
Abstract
Training machine-learning models with synthetically generated data can alleviate the problem of data scarcity when acquiring diverse and sufficiently large datasets is costly and challenging. Here we show that cascaded diffusion models can be used to synthesize realistic whole-slide image tiles from latent representations of RNA-sequencing data from human tumours. Alterations in gene expression affected the composition of cell types in the generated synthetic image tiles, which accurately preserved the distribution of cell types and maintained the cell fraction observed in bulk RNA-sequencing data, as we show for lung adenocarcinoma, kidney renal papillary cell carcinoma, cervical squamous cell carcinoma, colon adenocarcinoma and glioblastoma. Machine-learning models pretrained with the generated synthetic data performed better than models trained from scratch. Synthetic data may accelerate the development of machine-learning models in scarce-data settings and allow for the imputation of missing data modalities.
Collapse
Affiliation(s)
- Francisco Carrillo-Perez
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
| | - Marija Pizurica
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
- Internet technology and Data science Lab (IDLab), Ghent University, Ghent, Belgium
| | - Yuanning Zheng
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA
| | - Tarak Nath Nandi
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Ravi Madduri
- Data Science and Learning Division, Argonne National Laboratory, Lemont, IL, USA
| | - Jeanne Shen
- Department of Pathology, Stanford University, School of Medicine, Palo Alto, CA, USA
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Stanford University, School of Medicine, Stanford, CA, USA.
- Department of Biomedical Data Science, Stanford University, School of Medicine, Stanford, CA, USA.
| |
Collapse
|
13
|
Wang M, Fan S, Li Y, Gao B, Xie Z, Chen H. Robust multi-modal fusion architecture for medical data with knowledge distillation. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 260:108568. [PMID: 39709743 DOI: 10.1016/j.cmpb.2024.108568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/01/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND The fusion of multi-modal data has been shown to significantly enhance the performance of deep learning models, particularly on medical data. However, missing modalities are common in medical data due to patient specificity, which poses a substantial challenge to the application of these models. OBJECTIVE This study aimed to develop a novel and efficient multi-modal fusion framework for medical datasets that maintains consistent performance, even in the absence of one or more modalities. METHODS In this paper, we fused three modalities: chest X-ray radiographs, history of present illness text, and tabular data such as demographics and laboratory tests. A multi-modal fusion module based on pooled bottleneck (PB) attention was proposed in conjunction with knowledge distillation (KD) for enhancing model inference in the case of missing modalities. In addition, we introduced a gradient modulation (GM) method to deal with the unbalanced optimization in multi-modal model training. Finally, we designed comparison and ablation experiments to evaluate the fusion effect, the model robustness to missing modalities, and the contribution of each component (PB, KD, and GM). The evaluation experiments were performed on the MIMIC-IV datasets with the task of predicting in-hospital mortality risk. Model performance was assessed using the area under the receiver operating characteristic curve (AUROC) and the area under the precision-recall curve (AUPRC). RESULTS The proposed multi-modal fusion framework achieved an AUROC of 0.886 and AUPRC of 0.459, significantly surpassing the performance of baseline models. Even when one or two modalities were missing, our model consistently outperformed the reference models. Ablation of each of the three components resulted in varying degrees of performance degradation, highlighting their distinct contributions to the model's overall effectiveness. CONCLUSIONS This innovative multi-modal fusion architecture has demonstrated robustness to missing modalities, and has shown excellent performance in fusing three medical modalities for patient outcome prediction. This study provides a novel idea for addressing the challenge of missing modalities and has the potential be scaled to additional modalities.
Collapse
Affiliation(s)
- Muyu Wang
- School of Biomedical Engineering, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China
| | - Shiyu Fan
- School of Biomedical Engineering, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China
| | - Yichen Li
- School of Biomedical Engineering, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China
| | - Binyu Gao
- School of Biomedical Engineering, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China
| | - Zhongrang Xie
- School of Biomedical Engineering, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China
| | - Hui Chen
- School of Biomedical Engineering, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China; Beijing Key Laboratory of Fundamental Research on Biomechanics in Clinical Application, Capital Medical University, No.10, Xitoutiao, You An Men, Fengtai District, Beijing 100069, China.
| |
Collapse
|
14
|
Keyl J, Keyl P, Montavon G, Hosch R, Brehmer A, Mochmann L, Jurmeister P, Dernbach G, Kim M, Koitka S, Bauer S, Bechrakis N, Forsting M, Führer-Sakel D, Glas M, Grünwald V, Hadaschik B, Haubold J, Herrmann K, Kasper S, Kimmig R, Lang S, Rassaf T, Roesch A, Schadendorf D, Siveke JT, Stuschke M, Sure U, Totzeck M, Welt A, Wiesweg M, Baba HA, Nensa F, Egger J, Müller KR, Schuler M, Klauschen F, Kleesiek J. Decoding pan-cancer treatment outcomes using multimodal real-world data and explainable artificial intelligence. NATURE CANCER 2025; 6:307-322. [PMID: 39885364 PMCID: PMC11864985 DOI: 10.1038/s43018-024-00891-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 12/06/2024] [Indexed: 02/01/2025]
Abstract
Despite advances in precision oncology, clinical decision-making still relies on limited variables and expert knowledge. To address this limitation, we combined multimodal real-world data and explainable artificial intelligence (xAI) to introduce AI-derived (AID) markers for clinical decision support. We used xAI to decode the outcome of 15,726 patients across 38 solid cancer entities based on 350 markers, including clinical records, image-derived body compositions, and mutational tumor profiles. xAI determined the prognostic contribution of each clinical marker at the patient level and identified 114 key markers that accounted for 90% of the neural network's decision process. Moreover, xAI enabled us to uncover 1,373 prognostic interactions between markers. Our approach was validated in an independent cohort of 3,288 patients with lung cancer from a US nationwide electronic health record-derived database. These results show the potential of xAI to transform the assessment of clinical variables and enable personalized, data-driven cancer care.
Collapse
Affiliation(s)
- Julius Keyl
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
- Institute of Pathology, University Hospital Essen (AöR), Essen, Germany
| | - Philipp Keyl
- Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | - Grégoire Montavon
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
- Machine Learning Group, Technical University of Berlin, Berlin, Germany
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - René Hosch
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
| | - Alexander Brehmer
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
| | - Liliana Mochmann
- Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Philipp Jurmeister
- Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Gabriel Dernbach
- Machine Learning Group, Technical University of Berlin, Berlin, Germany
| | - Moon Kim
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
| | - Sven Koitka
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen (AöR), Essen, Germany
| | - Sebastian Bauer
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
| | - Nikolaos Bechrakis
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Ophthalmology, University Hospital Essen (AöR), Essen, Germany
| | - Michael Forsting
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
| | - Dagmar Führer-Sakel
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Essen (AöR), Essen, Germany
| | - Martin Glas
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Division of Clinical Neurooncology, Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Medicine Essen, University Duisburg-Essen, Essen, Germany
| | - Viktor Grünwald
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Urology, University Hospital Essen (AöR), Essen, Germany
| | - Boris Hadaschik
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Urology, University Hospital Essen (AöR), Essen, Germany
| | - Johannes Haubold
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Nuclear Medicine, University Hospital Essen (AöR), Essen, Germany
| | - Stefan Kasper
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
| | - Rainer Kimmig
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- Department of Gynecology and Obstetrics, University Hospital Essen (AöR), Essen, Germany
| | - Stephan Lang
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Department of Otorhinolaryngology, University Hospital Essen (AöR), Essen, Germany
| | - Tienush Rassaf
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen (AöR), Essen, Germany
| | - Alexander Roesch
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Dermatology, University Hospital Essen (AöR), Essen, Germany
| | - Dirk Schadendorf
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Dermatology, University Hospital Essen (AöR), Essen, Germany
- Research Alliance Ruhr, Research Center One Health, University of Duisburg-Essen, Essen, Germany
| | - Jens T Siveke
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany
- Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK Partner Site Essen) and German Cancer Research Center, DKFZ, Heidelberg, Germany
| | - Martin Stuschke
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Radiotherapy, University Hospital Essen (AöR), Essen, Germany
| | - Ulrich Sure
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
- Department of Neurosurgery and Spine Surgery, University Hospital Essen (AöR), Essen, Germany
| | - Matthias Totzeck
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen (AöR), Essen, Germany
| | - Anja Welt
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - Marcel Wiesweg
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Felix Nensa
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
- Institute for Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen (AöR), Essen, Germany
- Medical Faculty, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany
| | - Jan Egger
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany
| | - Klaus-Robert Müller
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany.
- Machine Learning Group, Technical University of Berlin, Berlin, Germany.
- Department of Artificial Intelligence, Korea University, Seoul, South Korea.
- MPI for Informatics, Saarbrücken, Germany.
| | - Martin Schuler
- Department of Medical Oncology, University Hospital Essen (AöR), Essen, Germany.
- Medical Faculty, University of Duisburg-Essen, Essen, Germany.
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany.
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany.
| | - Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-University Munich, Munich, Germany.
- BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Berlin partner site, Berlin, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Munich partner site, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Jens Kleesiek
- Institute for Artificial Intelligence in Medicine, University Hospital Essen (AöR), Essen, Germany.
- Medical Faculty, University of Duisburg-Essen, Essen, Germany.
- West German Cancer Center, University Hospital Essen (AöR), Essen, Germany.
- German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany.
| |
Collapse
|
15
|
Zabaleta J, Aguinagalde B, Lopez I, Fernandez-Monge A, Lizarbe JA, Mainer M, Ferrer-Bonsoms JA, de Assas M. Utility of Artificial Intelligence for Decision Making in Thoracic Multidisciplinary Tumor Boards. J Clin Med 2025; 14:399. [PMID: 39860405 PMCID: PMC11765867 DOI: 10.3390/jcm14020399] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/28/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The aim of this study was to analyze whether the implementation of artificial intelligence (AI), specifically the Natural Language Processing (NLP) branch developed by OpenAI, could help a thoracic multidisciplinary tumor board (MTB) make decisions if provided with all of the patient data presented to the committee and supported by accepted clinical practice guidelines. Methods: This is a retrospective comparative study. The inclusion criteria were defined as all patients who presented at the thoracic MTB with a suspicious or first diagnosis of non-small-cell lung cancer between January 2023 and June 2023. Intervention: GPT 3.5 turbo chat was used, providing the clinical case summary presented in committee proceedings and the latest SEPAR lung cancer treatment guidelines. The application was asked to issue one of the following recommendations: follow-up, surgery, chemotherapy, radiotherapy, or chemoradiotherapy. Statistical analysis: A concordance analysis was performed by measuring the Kappa coefficient to evaluate the consistency between the results of the AI and the committee's decision. Results: Fifty-two patients were included in the study. The AI had an overall concordance of 76%, with a Kappa index of 0.59 and a consistency and replicability of 92.3% for the patients in whom it recommended surgery (after repeating the cases four times). Conclusions: AI is an interesting tool which could help in decision making in MTBs.
Collapse
Affiliation(s)
- Jon Zabaleta
- Department of Thoracic Surgery, Basque Health Service, Donostialdea Integrated Health Organisation, 20014 San Sebastian, Spain; (B.A.)
| | - Borja Aguinagalde
- Department of Thoracic Surgery, Basque Health Service, Donostialdea Integrated Health Organisation, 20014 San Sebastian, Spain; (B.A.)
| | - Iker Lopez
- Department of Thoracic Surgery, Basque Health Service, Donostialdea Integrated Health Organisation, 20014 San Sebastian, Spain; (B.A.)
| | - Arantza Fernandez-Monge
- Department of Thoracic Surgery, Basque Health Service, Donostialdea Integrated Health Organisation, 20014 San Sebastian, Spain; (B.A.)
| | - Jon A. Lizarbe
- Department of Thoracic Surgery, Basque Health Service, Donostialdea Integrated Health Organisation, 20014 San Sebastian, Spain; (B.A.)
| | - Maria Mainer
- Department of Thoracic Surgery, Basque Health Service, Donostialdea Integrated Health Organisation, 20014 San Sebastian, Spain; (B.A.)
| | | | - Mateo de Assas
- Tecnun, School of Engineering, University of Navarra, 20018 San Sebastian, Spain
| |
Collapse
|
16
|
Er AG, Aslan AT, Mikulska M, Akova M. Prevention and treatment of bacterial infections in patients with haematological cancers and haematopoietic stem cell transplantation: headways and shortcomings. Clin Microbiol Infect 2025; 31:24-28. [PMID: 39332598 DOI: 10.1016/j.cmi.2024.09.015] [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: 06/12/2024] [Revised: 08/20/2024] [Accepted: 09/19/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND There has been an unprecedented increase in the number of immunocompromised (IC) patients in clinical practice due to various reasons. Bacterial infections are a major cause of morbidity and mortality in this population. Emerging antibacterial resistance poses a significant challenge for prophylaxis and treatment. OBJECTIVES We aim to provide an update on antibacterial prophylaxis and management, particularly in high-risk IC patients, including those with acute leukaemia and haematopoietic stem cell transplantation. SOURCES We reviewed original articles, systematic reviews, metanalyses, and guidelines using PubMed, Scopus, and Web of Science. CONTENT We discussed the pros and cons of fluoroquinolone prophylaxis in neutropenic patients in the context of personalized medicine. We also attempted to give an outline of empirical treatment of presumed bacterial infections and targeted therapy options for documented bacterial infections, considering the recent surge of multiresistant bacteria in haematological cancer patients and local epidemiology. The shortcomings of the current strategies and future needs are discussed in detail. IMPLICATIONS Antibacterial prophylaxis with fluoroquinolones may still have a role in preventing bacterial infections in carefully selected patients with high-risk haematology. Empirical treatment algorithms still need to be adjusted according to host and local factors. The use of rapid diagnostic methods may lessen the need for broad-spectrum empirical antibiotic usage. However, these tests may not be easily available due to budget constraints in countries with limited resources but high rates of bacterial resistance. Although new antimicrobials provide opportunities for effective and less toxic treatment of highly resistant bacterial infections, large-scale data from IC patients are very limited. Using data-driven approaches with artificial intelligence tools may guide the selection of appropriate patients who would benefit most from such prophylactic and treatment regimens.
Collapse
Affiliation(s)
- Ahmet Görkem Er
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Abdullah Tarik Aslan
- Faculty of Medicine, University of Queensland Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Malgorzata Mikulska
- Department of Health Sciences, Division of Infectious Diseases, University of Genova, Genova, Italy; Division of Infectious Diseases, Italian Scientific Research and Care Institutes Ospedale Policlinico San Martino, Genova, Italy
| | - Murat Akova
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University School of Medicine, Ankara, Turkey.
| |
Collapse
|
17
|
Wen G, Li L. MMOSurv: meta-learning for few-shot survival analysis with multi-omics data. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 41:btae684. [PMID: 39563482 DOI: 10.1093/bioinformatics/btae684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 10/14/2024] [Accepted: 11/16/2024] [Indexed: 11/21/2024]
Abstract
MOTIVATION High-throughput techniques have produced a large amount of high-dimensional multi-omics data, which makes it promising to predict patient survival outcomes more accurately. Recent work has showed the superiority of multi-omics data in survival analysis. However, it remains challenging to integrate multi-omics data to solve few-shot survival prediction problem, with only a few available training samples, especially for rare cancers. RESULTS In this work, we propose a meta-learning framework for multi-omics few-shot survival analysis, namely MMOSurv, which enables to learn an effective multi-omics survival prediction model from a very few training samples of a specific cancer type, with the meta-knowledge across tasks from relevant cancer types. By assuming a deep Cox survival model with multiple omics, MMOSurv first learns an adaptable parameter initialization for the multi-omics survival model from abundant data of relevant cancers, and then adapts the parameters quickly and efficiently for the target cancer task with a very few training samples. Our experiments on eleven cancer types in The Cancer Genome Atlas datasets show that, compared to single-omics meta-learning methods, MMOSurv can better utilize the meta-information of similarities and relationships between different omics data from relevant cancer datasets to improve survival prediction of the target cancer with a very few multi-omics training samples. Furthermore, MMOSurv achieves better prediction performance than other state-of-the-art strategies such as multitask learning and pretraining. AVAILABILITY AND IMPLEMENTATION MMOSurv is freely available at https://github.com/LiminLi-xjtu/MMOSurv.
Collapse
Affiliation(s)
- Gang Wen
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Limin Li
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| |
Collapse
|
18
|
Wang H, Han X, Niu S, Cheng H, Ren J, Duan Y. DFASGCNS: A prognostic model for ovarian cancer prediction based on dual fusion channels and stacked graph convolution. PLoS One 2024; 19:e0315924. [PMID: 39680618 DOI: 10.1371/journal.pone.0315924] [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: 05/30/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Ovarian cancer is a malignant tumor with different clinicopathological and molecular characteristics. Due to its nonspecific early symptoms, the majority of patients are diagnosed with local or extensive metastasis, severely affecting treatment and prognosis. The occurrence of ovarian cancer is influenced by multiple complex mechanisms including genomics, transcriptomics, and proteomics. Integrating multiple types of omics data aids in predicting the survival rate of ovarian cancer patients. However, existing methods only fuse multi-omics data at the feature level, neglecting the shared and complementary neighborhood information among samples of multi-omics data, and failing to consider the potential interactions between different omics data at the molecular level. In this paper, we propose a prognostic model for ovarian cancer prediction named Dual Fusion Channels and Stacked Graph Convolutional Neural Network (DFASGCNS). The DFASGCNS utilizes dual fusion channels to learn feature representations of different omics data and the associations between samples. Stacked graph convolutional network is used to comprehensively learn the deep and intricate correlation networks present in multi-omics data, enhancing the model's ability to represent multi-omics data. An attention mechanism is introduced to allocate different weights to important features of different omics data, optimizing the feature representation of multi-omics data. Experimental results demonstrate that compared to existing methods, the DFASGCNS model exhibits significant advantages in ovarian cancer prognosis prediction and survival analysis. Kaplan-Meier curve analysis results indicate significant differences in the survival subgroups predicted by the DFASGCNS model, contributing to a deeper understanding of the pathogenesis of ovarian cancer and providing more reliable auxiliary diagnostic information for the prognosis assessment of ovarian cancer patients.
Collapse
Affiliation(s)
- Huiqing Wang
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Xiao Han
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Shuaijun Niu
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Hao Cheng
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Jianxue Ren
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| | - Yimeng Duan
- College of Computer Science and Technology (College of Data Science), Taiyuan University of Technology, Taiyuan, China
| |
Collapse
|
19
|
Yeghaian M, Bodalal Z, Tareco Bucho T, Kurilova I, Blank C, Smit E, van der Heijden M, Nguyen-Kim T, van den Broek D, Beets-Tan R, Trebeschi S. Integrated noninvasive diagnostics for prediction of survival in immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100723. [PMID: 39185322 PMCID: PMC11342748 DOI: 10.1016/j.iotech.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Background Integrating complementary diagnostic data sources promises enhanced robustness in the predictive performance of artificial intelligence (AI) models, a crucial requirement for future clinical validation/implementation. In this study, we investigate the potential value of integrating data from noninvasive diagnostic modalities, including chest computed tomography (CT) imaging, routine laboratory blood tests, and clinical parameters, to retrospectively predict 1-year survival in a cohort of patients with advanced non-small-cell lung cancer, melanoma, and urothelial cancer treated with immunotherapy. Patients and methods The study included 475 patients, of whom 444 had longitudinal CT scans and 475 had longitudinal laboratory data. An ensemble of AI models was trained on data from each diagnostic modality, and subsequently, a model-agnostic integration approach was adopted for combining the prediction probabilities of each modality and producing an integrated decision. Results Integrating different diagnostic data demonstrated a modest increase in predictive performance. The highest area under the curve (AUC) was achieved by CT and laboratory data integration (AUC of 0.83, 95% confidence interval 0.81-0.85, P < 0.001), whereas the performance of individual models trained on laboratory and CT data independently yielded AUCs of 0.81 and 0.73, respectively. Conclusions In our retrospective cohort, integrating different noninvasive data modalities improved performance.
Collapse
Affiliation(s)
- M. Yeghaian
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Z. Bodalal
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - T.M. Tareco Bucho
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - I. Kurilova
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - C.U. Blank
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - E.F. Smit
- Pulmonology Department, Leiden University Medical Center, Leiden, The Netherlands
| | - M.S. van der Heijden
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - T.D.L. Nguyen-Kim
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Institute of Diagnostic and Interventional Radiology, University Hospital of Zurich, Zurich, Switzerland
| | - D. van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R.G.H. Beets-Tan
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - S. Trebeschi
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
20
|
Aslan AT, Akova M. Recent updates in treating carbapenem-resistant infections in patients with hematological malignancies. Expert Rev Anti Infect Ther 2024; 22:1055-1071. [PMID: 39313753 DOI: 10.1080/14787210.2024.2408746] [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] [Revised: 09/19/2024] [Accepted: 09/22/2024] [Indexed: 09/25/2024]
Abstract
INTRODUCTION Patients with hematological malignancies (PHMs) are at increased risk for infections caused by carbapenem-resistant organisms (CROs) due to frequent exposure to broad-spectrum antibiotics and prolonged hospital stays. These infections result in high mortality and morbidity rates along with delays in chemotherapy, longer hospitalizations, and increased health care costs. AREAS COVERED Treatment alternatives for CRO infections in PHMs. EXPERT OPINION The best available treatment option for KPC and OXA-48 producers is ceftazidime/avibactam. Imipenem/cilastatin/relebactam and meropenem/vaborbactam remain as the alternative options. They can also be used as salvage therapy in KPC-positive Enterobacterales infections resistant to ceftazidime/avibactam, if in vitro susceptibility is shown. Treatment of metallo-β-lactamase producers is an unmet need. Ceftazidime/avibactam plus aztreonam or aztreonam/avibactam seems to be the most reliable option for metallo-β-lactamase producers. As a first-line option for carbapenem-resistant Pseudomonas aeruginosa infections, ceftolozane/tazobactam is preferable and ceftazidime/avibactam and imipenem/cilastatin/relebactam constitute alternative regimens. Although sulbactam/durlobactam is the most reliable option against carbapenem-resistant Acinetobacter baumannii infections, its utility as monotherapy and in PHMs is not yet known. Cefiderocol can be selected as a 'last-resort' option for CRO infections. New risk score models supported by artificial intelligence algorithms can be used to predict the exact risk of infections in previously colonized patients.
Collapse
Affiliation(s)
- Abdullah Tarık Aslan
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Murat Akova
- Faculty of Medicine, Infectious Diseases and Clinical Microbiology, Hacettepe University, Ankara, Türkiye
| |
Collapse
|
21
|
Palmal S, Saha S, Arya N, Tripathy S. CAGCL: Predicting Short- and Long-Term Breast Cancer Survival With Cross-Modal Attention and Graph Contrastive Learning. IEEE J Biomed Health Inform 2024; 28:7382-7391. [PMID: 39236140 DOI: 10.1109/jbhi.2024.3449756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
In breast cancer treatment, accurately predicting how long a patient will survive is crucial for decision-making. This information guides treatment choices and supports patients' psychological recovery. To address this challenge, we introduce a novel predictive model to forecast breast cancer prognosis by leveraging diverse data sources, including clinical records, copy number variation, gene expressions, DNA methylation, microRNA (miRSeq) sequencing, and whole slide image data from the TCGA Database. The methodology incorporates graph contrastive learning with cross-modality attention (CAGCL), considering all possible combinations of the six distinct data modalities. Feature embeddings are enhanced through graph contrastive learning, which identifies subtle differences and similarities among samples. Further, to learn the complementary nature of information across multiple data modalities, a cross-attention framework is proposed and applied to the graph contrastive learning-based extracted features from various data sources for breast cancer survival prediction. It performs a binary classification to anticipate the likelihood of short- and long-term breast cancer survivors, delineated by a five-year threshold. The proposed model (CAGCL) showcases superior performance compared to baseline models and other state-of-the-art models. The model attains an accuracy of 0.932, a sensitivity of 0.954, a precision of 0.958, an F1 score of 0.956, and an AUC of 0.948, underscoring its effectiveness in predicting breast cancer survival.
Collapse
|
22
|
Yang H, Yang M, Chen J, Yao G, Zou Q, Jia L. Multimodal deep learning approaches for precision oncology: a comprehensive review. Brief Bioinform 2024; 26:bbae699. [PMID: 39757116 DOI: 10.1093/bib/bbae699] [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: 11/12/2024] [Revised: 12/02/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
The burgeoning accumulation of large-scale biomedical data in oncology, alongside significant strides in deep learning (DL) technologies, has established multimodal DL (MDL) as a cornerstone of precision oncology. This review provides an overview of MDL applications in this field, based on an extensive literature survey. In total, 651 articles published before September 2024 are included. We first outline publicly available multimodal datasets that support cancer research. Then, we discuss key DL training methods, data representation techniques, and fusion strategies for integrating multimodal data. The review also examines MDL applications in tumor segmentation, detection, diagnosis, prognosis, treatment selection, and therapy response monitoring. Finally, we critically assess the limitations of current approaches and propose directions for future research. By synthesizing current progress and identifying challenges, this review aims to guide future efforts in leveraging MDL to advance precision oncology.
Collapse
Affiliation(s)
- Huan Yang
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Chengdian Road, Kecheng District, Quzhou 324000, Zhejiang, China
| | - Minglei Yang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Jianshe Dong Road, Erqi District, Zhengzhou 450052, Henan, China
| | - Jiani Chen
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Chengdian Road, Kecheng District, Quzhou 324000, Zhejiang, China
- School of Opto-electronic and Communication Engineering, Xiamen University of Technology, Ligong Road, Jimei District, Xiamen 361024, Fujian, China
| | - Guocong Yao
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Chengdian Road, Kecheng District, Quzhou 324000, Zhejiang, China
- School of Computer and Information Engineering, Henan University, Jinming Avenue, Longting District, Kaifeng 475001, Henan, China
| | - Quan Zou
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Chengdian Road, Kecheng District, Quzhou 324000, Zhejiang, China
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Section2, North Jianshe Road, Chenghua District, Chengdu 610054, Sichuan, China
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Changchun Street, Xicheng District, Beijing 100053, China
| |
Collapse
|
23
|
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
|
24
|
Katayama A, Aoki Y, Watanabe Y, Horiguchi J, Rakha EA, Oyama T. Current status and prospects of artificial intelligence in breast cancer pathology: convolutional neural networks to prospective Vision Transformers. Int J Clin Oncol 2024; 29:1648-1668. [PMID: 38619651 DOI: 10.1007/s10147-024-02513-3] [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: 01/16/2024] [Accepted: 03/12/2024] [Indexed: 04/16/2024]
Abstract
Breast cancer is the most prevalent cancer among women, and its diagnosis requires the accurate identification and classification of histological features for effective patient management. Artificial intelligence, particularly through deep learning, represents the next frontier in cancer diagnosis and management. Notably, the use of convolutional neural networks and emerging Vision Transformers (ViT) has been reported to automate pathologists' tasks, including tumor detection and classification, in addition to improving the efficiency of pathology services. Deep learning applications have also been extended to the prediction of protein expression, molecular subtype, mutation status, therapeutic efficacy, and outcome prediction directly from hematoxylin and eosin-stained slides, bypassing the need for immunohistochemistry or genetic testing. This review explores the current status and prospects of deep learning in breast cancer diagnosis with a focus on whole-slide image analysis. Artificial intelligence applications are increasingly applied to many tasks in breast pathology ranging from disease diagnosis to outcome prediction, thus serving as valuable tools for assisting pathologists and supporting breast cancer management.
Collapse
Affiliation(s)
- Ayaka Katayama
- Diagnostic Pathology, Gunma University Graduate School of Medicine, 3-39-22 Showamachi, Maebashi, Gunma, 371-8511, Japan.
| | - Yuki Aoki
- Center for Mathematics and Data Science, Gunma University, Maebashi, Japan
| | - Yukako Watanabe
- Clinical Training Center, Gunma University Hospital, Maebashi, Japan
| | - Jun Horiguchi
- Department of Breast Surgery, International University of Health and Welfare, Narita, Japan
| | - Emad A Rakha
- Department of Histopathology School of Medicine, University of Nottingham, University Park, Nottingham, UK
- Department of Pathology, Hamad Medical Corporation, Doha, Qatar
| | - Tetsunari Oyama
- Diagnostic Pathology, Gunma University Graduate School of Medicine, 3-39-22 Showamachi, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
25
|
Hansen ER, Sagi T, Hose K. Multimodal representation learning for medical analytics - a systematic literature review. Health Informatics J 2024; 30:14604582241290474. [PMID: 39511041 DOI: 10.1177/14604582241290474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Objectives: Machine learning-based analytics over uni-modal medical data has shown considerable promise and is now routinely deployed in diagnostic procedures. However, patient data consists of diverse types of data. By exploiting such data, multimodal approaches promise to revolutionize our ability to provide personalized care. Attempts to combine two modalities in a single diagnostic task have utilized the evolving field of multimodal representation learning (MRL), which learns a shared latent space between related modality samples. This new space can be used to improve the performance of machine-learning-based analytics. So far, however, our understanding of how modalities have been applied in MRL-based medical applications and which modalities are best suited for specific medical tasks is still unclear, as previous reviews have not addressed the medical analytics domain and its unique challenges and opportunities. Instead, this work aims to review the landscape of MRL for medical tasks to highlight opportunities for advancing medical applications. Methods: This paper presents a framework for positioning MRL techniques and medical modalities. More than 1000 papers related to medical analytics were reviewed, positioned, and classified using the proposed framework in the most extensive review to date. The paper further provides an online tool for researchers and developers of medical analytics to dive into the rapidly changing landscape of MRL for medical applications. Results: The main finding is that work in the domain has been sparse: only a few medical informatics tasks have been the target of much MRL-based work, with the overwhelming majority of tasks being diagnostic rather than prognostic. Similarly, numerous potentially compatible information modality combinations are unexplored or under-explored for most medical tasks. Conclusions: There is much to gain from using MRL in many unexplored combinations of medical tasks and modalities. This work can guide researchers working on a specific medical application to identify under-explored modality combinations and identify novel and emerging MRL techniques that can be adapted to the task at hand.
Collapse
Affiliation(s)
- Emil Riis Hansen
- Department of Computer Science, Aalborg University, Aalborg, Denmark
| | - Tomer Sagi
- Department of Computer Science, Aalborg University, Aalborg, Denmark
| | - Katja Hose
- Department of Computer Science, Aalborg University, Aalborg, Denmark; Department of Informatics, TU Wien, Wien, Austria
| |
Collapse
|
26
|
Vorontsov E, Bozkurt A, Casson A, Shaikovski G, Zelechowski M, Severson K, Zimmermann E, Hall J, Tenenholtz N, Fusi N, Yang E, Mathieu P, van Eck A, Lee D, Viret J, Robert E, Wang YK, Kunz JD, Lee MCH, Bernhard JH, Godrich RA, Oakley G, Millar E, Hanna M, Wen H, Retamero JA, Moye WA, Yousfi R, Kanan C, Klimstra DS, Rothrock B, Liu S, Fuchs TJ. A foundation model for clinical-grade computational pathology and rare cancers detection. Nat Med 2024; 30:2924-2935. [PMID: 39039250 PMCID: PMC11485232 DOI: 10.1038/s41591-024-03141-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/19/2024] [Indexed: 07/24/2024]
Abstract
The analysis of histopathology images with artificial intelligence aims to enable clinical decision support systems and precision medicine. The success of such applications depends on the ability to model the diverse patterns observed in pathology images. To this end, we present Virchow, the largest foundation model for computational pathology to date. In addition to the evaluation of biomarker prediction and cell identification, we demonstrate that a large foundation model enables pan-cancer detection, achieving 0.95 specimen-level area under the (receiver operating characteristic) curve across nine common and seven rare cancers. Furthermore, we show that with less training data, the pan-cancer detector built on Virchow can achieve similar performance to tissue-specific clinical-grade models in production and outperform them on some rare variants of cancer. Virchow's performance gains highlight the value of a foundation model and open possibilities for many high-impact applications with limited amounts of labeled training data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Ellen Yang
- Memorial Sloan Kettering Cancer Center, New York, NY, US
| | | | | | | | | | | | | | | | | | | | | | | | - Ewan Millar
- NSW Health Pathology, St George Hospital, Sydney, New South Wales, Australia
| | - Matthew Hanna
- Memorial Sloan Kettering Cancer Center, New York, NY, US
| | - Hannah Wen
- Memorial Sloan Kettering Cancer Center, New York, NY, US
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Kook L, Lundborg AR. Algorithm-agnostic significance testing in supervised learning with multimodal data. Brief Bioinform 2024; 25:bbae475. [PMID: 39323092 PMCID: PMC11424510 DOI: 10.1093/bib/bbae475] [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: 05/30/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
MOTIVATION Valid statistical inference is crucial for decision-making but difficult to obtain in supervised learning with multimodal data, e.g. combinations of clinical features, genomic data, and medical images. Multimodal data often warrants the use of black-box algorithms, for instance, random forests or neural networks, which impede the use of traditional variable significance tests. RESULTS We address this problem by proposing the use of COvariance MEasure Tests (COMETs), which are calibrated and powerful tests that can be combined with any sufficiently predictive supervised learning algorithm. We apply COMETs to several high-dimensional, multimodal data sets to illustrate (i) variable significance testing for finding relevant mutations modulating drug-activity, (ii) modality selection for predicting survival in liver cancer patients with multiomics data, and (iii) modality selection with clinical features and medical imaging data. In all applications, COMETs yield results consistent with domain knowledge without requiring data-driven pre-processing, which may invalidate type I error control. These novel applications with high-dimensional multimodal data corroborate prior results on the power and robustness of COMETs for significance testing. AVAILABILITY AND IMPLEMENTATION COMETs are implemented in the cometsR package available on CRAN and pycometsPython library available on GitHub. Source code for reproducing all results is available at https://github.com/LucasKook/comets. All data sets used in this work are openly available.
Collapse
Affiliation(s)
- Lucas Kook
- Institute for Statistics and Mathematics, Vienna University of Economics and Business, Welthandelsplatz 1, AT-1020 Vienna, Austria
| | - Anton Rask Lundborg
- Department of Mathematical Sciences, University of Copenhagen, Universitetsparken 5, DK-2100 Copenhagen, Denmark
| |
Collapse
|
28
|
Ogundipe O, Kurt Z, Woo WL. Deep neural networks integrating genomics and histopathological images for predicting stages and survival time-to-event in colon cancer. PLoS One 2024; 19:e0305268. [PMID: 39226289 PMCID: PMC11371203 DOI: 10.1371/journal.pone.0305268] [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: 09/15/2023] [Accepted: 05/26/2024] [Indexed: 09/05/2024] Open
Abstract
MOTIVATION There exists an unexplained diverse variation within the predefined colon cancer stages using only features from either genomics or histopathological whole slide images as prognostic factors. Unraveling this variation will bring about improved staging and treatment outcomes. Hence, motivated by the advancement of Deep Neural Network (DNN) libraries and complementary factors within some genomics datasets, we aggregate atypia patterns in histopathological images with diverse carcinogenic expression from mRNA, miRNA and DNA methylation as an integrative input source into a deep neural network for colon cancer stages classification, and samples stratification into low or high-risk survival groups. RESULTS The genomics-only and integrated input features return Area Under Curve-Receiver Operating Characteristic curve (AUC-ROC) of 0.97 compared with AUC-ROC of 0.78 obtained when only image features are used for the stage's classification. A further analysis of prediction accuracy using the confusion matrix shows that the integrated features have a weakly improved accuracy of 0.08% more than the accuracy obtained with genomics features. Also, the extracted features were used to split the patients into low or high-risk survival groups. Among the 2,700 fused features, 1,836 (68%) features showed statistically significant survival probability differences in aggregating samples into either low or high between the two risk survival groups. Availability and Implementation: https://github.com/Ogundipe-L/EDCNN.
Collapse
Affiliation(s)
- Olalekan Ogundipe
- Department of Computer and Information Sciences, University of Northumbria, Newcastle Upon Tyne, United Kingdom
| | - Zeyneb Kurt
- Information School, University of Sheffield, Sheffield, United Kingdom
| | - Wai Lok Woo
- Department of Computer and Information Sciences, University of Northumbria, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
29
|
Wang J, Liu G, Zhou C, Cui X, Wang W, Wang J, Huang Y, Jiang J, Wang Z, Tang Z, Zhang A, Cui D. Application of artificial intelligence in cancer diagnosis and tumor nanomedicine. NANOSCALE 2024; 16:14213-14246. [PMID: 39021117 DOI: 10.1039/d4nr01832j] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Cancer is a major health concern due to its high incidence and mortality rates. Advances in cancer research, particularly in artificial intelligence (AI) and deep learning, have shown significant progress. The swift evolution of AI in healthcare, especially in tools like computer-aided diagnosis, has the potential to revolutionize early cancer detection. This technology offers improved speed, accuracy, and sensitivity, bringing a transformative impact on cancer diagnosis, treatment, and management. This paper provides a concise overview of the application of artificial intelligence in the realms of medicine and nanomedicine, with a specific emphasis on the significance and challenges associated with cancer diagnosis. It explores the pivotal role of AI in cancer diagnosis, leveraging structured, unstructured, and multimodal fusion data. Additionally, the article delves into the applications of AI in nanomedicine sensors and nano-oncology drugs. The fundamentals of deep learning and convolutional neural networks are clarified, underscoring their relevance to AI-driven cancer diagnosis. A comparative analysis is presented, highlighting the accuracy and efficiency of traditional methods juxtaposed with AI-based approaches. The discussion not only assesses the current state of AI in cancer diagnosis but also delves into the challenges faced by AI in this context. Furthermore, the article envisions the future development direction and potential application of artificial intelligence in cancer diagnosis, offering a hopeful prospect for enhanced cancer detection and improved patient prognosis.
Collapse
Affiliation(s)
- Junhao Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Guan Liu
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Cheng Zhou
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Xinyuan Cui
- Imaging Department of Rui Jin Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Jiulin Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Yixin Huang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Jinlei Jiang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Zhitao Wang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Zengyi Tang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Amin Zhang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, China.
| | - Daxiang Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China.
- School of Medicine, Henan University, Henan, China
| |
Collapse
|
30
|
Zhang G, Ma C, Yan C, Luo H, Wang J, Liang W, Luo J. MSFN: a multi-omics stacked fusion network for breast cancer survival prediction. Front Genet 2024; 15:1378809. [PMID: 39161422 PMCID: PMC11331006 DOI: 10.3389/fgene.2024.1378809] [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: 01/30/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Introduction: Developing effective breast cancer survival prediction models is critical to breast cancer prognosis. With the widespread use of next-generation sequencing technologies, numerous studies have focused on survival prediction. However, previous methods predominantly relied on single-omics data, and survival prediction using multi-omics data remains a significant challenge. Methods: In this study, considering the similarity of patients and the relevance of multi-omics data, we propose a novel multi-omics stacked fusion network (MSFN) based on a stacking strategy to predict the survival of breast cancer patients. MSFN first constructs a patient similarity network (PSN) and employs a residual graph neural network (ResGCN) to obtain correlative prognostic information from PSN. Simultaneously, it employs convolutional neural networks (CNNs) to obtain specificity prognostic information from multi-omics data. Finally, MSFN stacks the prognostic information from these networks and feeds into AdaboostRF for survival prediction. Results: Experiments results demonstrated that our method outperformed several state-of-the-art methods, and biologically validated by Kaplan-Meier and t-SNE.
Collapse
Affiliation(s)
- Ge Zhang
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, Henan, China
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, Henan, China
| | - Chenwei Ma
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
| | - Chaokun Yan
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, Henan, China
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, Henan, China
| | - Huimin Luo
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, Henan, China
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, Henan, China
| | - Jianlin Wang
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, Henan, China
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, Henan, China
| | - Wenjuan Liang
- Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng, Henan, China
- School of Computer and Information Engineering, Henan University, Kaifeng, Henan, China
- Henan Key Laboratory of Big Data Analysis and Processing, Henan University, Kaifeng, Henan, China
| | - Junwei Luo
- College of Computer Science and Technology, Henan Polytechnic University, Jiaozuo, Henan, China
| |
Collapse
|
31
|
Liu F. Data Science Methods for Real-World Evidence Generation in Real-World Data. Annu Rev Biomed Data Sci 2024; 7:201-224. [PMID: 38748863 DOI: 10.1146/annurev-biodatasci-102423-113220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
In the healthcare landscape, data science (DS) methods have emerged as indispensable tools to harness real-world data (RWD) from various data sources such as electronic health records, claim and registry data, and data gathered from digital health technologies. Real-world evidence (RWE) generated from RWD empowers researchers, clinicians, and policymakers with a more comprehensive understanding of real-world patient outcomes. Nevertheless, persistent challenges in RWD (e.g., messiness, voluminousness, heterogeneity, multimodality) and a growing awareness of the need for trustworthy and reliable RWE demand innovative, robust, and valid DS methods for analyzing RWD. In this article, I review some common current DS methods for extracting RWE and valuable insights from complex and diverse RWD. This article encompasses the entire RWE-generation pipeline, from study design with RWD to data preprocessing, exploratory analysis, methods for analyzing RWD, and trustworthiness and reliability guarantees, along with data ethics considerations and open-source tools. This review, tailored for an audience that may not be experts in DS, aspires to offer a systematic review of DS methods and assists readers in selecting suitable DS methods and enhancing the process of RWE generation for addressing their specific challenges.
Collapse
Affiliation(s)
- Fang Liu
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, Indiana, USA;
| |
Collapse
|
32
|
Chowdhury A, Thway K, Pasquali S, Callegaro D, Gronchi A, Jones RL, Huang PH. Opportunities and Challenges in Soft Tissue Sarcoma Risk Stratification in the Era of Personalised Medicine. Curr Treat Options Oncol 2024; 25:1124-1135. [PMID: 39080193 DOI: 10.1007/s11864-024-01244-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] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 08/17/2024]
Abstract
OPINION STATEMENT Soft tissue sarcomas (STS) are a rare and heterogeneous group of cancers. Treatment options have changed little in the past thirty years, and the role of neoadjuvant chemotherapy is controversial. Accurate risk stratification is crucial in STS in order to facilitate clinical discussions around peri-operative treatment. Current risk stratification tools used in clinic, such as Sarculator, use clinicopathological characteristics and may be specific to anatomical site or to histology. More recently, risk stratification tools have been developed using molecular or immunological data. Combining Sarculator with other risk stratification tools may identify novel patient groups with differential clinical outcomes. There are several considerations when translating risk stratification tools into widespread clinical use, including establishing clinical utility, health economic value, being applicable to existing clinical pathways, having strong real-world performance, and being supported by investment into infrastructure. Future work may include incorporation of novel modalities and data integration techniques.
Collapse
Affiliation(s)
- Avirup Chowdhury
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK
| | - Khin Thway
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Sandro Pasquali
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Dario Callegaro
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Robin L Jones
- The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Paul H Huang
- Division of Molecular Pathology, The Institute of Cancer Research, 15 Cotswold Road Sutton, London, SM2 5NG, UK.
| |
Collapse
|
33
|
Mukund A, Afridi MA, Karolak A, Park MA, Permuth JB, Rasool G. Pancreatic Ductal Adenocarcinoma (PDAC): A Review of Recent Advancements Enabled by Artificial Intelligence. Cancers (Basel) 2024; 16:2240. [PMID: 38927945 PMCID: PMC11201559 DOI: 10.3390/cancers16122240] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most formidable challenges in oncology, characterized by its late detection and poor prognosis. Artificial intelligence (AI) and machine learning (ML) are emerging as pivotal tools in revolutionizing PDAC care across various dimensions. Consequently, many studies have focused on using AI to improve the standard of PDAC care. This review article attempts to consolidate the literature from the past five years to identify high-impact, novel, and meaningful studies focusing on their transformative potential in PDAC management. Our analysis spans a broad spectrum of applications, including but not limited to patient risk stratification, early detection, and prediction of treatment outcomes, thereby highlighting AI's potential role in enhancing the quality and precision of PDAC care. By categorizing the literature into discrete sections reflective of a patient's journey from screening and diagnosis through treatment and survivorship, this review offers a comprehensive examination of AI-driven methodologies in addressing the multifaceted challenges of PDAC. Each study is summarized by explaining the dataset, ML model, evaluation metrics, and impact the study has on improving PDAC-related outcomes. We also discuss prevailing obstacles and limitations inherent in the application of AI within the PDAC context, offering insightful perspectives on potential future directions and innovations.
Collapse
Affiliation(s)
- Ashwin Mukund
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| | - Muhammad Ali Afridi
- School of Electrical Engineering and Computer Science (SEECS), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan;
| | - Aleksandra Karolak
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| | - Margaret A. Park
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (M.A.P.); (J.B.P.)
| | - Jennifer B. Permuth
- Departments of Cancer Epidemiology and Gastrointestinal Oncology, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (M.A.P.); (J.B.P.)
| | - Ghulam Rasool
- Department of Machine Learning, Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA; (A.M.); (A.K.)
| |
Collapse
|
34
|
Wang T, Chen H, Chen Z, Li M, Lu Y. Prediction model of early recurrence of multimodal hepatocellular carcinoma with tensor fusion. Phys Med Biol 2024; 69:125003. [PMID: 38776945 DOI: 10.1088/1361-6560/ad4f45] [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: 12/24/2023] [Accepted: 05/22/2024] [Indexed: 05/25/2024]
Abstract
Objective.In oncology, clinical decision-making relies on a multitude of data modalities, including histopathological, radiological, and clinical factors. Despite the emergence of computer-aided multimodal decision-making systems for predicting hepatocellular carcinoma (HCC) recurrence post-hepatectomy, existing models often employ simplistic feature-level concatenation, leading to redundancy and suboptimal performance. Moreover, these models frequently lack effective integration with clinically relevant data and encounter challenges in integrating diverse scales and dimensions, as well as incorporating the liver background, which holds clinical significance but has been previously overlooked.Approach.To address these limitations, we propose two approaches. Firstly, we introduce the tensor fusion method to our model, which offers distinct advantages in handling multi-scale and multi-dimensional data fusion, potentially enhancing overall performance. Secondly, we pioneer the consideration of the liver background's impact, integrating it into the feature extraction process using a deep learning segmentation-based algorithm. This innovative inclusion aligns the model more closely with real-world clinical scenarios, as the liver background may contain crucial information related to postoperative recurrence.Main results.We collected radiomics (MRI) and histopathological images from 176 cases diagnosed by experienced clinicians across two independent centers. Our proposed network underwent training and 5-fold cross-validation on this dataset before validation on an external test dataset comprising 40 cases. Ultimately, our model demonstrated outstanding performance in predicting early recurrence of HCC postoperatively, achieving an AUC of 0.883.Significance.These findings signify significant progress in addressing challenges related to multimodal data fusion and hold promise for more accurate clinical outcome predictions. In this study, we exploited global 3D liver background into modelling which is crucial to to the prognosis assessment and analyzed the whole liver background in addition to the tumor region. Both MRI images and histopathological images of HCC were fused at high-dimensional feature space using tensor techniques to solve cross-scale data integration issue.
Collapse
Affiliation(s)
- Tianyi Wang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Haimei Chen
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zebin Chen
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Mingkai Li
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yao Lu
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
35
|
Wang Z, Lin R, Li Y, Zeng J, Chen Y, Ouyang W, Li H, Jia X, Lai Z, Yu Y, Yao H, Su W. Deep learning-based multi-modal data integration enhancing breast cancer disease-free survival prediction. PRECISION CLINICAL MEDICINE 2024; 7:pbae012. [PMID: 38912415 DOI: 10.1093/pcmedi] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND The prognosis of breast cancer is often unfavorable, emphasizing the need for early metastasis risk detection and accurate treatment predictions. This study aimed to develop a novel multi-modal deep learning model using preoperative data to predict disease-free survival (DFS). METHODS We retrospectively collected pathology imaging, molecular and clinical data from The Cancer Genome Atlas and one independent institution in China. We developed a novel Deep Learning Clinical Medicine Based Pathological Gene Multi-modal (DeepClinMed-PGM) model for DFS prediction, integrating clinicopathological data with molecular insights. The patients included the training cohort (n = 741), internal validation cohort (n = 184), and external testing cohort (n = 95). RESULT Integrating multi-modal data into the DeepClinMed-PGM model significantly improved area under the receiver operating characteristic curve (AUC) values. In the training cohort, AUC values for 1-, 3-, and 5-year DFS predictions increased to 0.979, 0.957, and 0.871, while in the external testing cohort, the values reached 0.851, 0.878, and 0.938 for 1-, 2-, and 3-year DFS predictions, respectively. The DeepClinMed-PGM's robust discriminative capabilities were consistently evident across various cohorts, including the training cohort [hazard ratio (HR) 0.027, 95% confidence interval (CI) 0.0016-0.046, P < 0.0001], the internal validation cohort (HR 0.117, 95% CI 0.041-0.334, P < 0.0001), and the external cohort (HR 0.061, 95% CI 0.017-0.218, P < 0.0001). Additionally, the DeepClinMed-PGM model demonstrated C-index values of 0.925, 0.823, and 0.864 within the three cohorts, respectively. CONCLUSION This study introduces an approach to breast cancer prognosis, integrating imaging and molecular and clinical data for enhanced predictive accuracy, offering promise for personalized treatment strategies.
Collapse
Affiliation(s)
- Zehua Wang
- Guangdong Key Laboratory of Cross-Application of Data Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai 519087, China
| | - Ruichong Lin
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao 999078, China
- Department of Computer and Information Engineering, Guangzhou Huali College, Guangzhou 511325, China
| | - Yanchun Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jin Zeng
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Han Li
- The Second Clinical Medical College, Southern Medical University, Guangzhou 510515, China
| | - Xueyan Jia
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Zijia Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Weifeng Su
- Guangdong Key Laboratory of Cross-Application of Data Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai 519087, China
| |
Collapse
|
36
|
Wang Z, Lin R, Li Y, Zeng J, Chen Y, Ouyang W, Li H, Jia X, Lai Z, Yu Y, Yao H, Su W. Deep learning-based multi-modal data integration enhancing breast cancer disease-free survival prediction. PRECISION CLINICAL MEDICINE 2024; 7:pbae012. [PMID: 38912415 PMCID: PMC11190375 DOI: 10.1093/pcmedi/pbae012] [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: 02/21/2024] [Revised: 05/19/2024] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND The prognosis of breast cancer is often unfavorable, emphasizing the need for early metastasis risk detection and accurate treatment predictions. This study aimed to develop a novel multi-modal deep learning model using preoperative data to predict disease-free survival (DFS). METHODS We retrospectively collected pathology imaging, molecular and clinical data from The Cancer Genome Atlas and one independent institution in China. We developed a novel Deep Learning Clinical Medicine Based Pathological Gene Multi-modal (DeepClinMed-PGM) model for DFS prediction, integrating clinicopathological data with molecular insights. The patients included the training cohort (n = 741), internal validation cohort (n = 184), and external testing cohort (n = 95). RESULT Integrating multi-modal data into the DeepClinMed-PGM model significantly improved area under the receiver operating characteristic curve (AUC) values. In the training cohort, AUC values for 1-, 3-, and 5-year DFS predictions increased to 0.979, 0.957, and 0.871, while in the external testing cohort, the values reached 0.851, 0.878, and 0.938 for 1-, 2-, and 3-year DFS predictions, respectively. The DeepClinMed-PGM's robust discriminative capabilities were consistently evident across various cohorts, including the training cohort [hazard ratio (HR) 0.027, 95% confidence interval (CI) 0.0016-0.046, P < 0.0001], the internal validation cohort (HR 0.117, 95% CI 0.041-0.334, P < 0.0001), and the external cohort (HR 0.061, 95% CI 0.017-0.218, P < 0.0001). Additionally, the DeepClinMed-PGM model demonstrated C-index values of 0.925, 0.823, and 0.864 within the three cohorts, respectively. CONCLUSION This study introduces an approach to breast cancer prognosis, integrating imaging and molecular and clinical data for enhanced predictive accuracy, offering promise for personalized treatment strategies.
Collapse
Affiliation(s)
- Zehua Wang
- Guangdong Key Laboratory of Cross-Application of Data Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai 519087, China
| | - Ruichong Lin
- Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao 999078, China
- Department of Computer and Information Engineering, Guangzhou Huali College, Guangzhou 511325, China
| | - Yanchun Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jin Zeng
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm 17177, Sweden
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Han Li
- The Second Clinical Medical College, Southern Medical University, Guangzhou 510515, China
| | - Xueyan Jia
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Zijia Lai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Weifeng Su
- Guangdong Key Laboratory of Cross-Application of Data Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai 519087, China
| |
Collapse
|
37
|
Er AG, Ding DY, Er B, Uzun M, Cakmak M, Sadee C, Durhan G, Ozmen MN, Tanriover MD, Topeli A, Aydin Son Y, Tibshirani R, Unal S, Gevaert O. Multimodal data fusion using sparse canonical correlation analysis and cooperative learning: a COVID-19 cohort study. NPJ Digit Med 2024; 7:117. [PMID: 38714751 PMCID: PMC11076490 DOI: 10.1038/s41746-024-01128-2] [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: 11/06/2023] [Accepted: 04/25/2024] [Indexed: 05/10/2024] Open
Abstract
Through technological innovations, patient cohorts can be examined from multiple views with high-dimensional, multiscale biomedical data to classify clinical phenotypes and predict outcomes. Here, we aim to present our approach for analyzing multimodal data using unsupervised and supervised sparse linear methods in a COVID-19 patient cohort. This prospective cohort study of 149 adult patients was conducted in a tertiary care academic center. First, we used sparse canonical correlation analysis (CCA) to identify and quantify relationships across different data modalities, including viral genome sequencing, imaging, clinical data, and laboratory results. Then, we used cooperative learning to predict the clinical outcome of COVID-19 patients: Intensive care unit admission. We show that serum biomarkers representing severe disease and acute phase response correlate with original and wavelet radiomics features in the LLL frequency channel (cor(Xu1, Zv1) = 0.596, p value < 0.001). Among radiomics features, histogram-based first-order features reporting the skewness, kurtosis, and uniformity have the lowest negative, whereas entropy-related features have the highest positive coefficients. Moreover, unsupervised analysis of clinical data and laboratory results gives insights into distinct clinical phenotypes. Leveraging the availability of global viral genome databases, we demonstrate that the Word2Vec natural language processing model can be used for viral genome encoding. It not only separates major SARS-CoV-2 variants but also allows the preservation of phylogenetic relationships among them. Our quadruple model using Word2Vec encoding achieves better prediction results in the supervised task. The model yields area under the curve (AUC) and accuracy values of 0.87 and 0.77, respectively. Our study illustrates that sparse CCA analysis and cooperative learning are powerful techniques for handling high-dimensional, multimodal data to investigate multivariate associations in unsupervised and supervised tasks.
Collapse
Affiliation(s)
- Ahmet Gorkem Er
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey.
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey.
| | - Daisy Yi Ding
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Berrin Er
- Department of Internal Medicine, Division of Intensive Care Medicine, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Mertcan Uzun
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Mehmet Cakmak
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Christoph Sadee
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Gamze Durhan
- Department of Radiology, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Mustafa Nasuh Ozmen
- Department of Radiology, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Mine Durusu Tanriover
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Arzu Topeli
- Department of Internal Medicine, Division of Intensive Care Medicine, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Yesim Aydin Son
- Department of Health Informatics, Graduate School of Informatics, Middle East Technical University, 06800, Ankara, Turkey
| | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
- Department of Statistics, Stanford University, Stanford, CA, 94305, USA
| | - Serhat Unal
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University Faculty of Medicine, 06230, Ankara, Turkey
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research (BMIR), Department of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
38
|
Wu H, Peng L, Du D, Xu H, Lin G, Zhou Z, Lu L, Lv W. BAF-Net: bidirectional attention-aware fluid pyramid feature integrated multimodal fusion network for diagnosis and prognosis. Phys Med Biol 2024; 69:105007. [PMID: 38593831 DOI: 10.1088/1361-6560/ad3cb2] [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: 12/27/2023] [Accepted: 04/09/2024] [Indexed: 04/11/2024]
Abstract
Objective. To go beyond the deficiencies of the three conventional multimodal fusion strategies (i.e. input-, feature- and output-level fusion), we propose a bidirectional attention-aware fluid pyramid feature integrated fusion network (BAF-Net) with cross-modal interactions for multimodal medical image diagnosis and prognosis.Approach. BAF-Net is composed of two identical branches to preserve the unimodal features and one bidirectional attention-aware distillation stream to progressively assimilate cross-modal complements and to learn supplementary features in both bottom-up and top-down processes. Fluid pyramid connections were adopted to integrate the hierarchical features at different levels of the network, and channel-wise attention modules were exploited to mitigate cross-modal cross-level incompatibility. Furthermore, depth-wise separable convolution was introduced to fuse the cross-modal cross-level features to alleviate the increase in parameters to a great extent. The generalization abilities of BAF-Net were evaluated in terms of two clinical tasks: (1) an in-house PET-CT dataset with 174 patients for differentiation between lung cancer and pulmonary tuberculosis. (2) A public multicenter PET-CT head and neck cancer dataset with 800 patients from nine centers for overall survival prediction.Main results. On the LC-PTB dataset, improved performance was found in BAF-Net (AUC = 0.7342) compared with input-level fusion model (AUC = 0.6825;p< 0.05), feature-level fusion model (AUC = 0.6968;p= 0.0547), output-level fusion model (AUC = 0.7011;p< 0.05). On the H&N cancer dataset, BAF-Net (C-index = 0.7241) outperformed the input-, feature-, and output-level fusion model, with 2.95%, 3.77%, and 1.52% increments of C-index (p= 0.3336, 0.0479 and 0.2911, respectively). The ablation experiments demonstrated the effectiveness of all the designed modules regarding all the evaluated metrics in both datasets.Significance. Extensive experiments on two datasets demonstrated better performance and robustness of BAF-Net than three conventional fusion strategies and PET or CT unimodal network in terms of diagnosis and prognosis.
Collapse
Affiliation(s)
- Huiqin Wu
- Department of Medical Imaging, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, 518037, People's Republic of China
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Lihong Peng
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Dongyang Du
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Hui Xu
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Guoyu Lin
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Zidong Zhou
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
| | - Lijun Lu
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, Guangdong, 510515, People's Republic of China
- Pazhou Lab, Guangzhou, Guangdong, 510330, People's Republic of China
| | - Wenbing Lv
- School of Information and Yunnan Key Laboratory of Intelligent Systems and Computing, Yunnan University, Kunming, Yunnan, 650504, People's Republic of China
| |
Collapse
|
39
|
Qiu L, Zhao L, Zhao W, Zhao J. Dual-space disentangled-multimodal network (DDM-net) for glioma diagnosis and prognosis with incomplete pathology and genomic data. Phys Med Biol 2024; 69:085028. [PMID: 38595094 DOI: 10.1088/1361-6560/ad37ec] [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: 11/14/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024]
Abstract
Objective. Effective fusion of histology slides and molecular profiles from genomic data has shown great potential in the diagnosis and prognosis of gliomas. However, it remains challenging to explicitly utilize the consistent-complementary information among different modalities and create comprehensive representations of patients. Additionally, existing researches mainly focus on complete multi-modality data and usually fail to construct robust models for incomplete samples.Approach. In this paper, we propose adual-space disentangled-multimodal network (DDM-net)for glioma diagnosis and prognosis. DDM-net disentangles the latent features generated by two separate variational autoencoders (VAEs) into common and specific components through a dual-space disentangled approach, facilitating the construction of comprehensive representations of patients. More importantly, DDM-net imputes the unavailable modality in the latent feature space, making it robust to incomplete samples.Main results. We evaluated our approach on the TCGA-GBMLGG dataset for glioma grading and survival analysis tasks. Experimental results demonstrate that the proposed method achieves superior performance compared to state-of-the-art methods, with a competitive AUC of 0.952 and a C-index of 0.768.Significance. The proposed model may help the clinical understanding of gliomas and can serve as an effective fusion model with multimodal data. Additionally, it is capable of handling incomplete samples, making it less constrained by clinical limitations.
Collapse
Affiliation(s)
- Lu Qiu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Lu Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Wangyuan Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Jun Zhao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| |
Collapse
|
40
|
Pan L, Peng Y, Li Y, Wang X, Liu W, Xu L, Liang Q, Peng S. SELECTOR: Heterogeneous graph network with convolutional masked autoencoder for multimodal robust prediction of cancer survival. Comput Biol Med 2024; 172:108301. [PMID: 38492453 DOI: 10.1016/j.compbiomed.2024.108301] [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: 11/01/2023] [Revised: 02/03/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
Accurately predicting the survival rate of cancer patients is crucial for aiding clinicians in planning appropriate treatment, reducing cancer-related medical expenses, and significantly enhancing patients' quality of life. Multimodal prediction of cancer patient survival offers a more comprehensive and precise approach. However, existing methods still grapple with challenges related to missing multimodal data and information interaction within modalities. This paper introduces SELECTOR, a heterogeneous graph-aware network based on convolutional mask encoders for robust multimodal prediction of cancer patient survival. SELECTOR comprises feature edge reconstruction, convolutional mask encoder, feature cross-fusion, and multimodal survival prediction modules. Initially, we construct a multimodal heterogeneous graph and employ the meta-path method for feature edge reconstruction, ensuring comprehensive incorporation of feature information from graph edges and effective embedding of nodes. To mitigate the impact of missing features within the modality on prediction accuracy, we devised a convolutional masked autoencoder (CMAE) to process the heterogeneous graph post-feature reconstruction. Subsequently, the feature cross-fusion module facilitates communication between modalities, ensuring that output features encompass all features of the modality and relevant information from other modalities. Extensive experiments and analysis on six cancer datasets from TCGA demonstrate that our method significantly outperforms state-of-the-art methods in both modality-missing and intra-modality information-confirmed cases. Our codes are made available at https://github.com/panliangrui/Selector.
Collapse
Affiliation(s)
- Liangrui Pan
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410083, Hunan, China.
| | - Yijun Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410083, Hunan, China.
| | - Yan Li
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410083, Hunan, China.
| | - Xiang Wang
- Department of Thoracic Surgery, The second xiangya hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Wenjuan Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410083, Hunan, China.
| | - Liwen Xu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410083, Hunan, China.
| | - Qingchun Liang
- Department of Pathology, The second xiangya hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, 410083, Hunan, China.
| |
Collapse
|
41
|
D'Souza NS, Wang H, Giovannini A, Foncubierta-Rodriguez A, Beck KL, Boyko O, Syeda-Mahmood TF. Fusing modalities by multiplexed graph neural networks for outcome prediction from medical data and beyond. Med Image Anal 2024; 93:103064. [PMID: 38219500 DOI: 10.1016/j.media.2023.103064] [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: 04/02/2023] [Revised: 09/09/2023] [Accepted: 12/11/2023] [Indexed: 01/16/2024]
Abstract
With the emergence of multimodal electronic health records, the evidence for diseases, events, or findings may be present across multiple modalities ranging from clinical to imaging and genomic data. Developing effective patient-tailored therapeutic guidance and outcome prediction will require fusing evidence across these modalities. Developing general-purpose frameworks capable of modeling fine-grained and multi-faceted complex interactions, both within and across modalities is an important open problem in multimodal fusion. Generalized multimodal fusion is extremely challenging as evidence for outcomes may not be uniform across all modalities, not all modality features may be relevant, or not all modalities may be present for all patients, due to which simple methods of early, late, or intermediate fusion may be inadequate. In this paper, we present a novel approach that uses the machinery of multiplexed graphs for fusion. This allows for modalities to be represented through their targeted encodings. We model their relationship between explicitly via multiplexed graphs derived from salient features in a combined latent space. We then derive a new graph neural network for multiplex graphs for task-informed reasoning. We compare our framework against several state-of-the-art approaches for multi-graph reasoning and multimodal fusion. As a sanity check on the neural network design, we evaluate the multiplexed GNN on two popular benchmark datasets, namely the AIFB and the MUTAG dataset against several state-of-the-art multi-relational GNNs for reasoning. Second, we evaluate our multiplexed framework against several state-of-the-art multimodal fusion frameworks on two large clinical datasets for two separate applications. The first is the NIH-TB portals dataset for treatment outcome prediction in Tuberculosis, and the second is the ABIDE dataset for Autism Spectrum Disorder classification. Through rigorous experimental evaluation, we demonstrate that the multiplexed GNN provides robust performance improvements in all of these diverse applications.
Collapse
Affiliation(s)
| | | | | | | | | | - Orest Boyko
- Department of Radiology, VA Southern Nevada Healthcare System, NV, USA
| | | |
Collapse
|
42
|
Dolezal JM, Kochanny S, Dyer E, Ramesh S, Srisuwananukorn A, Sacco M, Howard FM, Li A, Mohan P, Pearson AT. Slideflow: deep learning for digital histopathology with real-time whole-slide visualization. BMC Bioinformatics 2024; 25:134. [PMID: 38539070 PMCID: PMC10967068 DOI: 10.1186/s12859-024-05758-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 03/20/2024] [Indexed: 05/04/2024] Open
Abstract
Deep learning methods have emerged as powerful tools for analyzing histopathological images, but current methods are often specialized for specific domains and software environments, and few open-source options exist for deploying models in an interactive interface. Experimenting with different deep learning approaches typically requires switching software libraries and reprocessing data, reducing the feasibility and practicality of experimenting with new architectures. We developed a flexible deep learning library for histopathology called Slideflow, a package which supports a broad array of deep learning methods for digital pathology and includes a fast whole-slide interface for deploying trained models. Slideflow includes unique tools for whole-slide image data processing, efficient stain normalization and augmentation, weakly-supervised whole-slide classification, uncertainty quantification, feature generation, feature space analysis, and explainability. Whole-slide image processing is highly optimized, enabling whole-slide tile extraction at 40x magnification in 2.5 s per slide. The framework-agnostic data processing pipeline enables rapid experimentation with new methods built with either Tensorflow or PyTorch, and the graphical user interface supports real-time visualization of slides, predictions, heatmaps, and feature space characteristics on a variety of hardware devices, including ARM-based devices such as the Raspberry Pi.
Collapse
Affiliation(s)
- James M Dolezal
- 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
| | - Emma Dyer
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Siddhi Ramesh
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Andrew Srisuwananukorn
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Matteo Sacco
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Frederick M Howard
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Anran Li
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA
| | - Prajval Mohan
- Department of Computer Science, University of Chicago, Chicago, IL, USA
| | - Alexander T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, USA.
| |
Collapse
|
43
|
Unger M, Kather JN. Deep learning in cancer genomics and histopathology. Genome Med 2024; 16:44. [PMID: 38539231 PMCID: PMC10976780 DOI: 10.1186/s13073-024-01315-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2024] [Indexed: 07/08/2024] Open
Abstract
Histopathology and genomic profiling are cornerstones of precision oncology and are routinely obtained for patients with cancer. Traditionally, histopathology slides are manually reviewed by highly trained pathologists. Genomic data, on the other hand, is evaluated by engineered computational pipelines. In both applications, the advent of modern artificial intelligence methods, specifically machine learning (ML) and deep learning (DL), have opened up a fundamentally new way of extracting actionable insights from raw data, which could augment and potentially replace some aspects of traditional evaluation workflows. In this review, we summarize current and emerging applications of DL in histopathology and genomics, including basic diagnostic as well as advanced prognostic tasks. Based on a growing body of evidence, we suggest that DL could be the groundwork for a new kind of workflow in oncology and cancer research. However, we also point out that DL models can have biases and other flaws that users in healthcare and research need to know about, and we propose ways to address them.
Collapse
Affiliation(s)
- Michaela Unger
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
44
|
Subramanian V, Syeda-Mahmood T, Do MN. Modelling-based joint embedding of histology and genomics using canonical correlation analysis for breast cancer survival prediction. Artif Intell Med 2024; 149:102787. [PMID: 38462287 DOI: 10.1016/j.artmed.2024.102787] [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: 01/24/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 03/12/2024]
Abstract
Traditional approaches to predicting breast cancer patients' survival outcomes were based on clinical subgroups, the PAM50 genes, or the histological tissue's evaluation. With the growth of multi-modality datasets capturing diverse information (such as genomics, histology, radiology and clinical data) about the same cancer, information can be integrated using advanced tools and have improved survival prediction. These methods implicitly exploit the key observation that different modalities originate from the same cancer source and jointly provide a complete picture of the cancer. In this work, we investigate the benefits of explicitly modelling multi-modality data as originating from the same cancer under a probabilistic framework. Specifically, we consider histology and genomics as two modalities originating from the same breast cancer under a probabilistic graphical model (PGM). We construct maximum likelihood estimates of the PGM parameters based on canonical correlation analysis (CCA) and then infer the underlying properties of the cancer patient, such as survival. Equivalently, we construct CCA-based joint embeddings of the two modalities and input them to a learnable predictor. Real-world properties of sparsity and graph-structures are captured in the penalized variants of CCA (pCCA) and are better suited for cancer applications. For generating richer multi-dimensional embeddings with pCCA, we introduce two novel embedding schemes that encourage orthogonality to generate more informative embeddings. The efficacy of our proposed prediction pipeline is first demonstrated via low prediction errors of the hidden variable and the generation of informative embeddings on simulated data. When applied to breast cancer histology and RNA-sequencing expression data from The Cancer Genome Atlas (TCGA), our model can provide survival predictions with average concordance-indices of up to 68.32% along with interpretability. We also illustrate how the pCCA embeddings can be used for survival analysis through Kaplan-Meier curves.
Collapse
Affiliation(s)
- Vaishnavi Subramanian
- Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA.
| | | | - Minh N Do
- Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA
| |
Collapse
|
45
|
Meng X, Tian Y, Zhang X. [Screening of immune related gene and survival prediction of lung adenocarcinoma patients based on LightGBM model]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2024; 41:70-79. [PMID: 38403606 PMCID: PMC10894725 DOI: 10.7507/1001-5515.202305038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Lung cancer is one of the malignant tumors with the greatest threat to human health, and studies have shown that some genes play an important regulatory role in the occurrence and development of lung cancer. In this paper, a LightGBM ensemble learning method is proposed to construct a prognostic model based on immune relate gene (IRG) profile data and clinical data to predict the prognostic survival rate of lung adenocarcinoma patients. First, this method used the Limma package for differential gene expression, used CoxPH regression analysis to screen the IRG to prognosis, and then used XGBoost algorithm to score the importance of the IRG features. Finally, the LASSO regression analysis was used to select IRG that could be used to construct a prognostic model, and a total of 17 IRG features were obtained that could be used to construct model. LightGBM was trained according to the IRG screened. The K-means algorithm was used to divide the patients into three groups, and the area under curve (AUC) of receiver operating characteristic (ROC) of the model output showed that the accuracy of the model in predicting the survival rates of the three groups of patients was 96%, 98% and 96%, respectively. The experimental results show that the model proposed in this paper can divide patients with lung adenocarcinoma into three groups [5-year survival rate higher than 65% (group 1), lower than 65% but higher than 30% (group 2) and lower than 30% (group 3)] and can accurately predict the 5-year survival rate of lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Xiangfu Meng
- School of Electronics and Information Engineering, Liaoning Technical University, Huludao, Liaoning 125000, P. R. China
| | - Youfa Tian
- School of Electronics and Information Engineering, Liaoning Technical University, Huludao, Liaoning 125000, P. R. China
| | - Xiaoyan Zhang
- School of Electronics and Information Engineering, Liaoning Technical University, Huludao, Liaoning 125000, P. R. China
| |
Collapse
|
46
|
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
|
47
|
Feng X, Shu W, Li M, Li J, Xu J, He M. Pathogenomics for accurate diagnosis, treatment, prognosis of oncology: a cutting edge overview. J Transl Med 2024; 22:131. [PMID: 38310237 PMCID: PMC10837897 DOI: 10.1186/s12967-024-04915-3] [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: 10/31/2023] [Accepted: 01/20/2024] [Indexed: 02/05/2024] Open
Abstract
The capability to gather heterogeneous data, alongside the increasing power of artificial intelligence to examine it, leading a revolution in harnessing multimodal data in the life sciences. However, most approaches are limited to unimodal data, leaving integrated approaches across modalities relatively underdeveloped in computational pathology. Pathogenomics, as an invasive method to integrate advanced molecular diagnostics from genomic data, morphological information from histopathological imaging, and codified clinical data enable the discovery of new multimodal cancer biomarkers to propel the field of precision oncology in the coming decade. In this perspective, we offer our opinions on synthesizing complementary modalities of data with emerging multimodal artificial intelligence methods in pathogenomics. It includes correlation between the pathological and genomic profile of cancer, fusion of histology, and genomics profile of cancer. We also present challenges, opportunities, and avenues for future work.
Collapse
Affiliation(s)
- Xiaobing Feng
- College of Electrical and Information Engineering, Hunan University, Changsha, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Wen Shu
- College of Electrical and Information Engineering, Hunan University, Changsha, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Mingya Li
- College of Electrical and Information Engineering, Hunan University, Changsha, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Junyu Li
- College of Electrical and Information Engineering, Hunan University, Changsha, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Junyao Xu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Min He
- College of Electrical and Information Engineering, Hunan University, Changsha, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
48
|
Cai H, Liao Y, Zhu L, Wang Z, Song J. Improving Cancer Survival Prediction via Graph Convolutional Neural Network Learning on Protein-Protein Interaction Networks. IEEE J Biomed Health Inform 2024; 28:1134-1143. [PMID: 37963003 DOI: 10.1109/jbhi.2023.3332640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Cancer is one of the most challenging health problems worldwide. Accurate cancer survival prediction is vital for clinical decision making. Many deep learning methods have been proposed to understand the association between patients' genomic features and survival time. In most cases, the gene expression matrix is fed directly to the deep learning model. However, this approach completely ignores the interactions between biomolecules, and the resulting models can only learn the expression levels of genes to predict patient survival. In essence, the interaction between biomolecules is the key to determining the direction and function of biological processes. Proteins are the building blocks and principal undertakings of life activities, and as such, their complex interaction network is potentially informative for deep learning methods. Therefore, a more reliable approach is to have the neural network learn both gene expression data and protein interaction networks. We propose a new computational approach, termed CRESCENT, which is a protein-protein interaction (PPI) prior knowledge graph-based convolutional neural network (GCN) to improve cancer survival prediction. CRESCENT relies on the gene expression networks rather than gene expression levels to predict patient survival. The performance of CRESCENT is evaluated on a large-scale pan-cancer dataset consisting of 5991 patients from 16 different types of cancers. Extensive benchmarking experiments demonstrate that our proposed method is competitive in terms of the evaluation metric of the time-dependent concordance index( Ctd) when compared with several existing state-of-the-art approaches. Experiments also show that incorporating the network structure between genomic features effectively improves cancer survival prediction.
Collapse
|
49
|
Tripathi S, Tabari A, Mansur A, Dabbara H, Bridge CP, Daye D. From Machine Learning to Patient Outcomes: A Comprehensive Review of AI in Pancreatic Cancer. Diagnostics (Basel) 2024; 14:174. [PMID: 38248051 PMCID: PMC10814554 DOI: 10.3390/diagnostics14020174] [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: 09/19/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Pancreatic cancer is a highly aggressive and difficult-to-detect cancer with a poor prognosis. Late diagnosis is common due to a lack of early symptoms, specific markers, and the challenging location of the pancreas. Imaging technologies have improved diagnosis, but there is still room for improvement in standardizing guidelines. Biopsies and histopathological analysis are challenging due to tumor heterogeneity. Artificial Intelligence (AI) revolutionizes healthcare by improving diagnosis, treatment, and patient care. AI algorithms can analyze medical images with precision, aiding in early disease detection. AI also plays a role in personalized medicine by analyzing patient data to tailor treatment plans. It streamlines administrative tasks, such as medical coding and documentation, and provides patient assistance through AI chatbots. However, challenges include data privacy, security, and ethical considerations. This review article focuses on the potential of AI in transforming pancreatic cancer care, offering improved diagnostics, personalized treatments, and operational efficiency, leading to better patient outcomes.
Collapse
Affiliation(s)
- Satvik Tripathi
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Azadeh Tabari
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Arian Mansur
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Harvard Medical School, Boston, MA 02115, USA
| | - Harika Dabbara
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Christopher P. Bridge
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Dania Daye
- Department of Radiology, Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (A.T.); (A.M.); (C.P.B.)
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
50
|
Jiang L, Xu C, Bai Y, Liu A, Gong Y, Wang YP, Deng HW. Autosurv: interpretable deep learning framework for cancer survival analysis incorporating clinical and multi-omics data. NPJ Precis Oncol 2024; 8:4. [PMID: 38182734 PMCID: PMC10770412 DOI: 10.1038/s41698-023-00494-6] [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: 01/26/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Accurate prognosis for cancer patients can provide critical information for optimizing treatment plans and improving life quality. Combining omics data and demographic/clinical information can offer a more comprehensive view of cancer prognosis than using omics or clinical data alone and can also reveal the underlying disease mechanisms at the molecular level. In this study, we developed and validated a deep learning framework to extract information from high-dimensional gene expression and miRNA expression data and conduct prognosis prediction for breast cancer and ovarian-cancer patients using multiple independent multi-omics datasets. Our model achieved significantly better prognosis prediction than the current machine learning and deep learning approaches in various settings. Moreover, an interpretation method was applied to tackle the "black-box" nature of deep neural networks and we identified features (i.e., genes, miRNA, demographic/clinical variables) that were important to distinguish predicted high- and low-risk patients. The significance of the identified features was partially supported by previous studies.
Collapse
Affiliation(s)
- Lindong Jiang
- Tulane Center of Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yuntong Bai
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Anqi Liu
- Tulane Center of Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Yun Gong
- Tulane Center of Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Yu-Ping Wang
- Department of Biomedical Engineering, School of Science and Engineering, Tulane University, New Orleans, LA, 70118, USA
| | - Hong-Wen Deng
- Tulane Center of Biomedical Informatics and Genomics, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|