101
|
Klauschen F, Dippel J, Keyl P, Jurmeister P, Bockmayr M, Mock A, Buchstab O, Alber M, Ruff L, Montavon G, Müller KR. Toward Explainable Artificial Intelligence for Precision Pathology. ANNUAL REVIEW OF PATHOLOGY 2024; 19:541-570. [PMID: 37871132 DOI: 10.1146/annurev-pathmechdis-051222-113147] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The rapid development of precision medicine in recent years has started to challenge diagnostic pathology with respect to its ability to analyze histological images and increasingly large molecular profiling data in a quantitative, integrative, and standardized way. Artificial intelligence (AI) and, more precisely, deep learning technologies have recently demonstrated the potential to facilitate complex data analysis tasks, including clinical, histological, and molecular data for disease classification; tissue biomarker quantification; and clinical outcome prediction. This review provides a general introduction to AI and describes recent developments with a focus on applications in diagnostic pathology and beyond. We explain limitations including the black-box character of conventional AI and describe solutions to make machine learning decisions more transparent with so-called explainable AI. The purpose of the review is to foster a mutual understanding of both the biomedical and the AI side. To that end, in addition to providing an overview of the relevant foundations in pathology and machine learning, we present worked-through examples for a better practical understanding of what AI can achieve and how it should be done.
Collapse
Affiliation(s)
- Frederick Klauschen
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- German Cancer Consortium, German Cancer Research Center (DKTK/DKFZ), Munich Partner Site, Munich, Germany
| | - Jonas Dippel
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- Machine Learning Group, Department of Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany;
| | - Philipp Keyl
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
| | - Philipp Jurmeister
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
- German Cancer Consortium, German Cancer Research Center (DKTK/DKFZ), Munich Partner Site, Munich, Germany
| | - Michael Bockmayr
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Andreas Mock
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
- German Cancer Consortium, German Cancer Research Center (DKTK/DKFZ), Munich Partner Site, Munich, Germany
| | - Oliver Buchstab
- Institute of Pathology, Ludwig-Maximilians-Universität München, Munich, Germany;
| | - Maximilian Alber
- Institute of Pathology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Aignostics, Berlin, Germany
| | | | - Grégoire Montavon
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- Machine Learning Group, Department of Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany;
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
| | - Klaus-Robert Müller
- Berlin Institute for the Foundations of Learning and Data (BIFOLD), Berlin, Germany
- Machine Learning Group, Department of Electrical Engineering and Computer Science, Technische Universität Berlin, Berlin, Germany;
- Department of Artificial Intelligence, Korea University, Seoul, Korea
- Max Planck Institute for Informatics, Saarbrücken, Germany
| |
Collapse
|
102
|
Yin G, Liu L, Yu T, Yu L, Feng M, Zhou C, Wang X, Teng G, Ma Z, Zhou W, Ye C, Zhang J, Ji C, Zhao L, Zhou P, Guo Y, Meng X, Fu Q, Zhang Q, Li L, Zhou F, Zheng C, Xiang Y, Guo M, Wang Y, Wang F, Huang S, Yu Z. Genomic and transcriptomic analysis of breast cancer identifies novel signatures associated with response to neoadjuvant chemotherapy. Genome Med 2024; 16:11. [PMID: 38217005 PMCID: PMC10787499 DOI: 10.1186/s13073-024-01286-8] [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: 07/24/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NAC) has become a standard treatment strategy for breast cancer (BC). However, owing to the high heterogeneity of these tumors, it is unclear which patient population most likely benefit from NAC. Multi-omics offer an improved approach to uncovering genomic and transcriptomic changes before and after NAC in BC and to identifying molecular features associated with NAC sensitivity. METHODS We performed whole-exome and RNA sequencing on 233 samples (including matched pre- and post-treatment tumors) from 50 BC patients with rigorously defined responses to NAC and analyzed changes in the multi-omics landscape. Molecular features associated with NAC response were identified and validated in a larger internal, and two external validation cohorts, as well as in vitro experiments. RESULTS The most frequently altered genes were TP53, TTN, and MUC16 in both pre- and post-treatment tumors. In comparison with pre-treatment tumors, there was a significant decrease in C > A transversion mutations in post-treatment tumors (P = 0.020). NAC significantly decreased the mutation rate (P = 0.006) of the DNA repair pathway and gene expression levels (FDR = 0.007) in this pathway. NAC also significantly changed the expression level of immune checkpoint genes and the abundance of tumor-infiltrating immune and stroma cells, including B cells, activated dendritic cells, γδT cells, M2 macrophages and endothelial cells. Furthermore, there was a higher rate of C > T substitutions in NAC nonresponsive tumors than responsive ones, especially when the substitution site was flanked by C and G. Importantly, there was a unique amplified region at 8p11.23 (containing ADGRA2 and ADRB3) and a deleted region at 3p13 (harboring FOXP1) in NAC nonresponsive and responsive tumors, respectively. Particularly, the CDKAL1 missense variant P409L (p.Pro409Leu, c.1226C > T) decreased BC cell sensitivity to docetaxel, and ADGRA2 or ADRB3 gene amplifications were associated with worse NAC response and poor prognosis in BC patients. CONCLUSIONS Our study has revealed genomic and transcriptomic landscape changes following NAC in BC, and identified novel biomarkers (CDKAL1P409L, ADGRA2 and ADRB3) underlying chemotherapy resistance and poor prognosis, which could guide the development of personalized treatments for BC.
Collapse
Affiliation(s)
- Gengshen Yin
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Liyuan Liu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Ting Yu
- Research Center for Mathematics and Interdisciplinary Sciences, Shandong University, Qingdao, 266237, China
| | - Lixiang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Man Feng
- Department of Pathology, The Third Affiliated Hospital of Shandong First Medical University (Affiliated Hospital of Shandong Academy of Medical Sciences), Jinan, 250031, China
| | - Chengjun Zhou
- Department of Pathology, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Xiaoying Wang
- Department of Pathology, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Guoxin Teng
- Department of Pathology, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Zhongbing Ma
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Wenzhong Zhou
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Chunmiao Ye
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Jialin Zhang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Changhua Ji
- Department of Pathology, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Linfeng Zhao
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Peng Zhou
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Yaxun Guo
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Xingchen Meng
- Department of Breast Surgery, Weifang People's Hospital, Weifang, 261041, China
| | - Qinye Fu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Qiang Zhang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Liang Li
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Fei Zhou
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Chao Zheng
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Yujuan Xiang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Mingming Guo
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Yongjiu Wang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China
| | - Fei Wang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China.
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China.
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China.
| | - Shuya Huang
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China.
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China.
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China.
| | - Zhigang Yu
- Department of Breast Surgery, The Second Hospital of Shandong University, Jinan, 250033, China.
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, 250033, China.
- Shandong Provincial Engineering Laboratory of Translational Research On Prevention and Treatment of Breast Disease, Jinan, 250033, China.
| |
Collapse
|
103
|
Tong L, Shi W, Isgut M, Zhong Y, Lais P, Gloster L, Sun J, Swain A, Giuste F, Wang MD. Integrating Multi-Omics Data With EHR for Precision Medicine Using Advanced Artificial Intelligence. IEEE Rev Biomed Eng 2024; 17:80-97. [PMID: 37824325 DOI: 10.1109/rbme.2023.3324264] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
With the recent advancement of novel biomedical technologies such as high-throughput sequencing and wearable devices, multi-modal biomedical data ranging from multi-omics molecular data to real-time continuous bio-signals are generated at an unprecedented speed and scale every day. For the first time, these multi-modal biomedical data are able to make precision medicine close to a reality. However, due to data volume and the complexity, making good use of these multi-modal biomedical data requires major effort. Researchers and clinicians are actively developing artificial intelligence (AI) approaches for data-driven knowledge discovery and causal inference using a variety of biomedical data modalities. These AI-based approaches have demonstrated promising results in various biomedical and healthcare applications. In this review paper, we summarize the state-of-the-art AI models for integrating multi-omics data and electronic health records (EHRs) for precision medicine. We discuss the challenges and opportunities in integrating multi-omics data with EHRs and future directions. We hope this review can inspire future research and developing in integrating multi-omics data with EHRs for precision medicine.
Collapse
|
104
|
Dziubańska-Kusibab PJ, Nevedomskaya E, Haendler B. Preclinical Anticipation of On- and Off-Target Resistance Mechanisms to Anti-Cancer Drugs: A Systematic Review. Int J Mol Sci 2024; 25:705. [PMID: 38255778 PMCID: PMC10815614 DOI: 10.3390/ijms25020705] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/22/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
The advent of targeted therapies has led to tremendous improvements in treatment options and their outcomes in the field of oncology. Yet, many cancers outsmart precision drugs by developing on-target or off-target resistance mechanisms. Gaining the ability to resist treatment is the rule rather than the exception in tumors, and it remains a major healthcare challenge to achieve long-lasting remission in most cancer patients. Here, we discuss emerging strategies that take advantage of innovative high-throughput screening technologies to anticipate on- and off-target resistance mechanisms before they occur in treated cancer patients. We divide the methods into non-systematic approaches, such as random mutagenesis or long-term drug treatment, and systematic approaches, relying on the clustered regularly interspaced short palindromic repeats (CRISPR) system, saturated mutagenesis, or computational methods. All these new developments, especially genome-wide CRISPR-based screening platforms, have significantly accelerated the processes for identification of the mechanisms responsible for cancer drug resistance and opened up new avenues for future treatments.
Collapse
Affiliation(s)
| | | | - Bernard Haendler
- Research and Early Development Oncology, Pharmaceuticals, Bayer AG, Müllerstr. 178, 13353 Berlin, Germany; (P.J.D.-K.); (E.N.)
| |
Collapse
|
105
|
Donati B, Reggiani F, Torricelli F, Santandrea G, Rossi T, Bisagni A, Gasparini E, Neri A, Cortesi L, Ferrari G, Bisagni G, Ragazzi M, Ciarrocchi A. Spatial Distribution of Immune Cells Drives Resistance to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Cancer Immunol Res 2024; 12:120-134. [PMID: 37856875 DOI: 10.1158/2326-6066.cir-23-0076] [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: 01/26/2023] [Revised: 06/22/2023] [Accepted: 10/19/2023] [Indexed: 10/21/2023]
Abstract
Neoadjuvant chemotherapy (NAC) alone or combined with target therapies represents the standard of care for localized triple-negative breast cancer (TNBC). However, only a fraction of patients have a response, necessitating better understanding of the complex elements in the TNBC ecosystem that establish continuous and multidimensional interactions. Resolving such complexity requires new spatially-defined approaches. Here, we used spatial transcriptomics to investigate the multidimensional organization of TNBC at diagnosis and explore the contribution of each cell component to response to NAC. Starting from a consecutive retrospective series of TNBC cases, we designed a case-control study including 24 patients with TNBC of which 12 experienced a pathologic complete response (pCR) and 12 no-response or progression (pNR) after NAC. Over 200 regions of interest (ROI) were profiled. Our computational approaches described a model that recapitulates clinical response to therapy. The data were validated in an independent cohort of patients. Differences in the transcriptional program were detected in the tumor, stroma, and immune infiltrate comparing patients with a pCR with those with pNR. In pCR, spatial contamination between the tumor mass and the infiltrating lymphocytes was observed, sustained by a massive activation of IFN-signaling. Conversely, pNR lesions displayed increased pro-angiogenetic signaling and oxygen-based metabolism. Only modest differences were observed in the stroma, revealing a topology-based functional heterogeneity of the immune infiltrate. Thus, spatial transcriptomics provides fundamental information on the multidimensionality of TNBC and allows an effective prediction of tumor behavior. These results open new perspectives for the improvement and personalization of therapeutic approaches to TNBCs.
Collapse
Affiliation(s)
- Benedetta Donati
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Francesca Reggiani
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Federica Torricelli
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giacomo Santandrea
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Teresa Rossi
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandra Bisagni
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisa Gasparini
- Oncology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonino Neri
- Scientific Directorate, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Laura Cortesi
- Department of Oncology and Hematology, Azienda Ospedaliera Policlinico di Modena, Modena, Italy
| | - Guglielmo Ferrari
- Breast Surgery Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giancarlo Bisagni
- Oncology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Moira Ragazzi
- Pathology Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
106
|
Huang Y, Yang H, Li J, Wang F, Liu W, Liu Y, Wang R, Duan L, Wu J, Gao Z, Cao J, Bian F, Zhang J, Zhao F, Yang S, Cao S, Yang A, Wang X, Geng M, Hao A, Li J, Cao J, Li C, Zhang Z, Zhang N, Huang Y, Zhang Y, Qian K, Zhou F. Diagnosis of Esophageal Squamous Cell Carcinoma by High-Performance Serum Metabolic Fingerprints: A Retrospective Study. SMALL METHODS 2024; 8:e2301046. [PMID: 37803160 DOI: 10.1002/smtd.202301046] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/22/2023] [Indexed: 10/08/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly prevalent and aggressive malignancy, and timely diagnosis of ESCC contributes to an increased cancer survival rate. However, current detection methods for ESCC mainly rely on endoscopic examination, limited by a relatively low participation rate. Herein, ferric-particle-enhanced laser desorption/ionization mass spectrometry (FPELDI MS) is utilized to record the serum metabolic fingerprints (SMFs) from a retrospective cohort (523 non-ESCC participants and 462 ESCC patients) to build diagnostic models toward ESCC. The PFELDI MS achieved high speed (≈30 s per sample), desirable reproducibility (coefficients of variation < 15%), and high throughput (985 samples with ≈124 200 data points for each spectrum). Desirable diagnostic performance with area-under-the-curves (AUCs) of 0.925-0.966 is obtained through machine learning of SMFs. Further, a metabolic biomarker panel is constructed, exhibiting superior diagnostic sensitivity (72.2-79.4%, p < 0.05) as compared with clinical protein biomarker tests (4.3-22.9%). Notably, the biomarker panel afforded an AUC of 0.844 (95% confidence interval [CI]: 0.806-0.880) toward early ESCC diagnosis. This work highlighted the potential of metabolic analysis for accurate screening and early detection of ESCC and offered insights into the metabolic characterization of diseases including but not limited to ESCC.
Collapse
Affiliation(s)
- Yida Huang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Haijun Yang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Junkuo Li
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Fuqiang Wang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Wanshan Liu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Yiwen Liu
- The First Affiliated Hospital, Henan Key Laboratory of Cancer Epigenetics, Henan University of Science and Technology, Luoyang, 471003, P. R. China
| | - Ruimin Wang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Lijuan Duan
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Jiao Wu
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Zhaowei Gao
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Jing Cao
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Fang Bian
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Juxiang Zhang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Fang Zhao
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Shouzhi Yang
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Shasha Cao
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Aihua Yang
- Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, 200433, P. R. China
| | - Xueliang Wang
- Shanghai Center for Clinical Laboratory, Shanghai Academy of Experimental Medicine, Shanghai, 200126, P. R. China
| | - Mingfei Geng
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Anlin Hao
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Jian Li
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Jianwei Cao
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Chaowei Li
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Zheyuan Zhang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Ning Zhang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Yanlin Huang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Yaowen Zhang
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
- Department of Obstetrics and Gynecology, Shanghai Key Laboratory of Gynecologic Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
- Division of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Fuyou Zhou
- Anyang Tumor Hospital, Anyang Tumor Hospital affiliated to Henan University of Science and Technology, Henan Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455001, P. R. China
| |
Collapse
|
107
|
Li Y, Wu F, Ge W, Zhang Y, Hu Y, Zhao L, Gou W, Shi J, Ni Y, Li L, Fu W, Lin X, Yu Y, Han Z, Chen C, Xu R, Zhang S, Zhou L, Pan G, Peng Y, Mao L, Zhou T, Zheng J, Zheng H, Sun Y, Guo T, Luo D. Risk stratification of papillary thyroid cancers using multidimensional machine learning. Int J Surg 2024; 110:372-384. [PMID: 37916932 PMCID: PMC10793787 DOI: 10.1097/js9.0000000000000814] [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: 07/10/2023] [Accepted: 09/18/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND Papillary thyroid cancer (PTC) is one of the most common endocrine malignancies with different risk levels. However, preoperative risk assessment of PTC is still a challenge in the worldwide. Here, the authors first report a Preoperative Risk Assessment Classifier for PTC (PRAC-PTC) by multidimensional features including clinical indicators, immune indices, genetic feature, and proteomics. MATERIALS AND METHODS The 558 patients collected from June 2013 to November 2020 were allocated to three groups: the discovery set [274 patients, 274 formalin-fixed paraffin-embedded (FFPE)], the retrospective test set (166 patients, 166 FFPE), and the prospective test set (118 patients, 118 fine-needle aspiration). Proteomic profiling was conducted by FFPE and fine-needle aspiration tissues from the patients. Preoperative clinical information and blood immunological indices were collected. The BRAFV600E mutation were detected by the amplification refractory mutation system. RESULTS The authors developed a machine learning model of 17 variables based on the multidimensional features of 274 PTC patients from a retrospective cohort. The PRAC-PTC achieved areas under the curve (AUC) of 0.925 in the discovery set and was validated externally by blinded analyses in a retrospective cohort of 166 PTC patients (0.787 AUC) and a prospective cohort of 118 PTC patients (0.799 AUC) from two independent clinical centres. Meanwhile, the preoperative predictive risk effectiveness of clinicians was improved with the assistance of PRAC-PTC, and the accuracies reached at 84.4% (95% CI: 82.9-84.4) and 83.5% (95% CI: 82.2-84.2) in the retrospective and prospective test sets, respectively. CONCLUSION This study demonstrated that the PRAC-PTC that integrating clinical data, gene mutation information, immune indices, high-throughput proteomics and machine learning technology in multicentre retrospective and prospective clinical cohorts can effectively stratify the preoperative risk of PTC and may decrease unnecessary surgery or overtreatment.
Collapse
Affiliation(s)
| | - Fan Wu
- Department of Oncological Surgery
| | - Weigang Ge
- bWestlake Omics (Hangzhou) Biotechnology Co., Ltd
| | - Yu Zhang
- Department of Oncological Surgery
| | - Yifan Hu
- bWestlake Omics (Hangzhou) Biotechnology Co., Ltd
| | - Lingqian Zhao
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University
| | - Wanglong Gou
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang
| | | | - Yeqin Ni
- Department of Oncological Surgery
| | - Lu Li
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University
- Research Centre for Industries of the Future, Westlake University
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province
| | - Wenxin Fu
- bWestlake Omics (Hangzhou) Biotechnology Co., Ltd
| | - Xiangfeng Lin
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong Province, People’s Republic of China
| | - Yunxian Yu
- Department of Epidemiology and Health Statistics, School of Public Health, Zhejiang University
| | | | | | | | - Shirong Zhang
- Centre of Translational Medicine, Hangzhou First People’s Hospital
| | - Li Zhou
- Department of Oncological Surgery
| | - Gang Pan
- Department of Oncological Surgery
| | - You Peng
- Department of Oncological Surgery
| | | | - Tianhan Zhou
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University
| | - Jusheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang
| | - Haitao Zheng
- Department of Thyroid Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Shandong Province, People’s Republic of China
| | - Yaoting Sun
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University
- Research Centre for Industries of the Future, Westlake University
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province
| | - Tiannan Guo
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University
- Research Centre for Industries of the Future, Westlake University
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province
| | - Dingcun Luo
- Department of Oncological Surgery
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University
| |
Collapse
|
108
|
Yang H, Xu Y, Dong M, Zhang Y, Gong J, Huang D, He J, Wei L, Huang S, Zhao L. Automated Prediction of Neoadjuvant Chemoradiotherapy Response in Locally Advanced Cervical Cancer Using Hybrid Model-Based MRI Radiomics. Diagnostics (Basel) 2023; 14:5. [PMID: 38201314 PMCID: PMC10795804 DOI: 10.3390/diagnostics14010005] [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: 09/18/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND This study aimed to develop a model that automatically predicts the neoadjuvant chemoradiotherapy (nCRT) response for patients with locally advanced cervical cancer (LACC) based on T2-weighted MR images and clinical parameters. METHODS A total of 138 patients were enrolled, and T2-weighted MR images and clinical information of the patients before treatment were collected. Clinical information included age, stage, pathological type, squamous cell carcinoma (SCC) level, and lymph node status. A hybrid model extracted the domain-specific features from the computational radiomics system, the abstract features from the deep learning network, and the clinical parameters. Then, it employed an ensemble learning classifier weighted by logistic regression (LR) classifier, support vector machine (SVM) classifier, K-Nearest Neighbor (KNN) classifier, and Bayesian classifier to predict the pathologic complete response (pCR). The area under the receiver operating characteristics curve (AUC), accuracy (ACC), true positive rate (TPR), true negative rate (TNR), and precision were used as evaluation metrics. RESULTS Among the 138 LACC patients, 74 were in the pCR group, and 64 were in the non-pCR group. There was no significant difference between the two cohorts in terms of tumor diameter (p = 0.787), lymph node (p = 0.068), and stage before radiotherapy (p = 0.846), respectively. The 109-dimension domain features and 1472-dimension abstract features from MRI images were used to form a hybrid model. The average AUC, ACC, TPR, TNR, and precision of the proposed hybrid model were about 0.80, 0.71, 0.75, 0.66, and 0.71, while the AUC values of using clinical parameters, domain-specific features, and abstract features alone were 0.61, 0.67 and 0.76, respectively. The AUC value of the model without an ensemble learning classifier was 0.76. CONCLUSIONS The proposed hybrid model can predict the radiotherapy response of patients with LACC, which might help radiation oncologists create personalized treatment plans for patients.
Collapse
Affiliation(s)
- Hua Yang
- Department of Radiation Oncology, Xijing Hospital of Air Force Medical University, Xi’an 710032, China; (H.Y.); (Y.Z.); (J.G.)
- Department of Radiation Oncology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Yinan Xu
- Key Lab of Intelligent Perception and Image Understanding of Ministry of Education, Xidian University, Xi’an 710071, China;
| | - Mohan Dong
- Department of Medical Education, Xijing Hospital of Air Force Medical University, Xi’an 710032, China;
| | - Ying Zhang
- Department of Radiation Oncology, Xijing Hospital of Air Force Medical University, Xi’an 710032, China; (H.Y.); (Y.Z.); (J.G.)
| | - Jie Gong
- Department of Radiation Oncology, Xijing Hospital of Air Force Medical University, Xi’an 710032, China; (H.Y.); (Y.Z.); (J.G.)
| | - Dong Huang
- Department of Military Biomedical Engineering, Air Force Medical University, Xi’an 710012, China;
| | - Junhua He
- Department of Radiation Oncology, 986 Hospital of Air Force Medical University, Xi’an 710054, China;
| | - Lichun Wei
- Department of Radiation Oncology, Xijing Hospital of Air Force Medical University, Xi’an 710032, China; (H.Y.); (Y.Z.); (J.G.)
| | - Shigao Huang
- Department of Radiation Oncology, Xijing Hospital of Air Force Medical University, Xi’an 710032, China; (H.Y.); (Y.Z.); (J.G.)
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital of Air Force Medical University, Xi’an 710032, China; (H.Y.); (Y.Z.); (J.G.)
| |
Collapse
|
109
|
Alvarez-Frutos L, Barriuso D, Duran M, Infante M, Kroemer G, Palacios-Ramirez R, Senovilla L. Multiomics insights on the onset, progression, and metastatic evolution of breast cancer. Front Oncol 2023; 13:1292046. [PMID: 38169859 PMCID: PMC10758476 DOI: 10.3389/fonc.2023.1292046] [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: 09/10/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer is the most common malignant neoplasm in women. Despite progress to date, 700,000 women worldwide died of this disease in 2020. Apparently, the prognostic markers currently used in the clinic are not sufficient to determine the most appropriate treatment. For this reason, great efforts have been made in recent years to identify new molecular biomarkers that will allow more precise and personalized therapeutic decisions in both primary and recurrent breast cancers. These molecular biomarkers include genetic and post-transcriptional alterations, changes in protein expression, as well as metabolic, immunological or microbial changes identified by multiple omics technologies (e.g., genomics, epigenomics, transcriptomics, proteomics, glycomics, metabolomics, lipidomics, immunomics and microbiomics). This review summarizes studies based on omics analysis that have identified new biomarkers for diagnosis, patient stratification, differentiation between stages of tumor development (initiation, progression, and metastasis/recurrence), and their relevance for treatment selection. Furthermore, this review highlights the importance of clinical trials based on multiomics studies and the need to advance in this direction in order to establish personalized therapies and prolong disease-free survival of these patients in the future.
Collapse
Affiliation(s)
- Lucia Alvarez-Frutos
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Daniel Barriuso
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mercedes Duran
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Mar Infante
- Laboratory of Molecular Genetics of Hereditary Cancer, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, Paris, France
| | - Roberto Palacios-Ramirez
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
| | - Laura Senovilla
- Laboratory of Cell Stress and Immunosurveillance, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid – Centro Superior de Investigaciones Cientificas (CSIC), Valladolid, Spain
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
110
|
Song W, Wu F, Yan Y, Li Y, Wang Q, Hu X, Li Y. Gut microbiota landscape and potential biomarker identification in female patients with systemic lupus erythematosus using machine learning. Front Cell Infect Microbiol 2023; 13:1289124. [PMID: 38169617 PMCID: PMC10758415 DOI: 10.3389/fcimb.2023.1289124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Objectives Systemic Lupus Erythematosus (SLE) is a complex autoimmune disease that disproportionately affects women. Early diagnosis and prevention are crucial for women's health, and the gut microbiota has been found to be strongly associated with SLE. This study aimed to identify potential biomarkers for SLE by characterizing the gut microbiota landscape using feature selection and exploring the use of machine learning (ML) algorithms with significantly dysregulated microbiotas (SDMs) for early identification of SLE patients. Additionally, we used the SHapley Additive exPlanations (SHAP) interpretability framework to visualize the impact of SDMs on the risk of developing SLE in females. Methods Stool samples were collected from 54 SLE patients and 55 Negative Controls (NC) for microbiota analysis using 16S rRNA sequencing. Feature selection was performed using Elastic Net and Boruta on species-level taxonomy. Subsequently, four ML algorithms, namely logistic regression (LR), Adaptive Boosting (AdaBoost), Random Forest (RF), and eXtreme gradient boosting (XGBoost), were used to achieve early identification of SLE with SDMs. Finally, the best-performing algorithm was combined with SHAP to explore how SDMs affect the risk of developing SLE in females. Results Both alpha and beta diversity were found to be different in SLE group. Following feature selection, 68 and 21 microbiota were retained in Elastic Net and Boruta, respectively, with 16 microbiota overlapping between the two, i.e., SDMs for SLE. The four ML algorithms with SDMs could effectively identify SLE patients, with XGBoost performing the best, achieving Accuracy, Sensitivity, Specificity, Positive Predictive Value, Negative Predictive Value, and AUC values of 0.844, 0.750, 0.938, 0.923, 0.790, and 0.930, respectively. The SHAP interpretability framework showed a complex non-linear relationship between the relative abundance of SDMs and the risk of SLE, with Escherichia_fergusonii having the largest SHAP value. Conclusions This study revealed dysbiosis in the gut microbiota of female SLE patients. ML classifiers combined with SDMs can facilitate early identification of female patients with SLE, particularly XGBoost. The SHAP interpretability framework provides insight into the impact of SDMs on the risk of SLE and may inform future scientific treatment for SLE.
Collapse
Affiliation(s)
- Wenzhu Song
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Feng Wu
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yan Yan
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
| | - Yaheng Li
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, Shanxiuan, China
| | - Qian Wang
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, Shanxiuan, China
| | - Xueli Hu
- Department of Nephrology, Hejin People’s Hospital, Yuncheng, Shanxi, China
| | - Yafeng Li
- Department of Nephrology, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Kidney Disease, Taiyuan, Shanxiuan, China
- Core Laboratory, Shanxi Provincial People's Hospital (Fifth Hospital) of Shanxi Medical University, Taiyuan, China
- Academy of Microbial Ecology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
111
|
Lee S, Sun M, Hu Y, Wang Y, Islam MN, Goerlitz D, Lucas PC, Lee AV, Swain SM, Tang G, Wang XS. iGenSig-Rx: an integral genomic signature based white-box tool for modeling cancer therapeutic responses using multi-omics data. RESEARCH SQUARE 2023:rs.3.rs-3649238. [PMID: 38077030 PMCID: PMC10705599 DOI: 10.21203/rs.3.rs-3649238/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Multi-omics sequencing is expected to become clinically routine within the next decade and transform clinical care. However, there is a paucity of viable and interpretable genome-wide modeling methods that can facilitate rational selection of patients for tailored intervention. Here we develop an integral genomic signature-based method called iGenSig-Rx as a white-box tool for modeling therapeutic response based on clinical trial datasets with improved cross-dataset applicability and tolerance to sequencing bias. This method leverages high-dimensional redundant genomic features to address the challenges of cross-dataset modeling, a concept similar to the use of redundant steel rods to reinforce the pillars of a building. Using genomic datasets for HER2 targeted therapies, the iGenSig-Rx model demonstrates stable predictive power across four independent clinical trials. More importantly, the iGenSig-Rx model offers the level of transparency much needed for clinical application, allowing for clear explanations as to how the predictions are produced, how the features contribute to the prediction, and what are the key underlying pathways. We expect that iGenSig-Rx as a class of biologically interpretable multi-omics modeling methods will have broad applications in big-data based precision oncology. The R package is available: https://github.com/wangxlab/iGenSig-Rx. NOTE: the Github website will be released upon publication and the R package is available for review through google drive: https://drive.google.com/drive/folders/1KgecmUoon9-h2Dg1rPCyEGFPOp28Ols3?usp=sharing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Sandra M Swain
- National Surgical Adjuvant Breast and Bowel Project (NSABP)
| | | | | |
Collapse
|
112
|
Zhang C, Xu J, Tang R, Yang J, Wang W, Yu X, Shi S. Novel research and future prospects of artificial intelligence in cancer diagnosis and treatment. J Hematol Oncol 2023; 16:114. [PMID: 38012673 PMCID: PMC10680201 DOI: 10.1186/s13045-023-01514-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Research into the potential benefits of artificial intelligence for comprehending the intricate biology of cancer has grown as a result of the widespread use of deep learning and machine learning in the healthcare sector and the availability of highly specialized cancer datasets. Here, we review new artificial intelligence approaches and how they are being used in oncology. We describe how artificial intelligence might be used in the detection, prognosis, and administration of cancer treatments and introduce the use of the latest large language models such as ChatGPT in oncology clinics. We highlight artificial intelligence applications for omics data types, and we offer perspectives on how the various data types might be combined to create decision-support tools. We also evaluate the present constraints and challenges to applying artificial intelligence in precision oncology. Finally, we discuss how current challenges may be surmounted to make artificial intelligence useful in clinical settings in the future.
Collapse
Affiliation(s)
- Chaoyi Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jianhui Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, No. 399 Lingling Road, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
113
|
Zelli V, Manno A, Compagnoni C, Ibraheem RO, Zazzeroni F, Alesse E, Rossi F, Arbib C, Tessitore A. Classification of tumor types using XGBoost machine learning model: a vector space transformation of genomic alterations. J Transl Med 2023; 21:836. [PMID: 37990214 PMCID: PMC10664515 DOI: 10.1186/s12967-023-04720-4] [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: 05/16/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Machine learning (ML) represents a powerful tool to capture relationships between molecular alterations and cancer types and to extract biological information. Here, we developed a plain ML model aimed at distinguishing cancer types based on genetic lesions, providing an additional tool to improve cancer diagnosis, particularly for tumors of unknown origin. METHODS TCGA data from 9,927 samples spanning 32 different cancer types were downloaded from cBioportal. A vector space model type data transformation technique was designed to build consistently homogeneous new datasets containing, as predictive features, calls for somatic point mutations and copy number variations at chromosome arm-level, thus allowing the use of the XGBoost classifier models. Considering the imbalance in the dataset, due to large difference in the number of cases for each tumor, two preprocessing strategies were considered: i) setting a percentage cut-off threshold to remove less represented cancer types, ii) dividing cancer types into different groups based on biological criteria and training a specific XGBoost model for each of them. The performance of all trained models was mainly assessed by the out-of-sample balanced accuracy (BACC) and the AUC scores. RESULTS The XGBoost classifier achieved the best performance (BACC 77%; AUC 97%) on a dataset containing the 10 most represented tumor types. Moreover, dividing the 18 most represented cancers into three different groups (endocrine-related carcinomas, other carcinomas and other cancers),such analysis models achieved 78%, 71% and 86% BACC, respectively, with AUC scores greater than 96%. In addition, the model capable of linking each group to a specific cancer type reached 81% BACC and 94% AUC. Overall, the diagnostic potential of our model was comparable/higher with respect to others already described in literature and based on similar molecular data and ML approaches. CONCLUSIONS A boosted ML approach able to accurately discriminate different cancer types was developed. The methodology builds datasets simpler and more interpretable than the original data, while keeping enough information to accurately train standard ML models without resorting to sophisticated Deep Learning architectures. In combination with histopathological examinations, this approach could improve cancer diagnosis by using specific DNA alterations, processed by a replicable and easy-to-use automated technology. The study encourages new investigations which could further increase the classifier's performance, for example by considering more features and dividing tumors into their main molecular subtypes.
Collapse
Affiliation(s)
- Veronica Zelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
- Center for Molecular Diagnostics and Advanced Therapies, University of L'Aquila, Via Petrini, 67100, L'Aquila, Italy
| | - Andrea Manno
- Department of Information Engineering, Computer Science and Mathematics, Center of Excellence DEWS, University of L'Aquila, 67100, L'Aquila, Italy
| | - Chiara Compagnoni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Rasheed Oyewole Ibraheem
- Department of Information Engineering, Computer Science and Mathematics, Center of Excellence DEWS, University of L'Aquila, 67100, L'Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Fabrizio Rossi
- Department of Information Engineering, Computer Science and Mathematics, Center of Excellence DEWS, University of L'Aquila, 67100, L'Aquila, Italy
| | - Claudio Arbib
- Department of Information Engineering, Computer Science and Mathematics, Center of Excellence DEWS, University of L'Aquila, 67100, L'Aquila, Italy
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100, L'Aquila, Italy.
- Center for Molecular Diagnostics and Advanced Therapies, University of L'Aquila, Via Petrini, 67100, L'Aquila, Italy.
| |
Collapse
|
114
|
Tao Y, Xing S, Zuo S, Bao P, Jin Y, Li Y, Li M, Wu Y, Chen S, Wang X, Zhu Y, Feng Y, Zhang X, Wang X, Xi Q, Lu Q, Wang P, Lu ZJ. Cell-free multi-omics analysis reveals potential biomarkers in gastrointestinal cancer patients' blood. Cell Rep Med 2023; 4:101281. [PMID: 37992683 PMCID: PMC10694666 DOI: 10.1016/j.xcrm.2023.101281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/29/2023] [Accepted: 10/16/2023] [Indexed: 11/24/2023]
Abstract
During cancer progression, tumorigenic and immune signals are spread through circulating molecules, such as cell-free DNA (cfDNA) and cell-free RNA (cfRNA) in the blood. So far, they have not been comprehensively investigated in gastrointestinal cancers. Here, we profile 4 categories of cell-free omics data from patients with colorectal cancer and patients with stomach adenocarcinoma and then assay 15 types of genomic, epigenomic, and transcriptomic variations. We find that multi-omics data are more appropriate for detection of cancer genes compared with single-omics data. In particular, cfRNAs are more sensitive and informative than cfDNAs in terms of detection rate, enriched functional pathways, etc. Moreover, we identify several peripheral immune signatures that are suppressed in patients with cancer. Specifically, we establish a γδ-T cell score and a cancer-associated-fibroblast (CAF) score, providing insights into clinical statuses like cancer stage and survival. Overall, we reveal a cell-free multi-molecular landscape that is useful for blood monitoring in personalized cancer treatment.
Collapse
Affiliation(s)
- Yuhuan Tao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Shaozhen Xing
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Shuai Zuo
- Gastro-Intestinal Surgery, Peking University First Hospital, Beijing 100034, China
| | - Pengfei Bao
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Yunfan Jin
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Yu Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Mingyang Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Yingchao Wu
- Gastro-Intestinal Surgery, Peking University First Hospital, Beijing 100034, China
| | - Shanwen Chen
- Gastro-Intestinal Surgery, Peking University First Hospital, Beijing 100034, China
| | - Xiaojuan Wang
- Institute for Precision Medicine, Tsinghua University, Beijing 100084, China; Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, No. 168, Litang Road, Changping District, Beijing 102218, China
| | - Yumin Zhu
- Medical school, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Ying Feng
- Department of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiaohua Zhang
- Department of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xianbo Wang
- Department of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Qiaoran Xi
- MOE Key Laboratory of Protein Sciences, State Key Laboratory of Molecular Oncology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qian Lu
- Institute for Precision Medicine, Tsinghua University, Beijing 100084, China; Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, No. 168, Litang Road, Changping District, Beijing 102218, China.
| | - Pengyuan Wang
- Gastro-Intestinal Surgery, Peking University First Hospital, Beijing 100034, China.
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute for Precision Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
115
|
Li J, He D, Bi Y, Liu S. The Emerging Roles of Exosomal miRNAs in Breast Cancer Progression and Potential Clinical Applications. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:825-840. [PMID: 38020052 PMCID: PMC10658810 DOI: 10.2147/bctt.s432750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023]
Abstract
Breast cancer remains the leading malignancy in terms of morbidity and mortality today. The tumor microenvironment of breast cancer includes multiple cell types, secreted proteins, and signaling components such as exosomes. Among these, exosomes have a lipid bilayer structure. Exosomes can reflect the biological traits of the parent cell and carry a variety of biologically active components, including proteins, lipids, small molecules, and non-coding RNAs, which include miRNA, lncRNA, and circRNA. MiRNAs are a group of non-coding RNAs of approximately 20-23 nucleotides in length encoded by the genome, triggering silencing and functional repression of target genes. MiRNAs have been shown to play a significant role in the development of cancer owing to their role in the prognosis, pathogenesis, diagnosis, and treatment of cancer. MiRNAs in exosomes can serve as effective mediators of information transfer from parental cells to recipient cells and trigger changes in biological traits such as proliferation, invasion, migration, and drug resistance. These changes can profoundly alter the progression of breast cancer. Therefore, here, we systematically summarize the association of exosomal miRNAs on breast cancer progression, diagnosis, and treatment in the hope of providing novel strategies and directions for subsequent breast cancer treatment.
Collapse
Affiliation(s)
- Jie Li
- Department of Thyroid and Breast Surgery, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, People’s Republic of China
| | - Dejiao He
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, People’s Republic of China
| | - Yajun Bi
- Department of Pediatrics, Dalian Municipal Women and Children’s Medical Center (Group), Dalian Medical University, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Shengxuan Liu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, 430030, People’s Republic of China
| |
Collapse
|
116
|
Pescia C, Guerini-Rocco E, Viale G, Fusco N. Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies. Cancers (Basel) 2023; 15:5430. [PMID: 38001690 PMCID: PMC10670146 DOI: 10.3390/cancers15225430] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Early breast cancer (BC) is the definition applied to breast-confined tumors with or without limited involvement of locoregional lymph nodes. While risk stratification is essential for guiding clinical decisions, it can be a complex endeavor in these patients due to the absence of comprehensive guidelines. Histopathological analysis and biomarker assessment play a pivotal role in defining patient outcomes. Traditional histological criteria such as tumor size, lymph node involvement, histological type and grade, lymphovascular invasion, and immune cell infiltration are significant prognostic indicators. In addition to the hormone receptor, HER2, and-in specific scenarios-BRCA1/2 testing, molecular subtyping through gene expression profiling provides valuable insights to tailor clinical decision-making. The emergence of "omics" technologies, applicable to both tissue and liquid biopsy samples, has broadened our arsenal for evaluating the risk of early BC. However, a pressing need remains for standardized methodologies and integrated pathological models that encompass multiple analytical dimensions. In this study, we provide a detailed examination of the existing strategies for early BC risk stratification, intending to serve as a practical guide for histopathologists and molecular pathologists.
Collapse
Affiliation(s)
- Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- School of Pathology, University of Milan, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| |
Collapse
|
117
|
Justo-Garrido M, López-Saavedra A, Alcaraz N, Cortés-González CC, Oñate-Ocaña LF, Caro-Sánchez CHS, Castro-Hernández C, Arriaga-Canon C, Díaz-Chávez J, Herrera LA. Association of SLC12A1 and GLUR4 Ion Transporters with Neoadjuvant Chemoresistance in Luminal Locally Advanced Breast Cancer. Int J Mol Sci 2023; 24:16104. [PMID: 38003293 PMCID: PMC10670992 DOI: 10.3390/ijms242216104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/24/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Chemoresistance to standard neoadjuvant treatment commonly occurs in locally advanced breast cancer, particularly in the luminal subtype, which is hormone receptor-positive and represents the most common subtype of breast cancer associated with the worst outcomes. Identifying the genes associated with chemoresistance is crucial for understanding the underlying mechanisms and discovering effective treatments. In this study, we aimed to identify genes linked to neoadjuvant chemotherapy resistance in 62 retrospectively included patients with luminal breast cancer. Whole RNA sequencing of 12 patient biopsies revealed 269 differentially expressed genes in chemoresistant patients. We further validated eight highly correlated genes associated with resistance. Among these, solute carrier family 12 member 1 (SLC12A1) and glutamate ionotropic AMPA type subunit 4 (GRIA4), both implicated in ion transport, showed the strongest association with chemoresistance. Notably, SLC12A1 expression was downregulated, while protein levels of glutamate receptor 4 (GLUR4), encoded by GRIA4, were elevated in patients with a worse prognosis. Our results suggest a potential link between SLC12A1 gene expression and GLUR4 protein levels with chemoresistance in luminal breast cancer. In particular, GLUR4 protein could serve as a potential target for drug intervention to overcome chemoresistance.
Collapse
Affiliation(s)
- Montserrat Justo-Garrido
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
| | - Alejandro López-Saavedra
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
| | - Nicolás Alcaraz
- The Bioinformatics Centre, Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark;
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Carlo C. Cortés-González
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
| | - Luis F. Oñate-Ocaña
- Department of Gastroenterology, National Cancer Institute (INCan), Tlalpan, Mexico City 14080, Mexico;
| | | | - Clementina Castro-Hernández
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
| | - Cristian Arriaga-Canon
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
| | - José Díaz-Chávez
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
| | - Luis A. Herrera
- Cancer Research Unit, Institute of Biomedical Research, National Autonomous University of Mexico (UNAM)-National Institute of Cancerology, San Fernando Av #22, XVI Section, Mexico City 14080, Mexico; (M.J.-G.); (A.L.-S.); (C.C.C.-G.); (C.C.-H.); (C.A.-C.)
- School of Medicine and Health Sciences-Tecnológico de Monterrey, Mexico City 14380, Mexico
| |
Collapse
|
118
|
Rediti M, Fernandez-Martinez A, Venet D, Rothé F, Hoadley KA, Parker JS, Singh B, Campbell JD, Ballman KV, Hillman DW, Winer EP, El-Abed S, Piccart M, Di Cosimo S, Symmans WF, Krop IE, Salgado R, Loi S, Pusztai L, Perou CM, Carey LA, Sotiriou C. Immunological and clinicopathological features predict HER2-positive breast cancer prognosis in the neoadjuvant NeoALTTO and CALGB 40601 randomized trials. Nat Commun 2023; 14:7053. [PMID: 37923752 PMCID: PMC10624889 DOI: 10.1038/s41467-023-42635-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 10/16/2023] [Indexed: 11/06/2023] Open
Abstract
The identification of prognostic markers in patients receiving neoadjuvant therapy is crucial for treatment optimization in HER2-positive breast cancer, with the immune microenvironment being a key factor. Here, we investigate the complexity of B and T cell receptor (BCR and TCR) repertoires in the context of two phase III trials, NeoALTTO and CALGB 40601, evaluating neoadjuvant paclitaxel with trastuzumab and/or lapatinib in women with HER2-positive breast cancer. BCR features, particularly the number of reads and clones, evenness and Gini index, are heterogeneous according to hormone receptor status and PAM50 subtypes. Moreover, BCR measures describing clonal expansion, namely evenness and Gini index, are independent prognostic factors. We present a model developed in NeoALTTO and validated in CALGB 40601 that can predict event-free survival (EFS) by integrating hormone receptor and clinical nodal status, breast pathological complete response (pCR), stromal tumor-infiltrating lymphocyte levels (%) and BCR repertoire evenness. A prognostic score derived from the model and including those variables, HER2-EveNT, allows the identification of patients with 5-year EFS > 90%, and, in those not achieving pCR, of a subgroup of immune-enriched tumors with an excellent outcome despite residual disease.
Collapse
Affiliation(s)
- Mattia Rediti
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - David Venet
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Françoise Rothé
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | | | - Jordan D Campbell
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Karla V Ballman
- Alliance Statistics and Data Management Center, Weill Cornell Medicine, New York, NY, USA
| | - David W Hillman
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN, USA
| | - Eric P Winer
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | | | - Martine Piccart
- Medical Oncology Department, Institut Jules Bordet and l'Université Libre de Bruxelles (U.L.B.), Brussels, Belgium
| | - Serena Di Cosimo
- Integrated biology platform unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - William Fraser Symmans
- Department of Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ian E Krop
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Roberto Salgado
- Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sherene Loi
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lajos Pusztai
- Breast Medical Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Lisa A Carey
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (H.U.B), Université Libre de Bruxelles (ULB), Brussels, Belgium.
| |
Collapse
|
119
|
Wang Z, Yu L, Ding X, Liao X, Wang L. Shared-Specific Feature Learning With Bottleneck Fusion Transformer for Multi-Modal Whole Slide Image Analysis. IEEE TRANSACTIONS ON MEDICAL IMAGING 2023; 42:3374-3383. [PMID: 37335798 DOI: 10.1109/tmi.2023.3287256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
The fusion of multi-modal medical data is essential to assist medical experts to make treatment decisions for precision medicine. For example, combining the whole slide histopathological images (WSIs) and tabular clinical data can more accurately predict the lymph node metastasis (LNM) of papillary thyroid carcinoma before surgery to avoid unnecessary lymph node resection. However, the huge-sized WSI provides much more high-dimensional information than low-dimensional tabular clinical data, making the information alignment challenging in the multi-modal WSI analysis tasks. This paper presents a novel transformer-guided multi-modal multi-instance learning framework to predict lymph node metastasis from both WSIs and tabular clinical data. We first propose an effective multi-instance grouping scheme, named siamese attention-based feature grouping (SAG), to group high-dimensional WSIs into representative low-dimensional feature embeddings for fusion. We then design a novel bottleneck shared-specific feature transfer module (BSFT) to explore the shared and specific features between different modalities, where a few learnable bottleneck tokens are utilized for knowledge transfer between modalities. Moreover, a modal adaptation and orthogonal projection scheme were incorporated to further encourage BSFT to learn shared and specific features from multi-modal data. Finally, the shared and specific features are dynamically aggregated via an attention mechanism for slide-level prediction. Experimental results on our collected lymph node metastasis dataset demonstrate the efficiency of our proposed components and our framework achieves the best performance with AUC (area under the curve) of 97.34%, outperforming the state-of-the-art methods by over 1.27%.
Collapse
|
120
|
Zhang Y, Huo J, Yu S, Feng W, Tuersun A, Chen F, Lv Z, Liu W, Zhao J, Xu Z, Lu A, Zong Y. Colorectal cancer tissue-originated spheroids reveal tumor intrinsic signaling pathways and mimic patient clinical chemotherapeutic response as a rapid and valid model. Biomed Pharmacother 2023; 167:115585. [PMID: 37774672 DOI: 10.1016/j.biopha.2023.115585] [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: 07/19/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
Locally advanced colorectal cancer requires preoperative chemotherapy to reduce local recurrence and metastasis rates, but it remains difficult to predict the tumor will be sensitive to which treatments. The patient-derived organoids (PDOs) are considered an effective platform for predicting tumor drug responses in precision oncology. However, it has the limitation of being time-consuming in practical applications, especially in neoadjuvant treatment. Here we used cancer tissue-originated spheroids (CTOS) method to establish organoids from a heterogeneous population of colorectal cancer specimens, and evaluated the capacity of CTOS to predict clinical drug responses. By analyzing the relationship of the activities of drug-treated CTOS, drug targets and target-related pathways, tumor intrinsic effective-target-related pathways can be identified. These pathways were highly matched to the abnormal pathways indicated by whole-exome sequencing. Based on this, we used half effective concentration gradients to classify CTOS as sensitive or resistant to chemotherapy regimens within a week, for predicting neoadjuvant treatment outcomes for colorectal cancer patients. The drug sensitivity test results are highly matched to the clinical responses to treatment in individual patients. Thus, our data suggested that CTOS models can be effectively screened ex vivo to identify pathways sensitive to chemotherapies. These data also supported organoid research for personalized clinical medication guidance immediately after diagnosis in patients with advanced colorectal cancer.
Collapse
Affiliation(s)
- Yuchen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianting Huo
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Suyue Yu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenqing Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Abudumaimaitijiang Tuersun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fangqian Chen
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zeping Lv
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wangyi Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingkun Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhuoqing Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Aiguo Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Yaping Zong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
121
|
Zhou S, Sun D, Mao W, Liu Y, Cen W, Ye L, Liang F, Xu J, Shi H, Ji Y, Wang L, Chang W. Deep radiomics-based fusion model for prediction of bevacizumab treatment response and outcome in patients with colorectal cancer liver metastases: a multicentre cohort study. EClinicalMedicine 2023; 65:102271. [PMID: 37869523 PMCID: PMC10589780 DOI: 10.1016/j.eclinm.2023.102271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Background Accurate tumour response prediction to targeted therapy allows for personalised conversion therapy for patients with unresectable colorectal cancer liver metastases (CRLM). In this study, we aimed to develop and validate a multi-modal deep learning model to predict the efficacy of bevacizumab in patients with initially unresectable CRLM using baseline PET/CT, clinical data, and colonoscopy biopsy specimens. Methods In this multicentre cohort study, we retrospectively collected data of 307 patients with CRLM from the BECOME study (NCT01972490) (Zhongshan Hospital of Fudan University, Shanghai) and two independent Chinese cohorts (internal validation cohort from January 1, 2018 to December 31, 2018 at Zhongshan Hospital of Fudan University; external validation cohort from January 1, 2020 to December 31, 2020 at Zhongshan Hospital-Xiamen, Shanghai, and the First Hospital of Wenzhou Medical University, Wenzhou). The main inclusion criteria were that patients with CRLM had pre-treatment PET/CT images as well as colonoscopy specimens. After extracting PET/CT features with deep neural networks (DNN) and selecting related clinical factors using LASSO analysis, a random forest classifier was built as the Deep Radiomics Bevacizumab efficacy predicting model (DERBY). Furthermore, by combining histopathological biomarkers into DERBY, we established DERBY+. The performance of model was evaluated using area under the curve (AUC), sensitivity, specificity, positive predictive value, and negative predictive value. Findings DERBY achieved promising performance in predicting bevacizumab sensitivity with an AUC of 0.77 and 95% confidence interval (CI) [0.67-0.87]. After combining histopathological features, we developed DERBY+, which had more robust accuracy for predicting tumour response in external validation cohort (AUC 0.83 and 95% CI [0.75-0.92], sensitivity 80.4%, specificity 76.8%). DERBY+ also had prognostic value: the responders had longer progression-free survival (median progression-free survival: 9.6 vs 6.3 months, p = 0.002) and overall survival (median overall survival: 27.6 vs 18.5 months, p = 0.010) than non-responders. Interpretation This multi-modal deep radiomics model, using PET/CT, clinical data and histopathological data, was able to identify patients with bevacizumab-sensitive CRLM, providing a favourable approach for precise patient treatment. To further validate and explore the clinical impact of this work, future prospective studies with larger patient cohorts are warranted. Funding The National Natural Science Foundation of China; Fujian Provincial Health Commission Project; Xiamen Science and Technology Agency Program; Clinical Research Plan of SHDC; Shanghai Science and Technology Committee Project; Clinical Research Plan of SHDC; Zhejiang Provincial Natural Science Foundation of China; and National Science Foundation of Xiamen.
Collapse
Affiliation(s)
- Shizhao Zhou
- Department of General Surgery, Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dazhen Sun
- Department of Automation, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wujian Mao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yu Liu
- Department of General Surgery, Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Cen
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Lechi Ye
- Department of Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianmin Xu
- Department of General Surgery, Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lisheng Wang
- Department of Automation, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wenju Chang
- Department of General Surgery, Department of Colorectal Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of General Surgery, Zhongshan Hospital (Xiamen Branch), Fudan University, Xiamen, Fujian, 361015, China
| |
Collapse
|
122
|
Yan S, Li J, Wu W. Artificial intelligence in breast cancer: application and future perspectives. J Cancer Res Clin Oncol 2023; 149:16179-16190. [PMID: 37656245 DOI: 10.1007/s00432-023-05337-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023]
Abstract
Breast cancer is one of the most common cancers and is one of the leading causes of cancer-related deaths in women worldwide. Early diagnosis and treatment are the key for a favorable prognosis. The application of artificial intelligence technology in the medical field is increasingly extensive, including image analysis, automated diagnosis, intelligent pharmaceutical system, personalized treatment and so on. AI-based breast cancer imaging, pathology and adjuvant therapy technology cannot only reduce the workload of clinicians, but also continuously improve the accuracy and sensitivity of breast cancer diagnosis and treatment. This paper reviews the application of AI in breast cancer, as well as looks ahead and poses challenges to the future development of AI for breast cancer detection and therapeutic, so as to provide ideas for future research.
Collapse
Affiliation(s)
- Shuixin Yan
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Jiadi Li
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Weizhu Wu
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
123
|
Zhong X, Lin Y, Zhang W, Bi Q. Predicting diagnosis and survival of bone metastasis in breast cancer using machine learning. Sci Rep 2023; 13:18301. [PMID: 37880320 PMCID: PMC10600146 DOI: 10.1038/s41598-023-45438-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023] Open
Abstract
This study aimed at establishing more accurate predictive models based on novel machine learning algorithms, with the overarching goal of providing clinicians with effective decision-making assistance. We retrospectively analyzed the breast cancer patients recorded in the Surveillance, Epidemiology, and End Results (SEER) database from 2010 to 2016. Multivariable logistic regression analyses were used to identify risk factors for bone metastases in breast cancer, whereas Cox proportional hazards regression analyses were used to identify prognostic factors for breast cancer with bone metastasis (BCBM). Based on the identified risk and prognostic factors, we developed diagnostic and prognostic models that incorporate six machine learning classifiers. We then used the area under the receiver operating characteristic (ROC) curve (AUC), learning curve, precision curve, calibration plot, and decision curve analysis to evaluate performance of the machine learning models. Univariable and multivariable logistic regression analyses showed that bone metastases were significantly associated with age, race, sex, grade, T stage, N stage, surgery, radiotherapy, chemotherapy, tumor size, brain metastasis, liver metastasis, lung metastasis, breast subtype, and PR. Univariate and multivariate Cox regression analyses revealed that age, race, marital status, grade, surgery, radiotherapy, chemotherapy, brain metastasis, liver metastasis, lung metastasis, breast subtype, ER, and PR were closely associated with the prognosis of BCBM. Among the six machine learning models, the XGBoost algorithm predicted the most accurate results (Diagnostic model AUC = 0.98; Prognostic model AUC = 0.88). According to the Shapley additive explanations (SHAP), the most critical feature of the diagnostic model was surgery, followed by N stage. Interestingly, surgery was also the most critical feature of prognostic model, followed by liver metastasis. Based on the XGBoost algorithm, we could effectively predict the diagnosis and survival of bone metastasis in breast cancer and provide targeted references for the treatment of BCBM patients.
Collapse
Affiliation(s)
- Xugang Zhong
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital Affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Yanze Lin
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China
| | - Wei Zhang
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital Affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China.
- Department of Orthopedics, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People's Republic of China.
| | - Qing Bi
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital Affiliated to Qingdao University, Qingdao, Shandong, People's Republic of China.
- Center for Rehabilitation Medicine, Cancer Center, Department of Orthopedics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, People's Republic of China.
| |
Collapse
|
124
|
Blise KE, Sivagnanam S, Betts CB, Betre K, Kirchberger N, Tate B, Furth EE, Dias Costa A, Nowak JA, Wolpin BM, Vonderheide RH, Goecks J, Coussens LM, Byrne KT. Machine learning links T cell function and spatial localization to neoadjuvant immunotherapy and clinical outcome in pancreatic cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563335. [PMID: 37961410 PMCID: PMC10634700 DOI: 10.1101/2023.10.20.563335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Tumor molecular datasets are becoming increasingly complex, making it nearly impossible for humans alone to effectively analyze them. Here, we demonstrate the power of using machine learning to analyze a single-cell, spatial, and highly multiplexed proteomic dataset from human pancreatic cancer and reveal underlying biological mechanisms that may contribute to clinical outcome. A novel multiplex immunohistochemistry antibody panel was used to audit T cell functionality and spatial localization in resected tumors from treatment-naive patients with localized pancreatic ductal adenocarcinoma (PDAC) compared to a second cohort of patients treated with neoadjuvant agonistic CD40 (αCD40) monoclonal antibody therapy. In total, nearly 2.5 million cells from 306 tissue regions collected from 29 patients across both treatment cohorts were assayed, and more than 1,000 tumor microenvironment (TME) features were quantified. We then trained machine learning models to accurately predict αCD40 treatment status and disease-free survival (DFS) following αCD40 therapy based upon TME features. Through downstream interpretation of the machine learning models' predictions, we found αCD40 therapy to reduce canonical aspects of T cell exhaustion within the TME, as compared to treatment-naive TMEs. Using automated clustering approaches, we found improved DFS following αCD40 therapy to correlate with the increased presence of CD44+ CD4+ Th1 cells located specifically within cellular spatial neighborhoods characterized by increased T cell proliferation, antigen-experience, and cytotoxicity in immune aggregates. Overall, our results demonstrate the utility of machine learning in molecular cancer immunology applications, highlight the impact of αCD40 therapy on T cells within the TME, and identify potential candidate biomarkers of DFS for αCD40-treated patients with PDAC.
Collapse
Affiliation(s)
- Katie E. Blise
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR USA
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Shamilene Sivagnanam
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR USA
| | - Courtney B. Betts
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR USA
- Current affiliation: Akoya Biosciences, 100 Campus Drive, 6th Floor, Marlborough, MA USA
| | - Konjit Betre
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR USA
| | - Nell Kirchberger
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR USA
| | - Benjamin Tate
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
- Immune Monitoring and Cancer Omics Services, Oregon Health & Science University, Portland, OR USA
| | - Emma E. Furth
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Andressa Dias Costa
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA USA
| | - Jonathan A. Nowak
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Brian M. Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA USA
| | - Robert H. Vonderheide
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
- Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Jeremy Goecks
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR USA
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
- Current affiliation: Department of Machine Learning, H. Lee Moffitt Cancer Center, Tampa, FL USA
- Current affiliation: Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, FL USA
| | - Lisa M. Coussens
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR USA
| | - Katelyn T. Byrne
- The Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR USA
- Lead contact
| |
Collapse
|
125
|
Ivanisevic T, Sewduth RN. Multi-Omics Integration for the Design of Novel Therapies and the Identification of Novel Biomarkers. Proteomes 2023; 11:34. [PMID: 37873876 PMCID: PMC10594525 DOI: 10.3390/proteomes11040034] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023] Open
Abstract
Multi-omics is a cutting-edge approach that combines data from different biomolecular levels, such as DNA, RNA, proteins, metabolites, and epigenetic marks, to obtain a holistic view of how living systems work and interact. Multi-omics has been used for various purposes in biomedical research, such as identifying new diseases, discovering new drugs, personalizing treatments, and optimizing therapies. This review summarizes the latest progress and challenges of multi-omics for designing new treatments for human diseases, focusing on how to integrate and analyze multiple proteome data and examples of how to use multi-proteomics data to identify new drug targets. We also discussed the future directions and opportunities of multi-omics for developing innovative and effective therapies by deciphering proteome complexity.
Collapse
Affiliation(s)
| | - Raj N. Sewduth
- VIB-KU Leuven Center for Cancer Biology (VIB), 3000 Leuven, Belgium;
| |
Collapse
|
126
|
Liu J, Wang H, Wu P, Wang J, Wang J, Hou H, Wang J, Zhang Y. A simplified frailty index and nomogram to predict the postoperative complications and survival in older patients with upper urinary tract urothelial carcinoma. Front Oncol 2023; 13:1187677. [PMID: 37901313 PMCID: PMC10600399 DOI: 10.3389/fonc.2023.1187677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 09/07/2023] [Indexed: 10/31/2023] Open
Abstract
Purpose This study was designed to investigate the clinical value of a simplified five-item frailty index (sFI) for predicting short- and long-term outcomes in older patients with upper urinary tract urothelial carcinoma (UTUC) patients after radical nephroureterectomy (RNU). Method This retrospective study included 333 patients (aged ≥65 years) with UTUC. Patients were classified into five groups: 0, 1, 2, 3, and 3+, according to sFI score. The variable importance and minimum depth methods were used to screen for significant variables, and univariable and multivariable logistic regression models applied to investigated the relationships between significant variables and postoperative complications. Survival differences between groups were analyzed using Kaplan-Meier plots and log-rank tests. Cox proportional hazards regression was used to evaluate risk factors associated with overall survival (OS) and cancer-specific survival (CSS). Further, we developed a nomogram based on clinicopathological features and the sFI. The area under the curve (AUC), Harrel's concordance index (C-index), calibration curve, and decision curve analysis (DCA) were used to evaluate the nomogram. Result Of 333 cases identified, 31.2% experienced a Clavien-Dindo grade of 2 or greater complication. Random forest-logistic regression modeling showed that sFI significantly influenced the incidence of postoperative complications in older patients (AUC= 0.756). Compared with patients with low sFI score, those with high sFI scores had significantly lower OS and CSS (p < 0.001). Across all patients, the random survival forest-Cox regression model revealed that sFI score was an independent prognostic factor for OS and CSS, with AUC values of 0.815 and 0.823 for predicting 3-year OS and CSS, respectively. The nomogram developed was clinically valuable and had good ability to discriminate abilities for high-risk patients. Further, we developed a survival risk classification system that divided all patients into high-, moderate-, and low-risk groups based on total nomogram points for each patient. Conclusion A simple five-item frailty index may be considered a prognostic factor for the prognosis and postoperative complications of UTUC following RNU. By using this predictive model, clinicians may increase their accuracy in predicting complications and prognosis and improve preoperative decision-making.
Collapse
Affiliation(s)
- Jianyong Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Haoran Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Pengjie Wu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Jiawen Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Jianye Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Huimin Hou
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Jianlong Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| | - Yaoguang Zhang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Institute of the Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Hospital Continence Center, Beijing, China
| |
Collapse
|
127
|
Steenbruggen TG, Wolf DM, Campbell MJ, Sanders J, Cornelissen S, Thijssen B, Salgado RA, Yau C, O-Grady N, Basu A, Bhaskaran R, Mittempergher L, Hirst GL, Coppe JP, Kok M, Sonke GS, van 't Veer LJ, Horlings HM. B-cells and regulatory T-cells in the microenvironment of HER2+ breast cancer are associated with decreased survival: a real-world analysis of women with HER2+ metastatic breast cancer. Breast Cancer Res 2023; 25:117. [PMID: 37794508 PMCID: PMC10552219 DOI: 10.1186/s13058-023-01717-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 09/21/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND Despite major improvements in treatment of HER2-positive metastatic breast cancer (MBC), only few patients achieve complete remission and remain progression free for a prolonged time. The tumor immune microenvironment plays an important role in the response to treatment in HER2-positive breast cancer and could contain valuable prognostic information. Detailed information on the cancer-immune cell interactions in HER2-positive MBC is however still lacking. By characterizing the tumor immune microenvironment in patients with HER2-positive MBC, we aimed to get a better understanding why overall survival (OS) differs so widely and which alternative treatment approaches may improve outcome. METHODS We included all patients with HER2-positive MBC who were treated with trastuzumab-based palliative therapy in the Netherlands Cancer Institute between 2000 and 2014 and for whom pre-treatment tissue from the primary tumor or from metastases was available. Infiltrating immune cells and their spatial relationships to one another and to tumor cells were characterized by immunohistochemistry and multiplex immunofluorescence. We also evaluated immune signatures and other key pathways using next-generation RNA-sequencing data. With nine years median follow-up from initial diagnosis of MBC, we investigated the association between tumor and immune characteristics and outcome. RESULTS A total of 124 patients with 147 samples were included and evaluated. The different technologies showed high correlations between each other. T-cells were less prevalent in metastases compared to primary tumors, whereas B-cells and regulatory T-cells (Tregs) were comparable between primary tumors and metastases. Stromal tumor-infiltrating lymphocytes in general were not associated with OS. The infiltration of B-cells and Tregs in the primary tumor was associated with unfavorable OS. Four signatures classifying the extracellular matrix of primary tumors showed differential survival in the population as a whole. CONCLUSIONS In a real-world cohort of 124 patients with HER2-positive MBC, B-cells, and Tregs in primary tumors are associated with unfavorable survival. With this paper, we provide a comprehensive insight in the tumor immune microenvironment that could guide further research into development of novel immunomodulatory strategies.
Collapse
Affiliation(s)
- Tessa G Steenbruggen
- Department of Medical Oncology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands.
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94115, USA.
| | - Denise M Wolf
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Michael J Campbell
- Department of Surgery, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
| | - Sten Cornelissen
- Core Facility Molecular Pathology and Biobanking, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
| | - Bram Thijssen
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
| | - Roberto A Salgado
- Department of Pathology, GZA-ZNA Hospitals, 2020, Antwerp, Belgium
- Division of Research, Peter Mac Callum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Christina Yau
- Department of Surgery, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Nick O-Grady
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Amrita Basu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Rajith Bhaskaran
- Research and Development, Agendia N.V, 1043 NT, Amsterdam, North Holland, The Netherlands
| | - Lorenza Mittempergher
- Research and Development, Agendia N.V, 1043 NT, Amsterdam, North Holland, The Netherlands
| | - Gillian L Hirst
- Department of Surgery, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Jean-Philippe Coppe
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Marleen Kok
- Department of Medical Oncology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
- Department of Clinical Oncology, University of Amsterdam, 1012 WX, Amsterdam, North Holland, The Netherlands
| | - Laura J van 't Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, 94115, USA
| | - Hugo M Horlings
- Department of Pathology, The Netherlands Cancer Institute, 1066 CX, Amsterdam, North Holland, The Netherlands
| |
Collapse
|
128
|
Ahn JS, Shin S, Yang SA, Park EK, Kim KH, Cho SI, Ock CY, Kim S. Artificial Intelligence in Breast Cancer Diagnosis and Personalized Medicine. J Breast Cancer 2023; 26:405-435. [PMID: 37926067 PMCID: PMC10625863 DOI: 10.4048/jbc.2023.26.e45] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Breast cancer is a significant cause of cancer-related mortality in women worldwide. Early and precise diagnosis is crucial, and clinical outcomes can be markedly enhanced. The rise of artificial intelligence (AI) has ushered in a new era, notably in image analysis, paving the way for major advancements in breast cancer diagnosis and individualized treatment regimens. In the diagnostic workflow for patients with breast cancer, the role of AI encompasses screening, diagnosis, staging, biomarker evaluation, prognostication, and therapeutic response prediction. Although its potential is immense, its complete integration into clinical practice is challenging. Particularly, these challenges include the imperatives for extensive clinical validation, model generalizability, navigating the "black-box" conundrum, and pragmatic considerations of embedding AI into everyday clinical environments. In this review, we comprehensively explored the diverse applications of AI in breast cancer care, underlining its transformative promise and existing impediments. In radiology, we specifically address AI in mammography, tomosynthesis, risk prediction models, and supplementary imaging methods, including magnetic resonance imaging and ultrasound. In pathology, our focus is on AI applications for pathologic diagnosis, evaluation of biomarkers, and predictions related to genetic alterations, treatment response, and prognosis in the context of breast cancer diagnosis and treatment. Our discussion underscores the transformative potential of AI in breast cancer management and emphasizes the importance of focused research to realize the full spectrum of benefits of AI in patient care.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.
| |
Collapse
|
129
|
Wei L, Niraula D, Gates EDH, Fu J, Luo Y, Nyflot MJ, Bowen SR, El Naqa IM, Cui S. Artificial intelligence (AI) and machine learning (ML) in precision oncology: a review on enhancing discoverability through multiomics integration. Br J Radiol 2023; 96:20230211. [PMID: 37660402 PMCID: PMC10546458 DOI: 10.1259/bjr.20230211] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 09/05/2023] Open
Abstract
Multiomics data including imaging radiomics and various types of molecular biomarkers have been increasingly investigated for better diagnosis and therapy in the era of precision oncology. Artificial intelligence (AI) including machine learning (ML) and deep learning (DL) techniques combined with the exponential growth of multiomics data may have great potential to revolutionize cancer subtyping, risk stratification, prognostication, prediction and clinical decision-making. In this article, we first present different categories of multiomics data and their roles in diagnosis and therapy. Second, AI-based data fusion methods and modeling methods as well as different validation schemes are illustrated. Third, the applications and examples of multiomics research in oncology are demonstrated. Finally, the challenges regarding the heterogeneity data set, availability of omics data, and validation of the research are discussed. The transition of multiomics research to real clinics still requires consistent efforts in standardizing omics data collection and analysis, building computational infrastructure for data sharing and storing, developing advanced methods to improve data fusion and interpretability, and ultimately, conducting large-scale prospective clinical trials to fill the gap between study findings and clinical benefits.
Collapse
Affiliation(s)
- Lise Wei
- Department of Radiation Oncology, University of Michigan, Michigan, United States
| | - Dipesh Niraula
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Evan D. H. Gates
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Jie Fu
- Department of Radiation Oncology, Stanford University, Stanford, California, United States
| | - Yi Luo
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Matthew J. Nyflot
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Stephen R. Bowen
- Department of Radiation Oncology, University of Washington, Washington, United States
| | - Issam M El Naqa
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, United States
| | - Sunan Cui
- Department of Radiation Oncology, University of Washington, Washington, United States
| |
Collapse
|
130
|
Fürstberger A, Ikonomi N, Kestler AMR, Marienfeld R, Schwab JD, Kuhn P, Seufferlein T, Kestler HA. AMBAR - Interactive Alteration annotations for molecular tumor boards. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 240:107697. [PMID: 37441893 DOI: 10.1016/j.cmpb.2023.107697] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 05/23/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023]
Abstract
MOTIVATION Personalized decision-making for cancer therapy relies on molecular profiling from sequencing data in combination with database evidence and expert knowledge. Molecular tumor boards (MTBs) bring together clinicians and scientists with diverse expertise and are increasingly established in the clinical routine for therapeutic interventions. However, the analysis and documentation of patients data are still time-consuming and difficult to manage for MTBs, especially as few tools are available for the amount of information required. RESULTS To overcome these limitations, we developed an interactive web application AMBAR (Alteration annotations for Molecular tumor BoARds), for therapeutic decision-making support in MTBs. AMBAR is an R shiny-based application that allows customization, interactive filtering, visualization, adding expert knowledge, and export to clinical systems of annotated mutations. AVAILABILITY AMBAR is dockerized, open source and available at https://sysbio.uni-ulm.de/?Software:Ambar Contact:hans.kestler@uni-ulm.de.
Collapse
Affiliation(s)
- Axel Fürstberger
- Institute of Medical Systems Biology, Ulm University, Ulm 89081, Germany; Department of Pathology, Ulm University Hospital, Ulm 89081, Germany; Zentrum Personalisierte Medizin, Ulm University Hospital, Ulm 89081, Germany
| | - Nensi Ikonomi
- Institute of Medical Systems Biology, Ulm University, Ulm 89081, Germany
| | - Angelika M R Kestler
- Department of Internal Medicine I, Ulm University Hospital, Ulm 89081, Germany; Zentrum Personalisierte Medizin, Ulm University Hospital, Ulm 89081, Germany
| | - Ralf Marienfeld
- Department of Pathology, Ulm University Hospital, Ulm 89081, Germany; Zentrum Personalisierte Medizin, Ulm University Hospital, Ulm 89081, Germany
| | - Julian D Schwab
- Institute of Medical Systems Biology, Ulm University, Ulm 89081, Germany
| | - Peter Kuhn
- Comprehensive Cancer Center, Ulm University Hospital, Ulm 89081, Germany; Zentrum Personalisierte Medizin, Ulm University Hospital, Ulm 89081, Germany
| | - Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm 89081, Germany; Zentrum Personalisierte Medizin, Ulm University Hospital, Ulm 89081, Germany
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm 89081, Germany; Zentrum Personalisierte Medizin, Ulm University Hospital, Ulm 89081, Germany.
| |
Collapse
|
131
|
Luo J, Li X, Wei KL, Chen G, Xiong DD. Advances in the application of computational pathology in diagnosis, immunomicroenvironment recognition, and immunotherapy evaluation of breast cancer: a narrative review. J Cancer Res Clin Oncol 2023; 149:12535-12542. [PMID: 37389595 DOI: 10.1007/s00432-023-05002-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND Breast cancer (BC) is a prevalent and highly lethal malignancy affecting women worldwide. Immunotherapy has emerged as a promising therapeutic strategy for BC, offering potential improvements in patient survival. Neoadjuvant therapy (NAT) has also gained significant clinical traction. With the advancement of computer technology, Artificial Intelligence (AI) has been increasingly applied in pathology research, expanding and redefining the scope of the field. This narrative review aims to provide a comprehensive overview of the current literature on the application of computational pathology in BC, specifically focusing on diagnosis, immune microenvironment recognition, and the evaluation of immunotherapy and NAT response. METHODS A thorough examination of relevant literature was conducted, focusing on studies investigating the role of computational pathology in BC diagnosis, immune microenvironment recognition, and immunotherapy and NAT assessment. RESULTS The application of computational pathology has shown significant potential in BC management. AI-based techniques enable improved diagnosis and classification of BC subtypes, enhance the identification and characterization of the immune microenvironment, and facilitate the evaluation of immunotherapy and NAT response. However, challenges related to data quality, standardization, and algorithm development still need to be addressed. CONCLUSION The integration of computational pathology and AI has transformative implications for BC patient care. By leveraging AI-based technologies, clinicians can make more informed decisions in diagnosis, treatment planning, and therapeutic response assessment. Future research should focus on refining AI algorithms, addressing technical challenges, and conducting large-scale clinical validation studies to facilitate the translation of computational pathology into routine clinical practice for BC patients.
Collapse
Affiliation(s)
- Jie Luo
- Department of Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, People's Republic of China
| | - Xia Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Kang-Lai Wei
- Department of Pathology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, People's Republic of China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Dan-Dan Xiong
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
132
|
Zhou C, Xie H, Zhu F, Yan W, Yu R, Wang Y. Improving the malignancy prediction of breast cancer based on the integration of radiomics features from dual-view mammography and clinical parameters. Clin Exp Med 2023; 23:2357-2368. [PMID: 36413273 DOI: 10.1007/s10238-022-00944-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/05/2022] [Indexed: 11/23/2022]
Abstract
Radiomics has been a promising imaging biomarker for many malignant diseases. We developed a novel radiomics strategy that incorporating radiomics features extracted from dual-view mammograms and clinical parameters for identifying benign and malignant breast lesions, and validated whether the radiomics assessment could improve the accurate diagnosis of breast cancer. A total of 380 patients (mean age, 52 ± 7 years) with 621 breast lesions utilizing mammograms on craniocaudal (CC) and mediolateral oblique (MLO) views were randomly allocated into the training (n = 486) and testing (n = 135) sets in this retrospective study. A total of 1184 and 2368 radiomics features were extracted from single-position region of interest (ROI) and position-paired ROI, separately. Clinical parameters were then combined for better prediction. Recursive feature elimination and least absolute shrinkage and selection operator methods were applied to select optimal predictive features. Random forest was used to conduct the predictive model. Intraclass correlation coefficient test was used to assess repeatability and reproducibility of features. After preprocessing, 467 radiomics features and clinical parameters remained in the single-view and dual-view models. The performance and significance of models were quantified by the area under the curve (AUC), sensitivity, specificity, and accuracy. The correlation analysis between variables was evaluated using the correlation ratio and Pearson correlation coefficient. The model using a combination of dual-view radiomics and clinical parameters achieved a favorable performance (AUC: 0.804, 95% CI: 0.668-0.916), outperformed single-view model and model without clinical parameters. Incorporating with radiomics features of dual-view (CC&MLO) mammogram, age, breast density, and type of suspicious lesions can provide a noninvasive approach to evaluate the malignancy of breast lesions and facilitate clinical decision-making.
Collapse
Affiliation(s)
- Chenyi Zhou
- Department of Radiology, The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Hui Xie
- Department of Radiology, The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Fanglian Zhu
- Department of Radiology, The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China
| | - Wanying Yan
- Beijing Infervision Technology Co. Ltd., Beijing, 100025, Beijing, China
| | - Ruize Yu
- Beijing Infervision Technology Co. Ltd., Beijing, 100025, Beijing, China
| | - Yanling Wang
- Department of Radiology, The People's Hospital of Suzhou New District, Suzhou, 215129, Jiangsu, China.
| |
Collapse
|
133
|
Nukui T, Onogi A. An R package for ensemble learning stacking. BIOINFORMATICS ADVANCES 2023; 3:vbad139. [PMID: 37818175 PMCID: PMC10561990 DOI: 10.1093/bioadv/vbad139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/05/2023] [Accepted: 09/28/2023] [Indexed: 10/12/2023]
Abstract
Summary Supervised learning is widely used in biology for prediction, and ensemble learning, including stacking, is a promising technique for increasing and stabilizing the prediction accuracy. In this study, we developed an R package for stacking. This package depends on the R package caret and can handle models supported by caret. Stacking involves cross-validation of training data with multiple base learners, and the predicted values are used as explanatory variables for the meta-learner. In the prediction, the testing data were fed into the base models, and the returned values were averaged for each base learner. The averaged values were then fed into the meta-model, and the final predictions were returned. Using this package, the training and prediction procedures for stacking can be conducted using one-row scripts. Availability and implementation The R package stacking is available at the Comprehensive R Archive Network (CRAN) (https://cran.r-project.org/) and GitHub (https://github.com/Onogi/stacking). R scripts to reproduce the presented results are also reposited at GitHub.
Collapse
Affiliation(s)
- Taichi Nukui
- Department of Life Sciences, Faculty of Agriculture, Ryukoku University, Otsu, Shiga 520-2194, Japan
| | - Akio Onogi
- Department of Life Sciences, Faculty of Agriculture, Ryukoku University, Otsu, Shiga 520-2194, Japan
| |
Collapse
|
134
|
Affiliation(s)
- David J Hunter
- From the Nuffield Department of Population Health (D.J.H.) and the Department of Statistics and Nuffield Department of Medicine (C.H.), University of Oxford, Oxford, and the Alan Turing Institute, London (C.H.) - both in the United Kingdom
| | - Christopher Holmes
- From the Nuffield Department of Population Health (D.J.H.) and the Department of Statistics and Nuffield Department of Medicine (C.H.), University of Oxford, Oxford, and the Alan Turing Institute, London (C.H.) - both in the United Kingdom
| |
Collapse
|
135
|
Cosgrove N, Eustace AJ, O'Donovan P, Madden SF, Moran B, Crown J, Moulton B, Morris PG, Grogan L, Breathnach O, Power C, Allen M, Walshe JM, Hill AD, Blümel A, O'Connor D, Das S, Milewska M, Fay J, Kay E, Toomey S, Hennessy BT, Furney SJ. Predictive modelling of response to neoadjuvant therapy in HER2+ breast cancer. NPJ Breast Cancer 2023; 9:72. [PMID: 37758711 PMCID: PMC10533568 DOI: 10.1038/s41523-023-00572-9] [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: 12/21/2022] [Accepted: 07/26/2023] [Indexed: 09/29/2023] Open
Abstract
HER2-positive (HER2+) breast cancer accounts for 20-25% of all breast cancers. Predictive biomarkers of neoadjuvant therapy response are needed to better identify patients with early stage disease who may benefit from tailored treatments in the adjuvant setting. As part of the TCHL phase-II clinical trial (ICORG10-05/NCT01485926) whole exome DNA sequencing was carried out on normal-tumour pairs collected from 22 patients. Here we report predictive modelling of neoadjuvant therapy response using clinicopathological and genomic features of pre-treatment tumour biopsies identified age, estrogen receptor (ER) status and level of immune cell infiltration may together be important for predicting response. Clonal evolution analysis of longitudinally collected tumour samples show subclonal diversity and dynamics are evident with potential therapy resistant subclones detected. The sources of greater pre-treatment immunogenicity associated with a pathological complete response is largely unexplored in HER2+ tumours. However, here we point to the possibility of APOBEC associated mutagenesis, specifically in the ER-neg/HER2+ subtype as a potential mediator of this immunogenic phenotype.
Collapse
Affiliation(s)
- Nicola Cosgrove
- Genomic Oncology Research Group, Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Alex J Eustace
- School of Biotechnology, National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Peter O'Donovan
- Genomic Oncology Research Group, Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Stephen F Madden
- Data Science Centre, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Bruce Moran
- Conway Institute, University College Dublin, Dublin, Ireland
| | - John Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Brian Moulton
- Clinical Oncology Development Europe, Dublin, Ireland
| | - Patrick G Morris
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Liam Grogan
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Oscar Breathnach
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Colm Power
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Michael Allen
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Janice M Walshe
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - Arnold D Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Anna Blümel
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Darren O'Connor
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Sudipto Das
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Małgorzata Milewska
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, 9, Ireland
| | - Joanna Fay
- RCSI Biobank Service, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, 9, Ireland
| | - Elaine Kay
- Department of Pathology, RCSI University of Medicine and Health Sciences, Beaumont Hospital, Dublin, 9, Ireland
| | - Sinead Toomey
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, 9, Ireland
| | - Bryan T Hennessy
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland.
- Medical Oncology Group, Department of Molecular Medicine, Royal College of Surgeons in Ireland, Dublin, 9, Ireland.
| | - Simon J Furney
- Genomic Oncology Research Group, Department of Physiology and Medical Physics, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| |
Collapse
|
136
|
Tran KA, Addala V, Johnston RL, Lovell D, Bradley A, Koufariotis LT, Wood S, Wu SZ, Roden D, Al-Eryani G, Swarbrick A, Williams ED, Pearson JV, Kondrashova O, Waddell N. Performance of tumour microenvironment deconvolution methods in breast cancer using single-cell simulated bulk mixtures. Nat Commun 2023; 14:5758. [PMID: 37717006 PMCID: PMC10505141 DOI: 10.1038/s41467-023-41385-5] [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: 07/31/2022] [Accepted: 09/01/2023] [Indexed: 09/18/2023] Open
Abstract
Cells within the tumour microenvironment (TME) can impact tumour development and influence treatment response. Computational approaches have been developed to deconvolve the TME from bulk RNA-seq. Using scRNA-seq profiling from breast tumours we simulate thousands of bulk mixtures, representing tumour purities and cell lineages, to compare the performance of nine TME deconvolution methods (BayesPrism, Scaden, CIBERSORTx, MuSiC, DWLS, hspe, CPM, Bisque, and EPIC). Some methods are more robust in deconvolving mixtures with high tumour purity levels. Most methods tend to mis-predict normal epithelial for cancer epithelial as tumour purity increases, a finding that is validated in two independent datasets. The breast cancer molecular subtype influences this mis-prediction. BayesPrism and DWLS have the lowest combined numbers of false positives and false negatives, and have the best performance when deconvolving granular immune lineages. Our findings highlight the need for more single-cell characterisation of rarer cell types, and suggest that tumour cell compositions should be considered when deconvolving the TME.
Collapse
Affiliation(s)
- Khoa A Tran
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
| | - Venkateswar Addala
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Rebecca L Johnston
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - David Lovell
- School of Computer Science, Queensland University of Technology, Brisbane, QLD, 4000, Australia
- QUT Centre for Data Science, Brisbane, QLD, 4000, Australia
| | - Andrew Bradley
- Faculty of Engineering, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Lambros T Koufariotis
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Scott Wood
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Sunny Z Wu
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Daniel Roden
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Ghamdan Al-Eryani
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Alexander Swarbrick
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, QLD, 4000, Australia
| | - John V Pearson
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Olga Kondrashova
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Nicola Waddell
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia.
| |
Collapse
|
137
|
Hoang DT, Dinstag G, Hermida LC, Ben-Zvi DS, Elis E, Caley K, Sammut SJ, Sinha S, Sinha N, Dampier CH, Stossel C, Patil T, Rajan A, Lassoued W, Strauss J, Bailey S, Allen C, Redman J, Beker T, Jiang P, Golan T, Wilkinson S, Sowalsky AG, Pine SR, Caldas C, Gulley JL, Aldape K, Aharonov R, Stone EA, Ruppin E. Prediction of cancer treatment response from histopathology images through imputed transcriptomics. RESEARCH SQUARE 2023:rs.3.rs-3193270. [PMID: 37790315 PMCID: PMC10543028 DOI: 10.21203/rs.3.rs-3193270/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin (H&E)-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an approach for predicting response to multiple targeted and immunotherapies from H&E-slides. In difference from existing approaches that aim to predict treatment response directly from the slides, ENLIGHT-DeepPT is an indirect two-step approach consisting of (1) DeepPT, a new deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response based on the DeepPT inferred expression values. DeepPT successfully predicts transcriptomics in all 16 TCGA cohorts tested and generalizes well to two independent datasets. Our key contribution is showing that ENLIGHT-DeepPT successfully predicts true responders in five independent patients' cohorts involving four different treatments spanning six cancer types with an overall odds ratio of 2.44, increasing the baseline response rate by 43.47% among predicted responders, without the need for any treatment data for training. Furthermore, its prediction accuracy on these datasets is comparable to a supervised approach predicting the response directly from the images, which needs to be trained and tested on the same cohort. ENLIGHT-DeepPT future application could provide clinicians with rapid treatment recommendations to an array of different therapies and importantly, may contribute to advancing precision oncology in developing countries.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | | | - Leandro C. Hermida
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Katherine Caley
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neelam Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher H. Dampier
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Tejas Patil
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Arun Rajan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shania Bailey
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint Allen
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam G. Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sharon R. Pine
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - James L. Gulley
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric A. Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
138
|
Messiou C, Lee R, Salto-Tellez M. Multimodal analysis and the oncology patient: Creating a hospital system for integrated diagnostics and discovery. Comput Struct Biotechnol J 2023; 21:4536-4539. [PMID: 37767106 PMCID: PMC10520501 DOI: 10.1016/j.csbj.2023.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
We propose that an information technology and computational framework that would unify access to hospital digital information silos, and enable integration of this information using machine learning methods, would bring a new paradigm to patient management and research. This is the core principle of Integrated Diagnostics (ID): the amalgamation of multiple analytical modalities, with evolved information technology, applied to a defined patient cohort, and resulting in a synergistic effect in the clinical value of the individual diagnostic tools. This has the potential to transform the practice of personalized oncology at a time at which it is very much needed. In this article we present different models from the literature that contribute to the vision of ID and we provide published exemplars of ID tools. We briefly describe ongoing efforts within a universal healthcare system to create national clinical datasets. Following this, we argue the case to create "hospital units" to leverage this multi-modal analysis, data integration and holistic clinical decision-making. Finally, we describe the joint model created in our institutions.
Collapse
Affiliation(s)
- Christina Messiou
- Imaging and Data Science Theme lead and Director of the Imaging AI hub at The Royal Marsden and Institute of Cancer Research, National Institute for Health Research Biomedical Research Centre, Sutton SM2 5PT, UK
| | - Richard Lee
- Consultant Respiratory Physician & Champion for Early Diagnosis Early Diagnosis and Detection Centre, NIHR Biomedical Research Centre at the Royal Marsden and ICR, National Heart and Lung Institute, Imperial College London, UK
| | - Manuel Salto-Tellez
- The Integrated Pathology Unit, the Institute of Cancer Research & The Royal Marsden Hospital, Sutton SM2 5PT, UK
- Precision Medicine Centre, Queen’s University Belfast, UK
| |
Collapse
|
139
|
Kang W, Qiu X, Luo Y, Luo J, Liu Y, Xi J, Li X, Yang Z. Application of radiomics-based multiomics combinations in the tumor microenvironment and cancer prognosis. J Transl Med 2023; 21:598. [PMID: 37674169 PMCID: PMC10481579 DOI: 10.1186/s12967-023-04437-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/12/2023] [Indexed: 09/08/2023] Open
Abstract
The advent of immunotherapy, a groundbreaking advancement in cancer treatment, has given rise to the prominence of the tumor microenvironment (TME) as a critical area of research. The clinical implications of an improved understanding of the TME are significant and far-reaching. Radiomics has been increasingly utilized in the comprehensive assessment of the TME and cancer prognosis. Similarly, the advancement of pathomics, which is based on pathological images, can offer additional insights into the panoramic view and microscopic information of tumors. The combination of pathomics and radiomics has revolutionized the concept of a "digital biopsy". As genomics and transcriptomics continue to evolve, integrating radiomics with genomic and transcriptomic datasets can offer further insights into tumor and microenvironment heterogeneity and establish correlations with biological significance. Therefore, the synergistic analysis of digital image features (radiomics, pathomics) and genetic phenotypes (genomics) can comprehensively decode and characterize the heterogeneity of the TME as well as predict cancer prognosis. This review presents a comprehensive summary of the research on important radiomics biomarkers for predicting the TME, emphasizing the interplay between radiomics, genomics, transcriptomics, and pathomics, as well as the application of multiomics in decoding the TME and predicting cancer prognosis. Finally, we discuss the challenges and opportunities in multiomics research. In conclusion, this review highlights the crucial role of radiomics and multiomics associations in the assessment of the TME and cancer prognosis. The combined analysis of radiomics, pathomics, genomics, and transcriptomics is a promising research direction with substantial research significance and value for comprehensive TME evaluation and cancer prognosis assessment.
Collapse
Affiliation(s)
- Wendi Kang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiang Qiu
- Obstetrics and Gynecology Hospital of, Fudan University, Shanghai, 200011, China
| | - Yingen Luo
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Jianwei Luo
- Department of Diagnostic Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, 283 Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, China
| | - Yang Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junqing Xi
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Xiao Li
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China
| | - Zhengqiang Yang
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli 17# Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
140
|
Nuñez NA, Coombes BJ, Melhuish Beaupre L, Romo-Nava F, Gardea-Resendez M, Ozerdem A, Veldic M, Singh B, Sanchez Ruiz JA, Cuellar-Barboza A, Leung JG, Prieto ML, McElroy SL, Biernacka JM, Frye MA. Antidepressant-Associated Treatment Emergent Mania: A Meta-Analysis to Guide Risk Modeling Pharmacogenomic Targets of Potential Clinical Value. J Clin Psychopharmacol 2023; 43:428-433. [PMID: 37683232 PMCID: PMC10476595 DOI: 10.1097/jcp.0000000000001747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/09/2023] [Indexed: 08/17/2023]
Abstract
BACKGROUND The purpose of this study was to review the association between the SLC6A4 5-HTTLPR polymorphism and antidepressant (AD)-associated treatment emergent mania (TEM) in bipolar disorder alongside starting a discussion on the merits of developing risk stratification models to guide when not to provide AD treatment for bipolar depression. METHODS Studies that examined the association between clinical and genetic risk factors, specifically monoaminergic transporter genetic variation, and TEM were identified. A meta-analysis was performed using the odds ratio to estimate the effect size under the Der-Simonian and Laird model. RESULTS Seven studies, referencing the SLC6A4 5-HTTLPR polymorphism and TEM (total N = 1578; TEM+ =594, TEM- = 984), of 142 identified articles were included. The time duration between the start of the AD to emergence of TEM ranged from 4 to 12 weeks. There was a nominally significant association between the s allele of the 5-HTTLPR polymorphism and TEM (odds ratio, 1.434; 95% confidence interval, 1.001-2.055; P = 0.0493; I2 = 52%). No studies have investigated norepinephrine or dopamine transporters. CONCLUSION Although the serotonin transporter genetic variation is commercially available in pharmacogenomic decision support tools, greater efforts, more broadly, should focus on complete genome-wide approaches to determine genetic variants that may contribute to TEM. Moreover, these data are exemplary to the merits of developing risk stratification models, which include both clinical and biological risk factors, to guide when not to use ADs in bipolar disorder. Future studies will need to validate new risk models that best inform the development of personalized medicine best practices treating bipolar depression.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marin Veldic
- From the Departments of Psychiatry and Psychology
| | | | | | | | | | - Miguel L. Prieto
- Department of Psychiatry, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Susan L. McElroy
- Department of Psychiatry, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Joanna M. Biernacka
- From the Departments of Psychiatry and Psychology
- Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Mark A. Frye
- From the Departments of Psychiatry and Psychology
| |
Collapse
|
141
|
Liu ZP. Editorial: Insights in integrative bioinformatics-2021. FRONTIERS IN BIOINFORMATICS 2023; 3:1267370. [PMID: 37671243 PMCID: PMC10476083 DOI: 10.3389/fbinf.2023.1267370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 08/14/2023] [Indexed: 09/07/2023] Open
Affiliation(s)
- Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong, China
| |
Collapse
|
142
|
Pang W, Zhang B, Jin L, Yao Y, Han Q, Zheng X. Serological Biomarker-Based Machine Learning Models for Predicting the Relapse of Ulcerative Colitis. J Inflamm Res 2023; 16:3531-3545. [PMID: 37636275 PMCID: PMC10455884 DOI: 10.2147/jir.s423086] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/11/2023] [Indexed: 08/29/2023] Open
Abstract
Purpose To explore whether machine learning models using serological markers can predict the relapse of Ulcerative colitis (UC). Patients and Methods This clinical cohort study included 292 UC patients, and serological markers were obtained when patients were discharged from the hospital. Subsequently, four machine learning models including the random forest (RF) model, the logistic regression model, the decision tree, and the neural network were compared to predict the relapse of UC. A nomogram was constructed, and the performance of these models was evaluated by accuracy, sensitivity, specificity, and the area under the receiver operating characteristic curve (AUC). Results Based on the patients' characteristics and serological markers, we selected the relevant variables associated with relapse and developed a LR model. The novel model including gender, white blood cell count, percentage of leukomonocyte, percentage of monocyte, absolute value of neutrophilic granulocyte, and erythrocyte sedimentation rate was established for predicting the relapse. In addition, the average AUC of the four machine learning models was 0.828, of which the RF model was the best. The AUC of the test group was 0.889, the accuracy was 76.4%, the sensitivity was 78.5%, and the specificity was 76.4%. There were 45 variables in the RF models, and the relative weight coefficients of these variables were determined. Age has the greatest impact on classification results, followed by hemoglobin concentration, white blood cell count, and platelet distribution width. Conclusion Machine learning models based on serological markers had high accuracy in predicting the relapse of UC. The model can be used to noninvasively predict patient outcomes and can be an effective tool for determining personalized treatment plans.
Collapse
Affiliation(s)
- Wenwen Pang
- Department of Clinical Laboratory, Tianjin Union Medical Center, Nankai University, Tianjin, People’s Republic of China
| | - Bowei Zhang
- School of Medicine, Nankai University, Tianjin, People’s Republic of China
| | - Leixin Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Yao Yao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Qiurong Han
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Xiaoli Zheng
- Department of Clinical Laboratory, Tianjin Union Medical Center, Nankai University, Tianjin, People’s Republic of China
| |
Collapse
|
143
|
Zhu Z, Jiang L, Ding X. Advancing Breast Cancer Heterogeneity Analysis: Insights from Genomics, Transcriptomics and Proteomics at Bulk and Single-Cell Levels. Cancers (Basel) 2023; 15:4164. [PMID: 37627192 PMCID: PMC10452610 DOI: 10.3390/cancers15164164] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/23/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer continues to pose a significant healthcare challenge worldwide for its inherent molecular heterogeneity. This review offers an in-depth assessment of the molecular profiling undertaken to understand this heterogeneity, focusing on multi-omics strategies applied both in traditional bulk and single-cell levels. Genomic investigations have profoundly informed our comprehension of breast cancer, enabling its categorization into six intrinsic molecular subtypes. Beyond genomics, transcriptomics has rendered deeper insights into the gene expression landscape of breast cancer cells. It has also facilitated the formulation of more precise predictive and prognostic models, thereby enriching the field of personalized medicine in breast cancer. The comparison between traditional and single-cell transcriptomics has identified unique gene expression patterns and facilitated the understanding of cell-to-cell variability. Proteomics provides further insights into breast cancer subtypes by illuminating intricate protein expression patterns and their post-translational modifications. The adoption of single-cell proteomics has been instrumental in this regard, revealing the complex dynamics of protein regulation and interaction. Despite these advancements, this review underscores the need for a holistic integration of multiple 'omics' strategies to fully decipher breast cancer heterogeneity. Such integration not only ensures a comprehensive understanding of breast cancer's molecular complexities, but also promotes the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Zijian Zhu
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, China;
| | - Lai Jiang
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200025, China;
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200030, China;
- Department of Anesthesiology and Surgical Intensive Care Unit, Xinhua Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200025, China;
| |
Collapse
|
144
|
Martino F, Lupi M, Giraudo E, Lanzetti L. Breast cancers as ecosystems: a metabolic perspective. Cell Mol Life Sci 2023; 80:244. [PMID: 37561190 PMCID: PMC10415483 DOI: 10.1007/s00018-023-04902-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023]
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer and one of the major causes of cancer death. Despite enormous progress in its management, both from the therapeutic and early diagnosis viewpoints, still around 700,000 patients succumb to the disease each year, worldwide. Late recurrency is the major problem in BC, with many patients developing distant metastases several years after the successful eradication of the primary tumor. This is linked to the phenomenon of metastatic dormancy, a still mysterious trait of the natural history of BC, and of several other types of cancer, by which metastatic cells remain dormant for long periods of time before becoming reactivated to initiate the clinical metastatic disease. In recent years, it has become clear that cancers are best understood if studied as ecosystems in which the impact of non-cancer-cell-autonomous events-dependent on complex interaction between the cancer and its environment, both local and systemic-plays a paramount role, probably as significant as the cell-autonomous alterations occurring in the cancer cell. In adopting this perspective, a metabolic vision of the cancer ecosystem is bound to improve our understanding of the natural history of cancer, across space and time. In BC, many metabolic pathways are coopted into the cancer ecosystem, to serve the anabolic and energy demands of the cancer. Their study is shedding new light on the most critical aspect of BC management, of metastatic dissemination, and that of the related phenomenon of dormancy and fostering the application of the knowledge to the development of metabolic therapies.
Collapse
Affiliation(s)
- Flavia Martino
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Mariadomenica Lupi
- Department of Oncology, University of Torino Medical School, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Enrico Giraudo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Department of Science and Drug Technology, University of Torino, Turin, Italy
| | - Letizia Lanzetti
- Department of Oncology, University of Torino Medical School, Turin, Italy.
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
| |
Collapse
|
145
|
Marcinak CT, Parker WF, Parikh AA, Datta J, Maithel SK, Kooby DA, Burkard ME, Kim HJ, LeCompte MT, Afshar M, Churpek MM, Zafar SN. Accuracy of models to prognosticate survival after surgery for pancreatic cancer in the era of neoadjuvant therapy. J Surg Oncol 2023; 128:280-288. [PMID: 37073788 PMCID: PMC10330210 DOI: 10.1002/jso.27287] [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: 02/02/2023] [Revised: 03/10/2023] [Accepted: 04/09/2023] [Indexed: 04/20/2023]
Abstract
BACKGROUND Outcomes for pancreatic adenocarcinoma (PDAC) remain difficult to prognosticate. Multiple models attempt to predict survival following the resection of PDAC, but their utility in the neoadjuvant population is unknown. We aimed to assess their accuracy among patients that received neoadjuvant chemotherapy (NAC). METHODS We performed a multi-institutional retrospective analysis of patients who received NAC and underwent resection of PDAC. Two prognostic systems were evaluated: the Memorial Sloan Kettering Cancer Center Pancreatic Adenocarcinoma Nomogram (MSKCCPAN) and the American Joint Committee on Cancer (AJCC) staging system. Discrimination between predicted and actual disease-specific survival was assessed using the Uno C-statistic and Kaplan-Meier method. Calibration of the MSKCCPAN was assessed using the Brier score. RESULTS A total of 448 patients were included. There were 232 (51.8%) females, and the mean age was 64.1 years (±9.5). Most had AJCC Stage I or II disease (77.7%). For the MSKCCPAN, the Uno C-statistic at 12-, 24-, and 36-month time points was 0.62, 0.63, and 0.62, respectively. The AJCC system demonstrated similarly mediocre discrimination. The Brier score for the MSKCCPAN was 0.15 at 12 months, 0.26 at 24 months, and 0.30 at 36 months, demonstrating modest calibration. CONCLUSIONS Current survival prediction models and staging systems for patients with PDAC undergoing resection after NAC have limited accuracy.
Collapse
Affiliation(s)
- Clayton T. Marcinak
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
| | - William F. Parker
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Alexander A. Parikh
- Division of Surgical Oncology and Endocrine Surgery, UT Health San Antonio MD Anderson – Mays Cancer Center, San Antonio, TX, USA
| | - Jashodeep Datta
- Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shishir K. Maithel
- Division of Surgical Oncology, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - David A. Kooby
- Division of Surgical Oncology, Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Mark E. Burkard
- Division of Hematology, Oncology, and Palliative Care, Department of Medicine, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
| | - Hong Jin Kim
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Michael T. LeCompte
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Majid Afshar
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
| | - Matthew M. Churpek
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
| | - Syed Nabeel Zafar
- Division of Surgical Oncology, Department of Surgery, School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
146
|
Wang H, Fu T, Du Y, Gao W, Huang K, Liu Z, Chandak P, Liu S, Van Katwyk P, Deac A, Anandkumar A, Bergen K, Gomes CP, Ho S, Kohli P, Lasenby J, Leskovec J, Liu TY, Manrai A, Marks D, Ramsundar B, Song L, Sun J, Tang J, Veličković P, Welling M, Zhang L, Coley CW, Bengio Y, Zitnik M. Scientific discovery in the age of artificial intelligence. Nature 2023; 620:47-60. [PMID: 37532811 DOI: 10.1038/s41586-023-06221-2] [Citation(s) in RCA: 179] [Impact Index Per Article: 89.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2023] [Indexed: 08/04/2023]
Abstract
Artificial intelligence (AI) is being increasingly integrated into scientific discovery to augment and accelerate research, helping scientists to generate hypotheses, design experiments, collect and interpret large datasets, and gain insights that might not have been possible using traditional scientific methods alone. Here we examine breakthroughs over the past decade that include self-supervised learning, which allows models to be trained on vast amounts of unlabelled data, and geometric deep learning, which leverages knowledge about the structure of scientific data to enhance model accuracy and efficiency. Generative AI methods can create designs, such as small-molecule drugs and proteins, by analysing diverse data modalities, including images and sequences. We discuss how these methods can help scientists throughout the scientific process and the central issues that remain despite such advances. Both developers and users of AI toolsneed a better understanding of when such approaches need improvement, and challenges posed by poor data quality and stewardship remain. These issues cut across scientific disciplines and require developing foundational algorithmic approaches that can contribute to scientific understanding or acquire it autonomously, making them critical areas of focus for AI innovation.
Collapse
Affiliation(s)
- Hanchen Wang
- Department of Engineering, University of Cambridge, Cambridge, UK
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
- Department of Research and Early Development, Genentech Inc, South San Francisco, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Tianfan Fu
- Department of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yuanqi Du
- Department of Computer Science, Cornell University, Ithaca, NY, USA
| | - Wenhao Gao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kexin Huang
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ziming Liu
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Payal Chandak
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA, USA
| | - Shengchao Liu
- Mila - Quebec AI Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Peter Van Katwyk
- Department of Earth, Environmental and Planetary Sciences, Brown University, Providence, RI, USA
- Data Science Institute, Brown University, Providence, RI, USA
| | - Andreea Deac
- Mila - Quebec AI Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Anima Anandkumar
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
- NVIDIA, Santa Clara, CA, USA
| | - Karianne Bergen
- Department of Earth, Environmental and Planetary Sciences, Brown University, Providence, RI, USA
- Data Science Institute, Brown University, Providence, RI, USA
| | - Carla P Gomes
- Department of Computer Science, Cornell University, Ithaca, NY, USA
| | - Shirley Ho
- Center for Computational Astrophysics, Flatiron Institute, New York, NY, USA
- Department of Astrophysical Sciences, Princeton University, Princeton, NJ, USA
- Department of Physics, Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Physics and Center for Data Science, New York University, New York, NY, USA
| | | | - Joan Lasenby
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | | | - Arjun Manrai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Debora Marks
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Le Song
- BioMap, Beijing, China
- Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Jimeng Sun
- University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jian Tang
- Mila - Quebec AI Institute, Montreal, Quebec, Canada
- HEC Montréal, Montreal, Quebec, Canada
- CIFAR AI Chair, Toronto, Ontario, Canada
| | - Petar Veličković
- Google DeepMind, London, UK
- Department of Computer Science and Technology, University of Cambridge, Cambridge, UK
| | - Max Welling
- University of Amsterdam, Amsterdam, Netherlands
- Microsoft Research Amsterdam, Amsterdam, Netherlands
| | - Linfeng Zhang
- DP Technology, Beijing, China
- AI for Science Institute, Beijing, China
| | - Connor W Coley
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yoshua Bengio
- Mila - Quebec AI Institute, Montreal, Quebec, Canada
- Université de Montréal, Montreal, Quebec, Canada
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Data Science Initiative, Cambridge, MA, USA.
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
147
|
Wu J, Liang C, Wang X, Huang Y, Liu W, Wang R, Cao J, Su X, Yin T, Wang X, Zhang Z, Shen L, Li D, Zou W, Wu J, Qiu L, Di W, Cao Y, Ji D, Qian K. Efficient Metabolic Fingerprinting of Follicular Fluid Encodes Ovarian Reserve and Fertility. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302023. [PMID: 37311196 PMCID: PMC10427401 DOI: 10.1002/advs.202302023] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/30/2023] [Indexed: 06/15/2023]
Abstract
Ovarian reserve (OR) and fertility are critical in women's healthcare. Clinical methods for encoding OR and fertility rely on the combination of tests, which cannot serve as a multi-functional platform with limited information from specific biofluids. Herein, metabolic fingerprinting of follicular fluid (MFFF) from follicles is performed, using particle-assisted laser desorption/ionization mass spectrometry (PALDI-MS) to encode OR and fertility. PALDI-MS allows efficient MFFF, showing fast speed (≈30 s), high sensitivity (≈60 fmol), and desirable reproducibility (coefficients of variation <15%). Further, machine learning of MFFF is applied to diagnose diminished OR (area under the curve of 0.929) and identify high-quality oocytes/embryos (p < 0.05) by a single PALDI-MS test. Meanwhile, metabolic biomarkers from MFFF are identified, which also determine oocyte/embryo quality (p < 0.05) from the sampling follicles toward fertility prediction in clinics. This approach offers a powerful platform in women's healthcare, not limited to OR and fertility.
Collapse
|
148
|
Dalal S, Onyema EM, Kumar P, Maryann DC, Roselyn AO, Obichili MI. A hybrid machine learning model for timely prediction of breast cancer. INTERNATIONAL JOURNAL OF MODELING, SIMULATION, AND SCIENTIFIC COMPUTING 2023; 14. [DOI: 10.1142/s1793962323410234] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Breast cancer is one of the leading causes of untimely deaths among women in various countries across the world. This can be attributed to many factors including late detection which often increase its severity. Thus, detecting the disease early would help mitigate its mortality rate and other risks associated with it. This study developed a hybrid machine learning model for timely prediction of breast cancer to help combat the disease. The dataset from Kaggle was adopted to predict the breast tumor growth and sizes using random tree classification, logistic regression, XBoost tree and multilayer perceptron on the dataset. The implementation of these machine learning algorithms and visualization of the results was done using Python. The results achieved a high accuracy (99.65%) on training and testing datasets which is far better than traditional means. The predictive model has good potential to enhance early detection and diagnosis of breast cancer and improvement of treatment outcome. It could also assist patients to timely deal with their condition or life patterns to support their recovery or survival.
Collapse
Affiliation(s)
- Surjeet Dalal
- College of Computing Science and IT, Teerthanker Mahaveer University, Moradabad, UP, India
| | - Edeh Michael Onyema
- Department of Mathematics and Computer Science, Coal City University, Enugu, Nigeria
- Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Pawan Kumar
- College of Computing Science and IT, Teerthanker Mahaveer University, Moradabad, UP, India
| | | | | | - Mercy Ifeyinwa Obichili
- Department of Mass Communication, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, Ebonyi State, Nigeria
| |
Collapse
|
149
|
Hill HA, Jain P, Ok CY, Sasaki K, Chen H, Wang ML, Chen K. Integrative Prognostic Machine Learning Models in Mantle Cell Lymphoma. CANCER RESEARCH COMMUNICATIONS 2023; 3:1435-1446. [PMID: 37538987 PMCID: PMC10395375 DOI: 10.1158/2767-9764.crc-23-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/17/2023] [Accepted: 06/27/2023] [Indexed: 08/05/2023]
Abstract
Patients with mantle cell lymphoma (MCL), an incurable B-cell malignancy, benefit from accurate pretreatment disease stratification. We curated an extensive database of 862 patients diagnosed between 2014 and 2022. A machine learning (ML) gradient-boosted model incorporated baseline features from clinicopathologic, cytogenetic, and genomic data with high predictive power discriminating between patients with indolent or responsive MCL and those with aggressive disease (AUC ROC = 0.83). In addition, we utilized the gradient-boosted framework as a robust feature selection method for multivariate logistic and survival modeling. The best ML models incorporated features from clinical and genomic data types highlighting the need for correlative molecular studies in precision oncology. As proof of concept, we launched our most accurate and practical models using an application interface, which has potential for clinical implementation. We designated the 20-feature ML model-based index the "integrative MIPI" or iMIPI and a similar 10-feature ML index the "integrative simplified MIPI" or iMIPI-s. The top 10 baseline prognostic features represented in the iMIPI-s are: lactase dehydrogenase (LDH), Ki-67%, platelet count, bone marrow involvement percentage, hemoglobin levels, the total number of observed somatic mutations, TP53 mutational status, Eastern Cooperative Oncology Group performance level, beta-2 microglobulin, and morphology. Our findings emphasize that prognostic applications and indices should include molecular features, especially TP53 mutational status. This work demonstrates the clinical utility of complex ML models and provides further evidence for existing prognostic markers in MCL. Significance Our model is the first to integrate a dynamic algorithm with multiple clinical and molecular features, allowing for accurate predictions of MCL disease outcomes in a large patient cohort.
Collapse
Affiliation(s)
- Holly A. Hill
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas
| | - Preetesh Jain
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chi Young Ok
- Department of Hematopathology, Division of Pathology-Lab Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Han Chen
- Department of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston School of Public Health, Houston, Texas
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Michael L. Wang
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, Division of Quantitative Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
150
|
Scott EC, Baines AC, Gong Y, Moore R, Pamuk GE, Saber H, Subedee A, Thompson MD, Xiao W, Pazdur R, Rao VA, Schneider J, Beaver JA. Trends in the approval of cancer therapies by the FDA in the twenty-first century. Nat Rev Drug Discov 2023; 22:625-640. [PMID: 37344568 DOI: 10.1038/s41573-023-00723-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/23/2023]
Abstract
The cancer treatment landscape has changed dramatically since the turn of the century, resulting in substantial improvements in outcomes for patients. This Review summarizes trends in the approval of oncology therapeutic products by the United States Food and Drug Administration (FDA) from January 2000 to October 2022, based on a categorization of these products by their mechanism of action and primary target. Notably, the rate of oncology indication approvals has increased in this time, driven by approvals for targeted therapies, as has the rate of introduction of new therapeutic approaches. Kinase inhibitors are the dominant product class by number of approved products and indications, yet immune checkpoint inhibitors have the second most approvals despite not entering the market until 2011. Other trends include a slight increase in the share of approvals for biomarker-defined populations and the emergence of tumour-site-agnostic approvals. Finally, we consider the implications of the trends for the future of oncology therapeutic product development, including the impact of novel therapeutic approaches and technologies.
Collapse
Affiliation(s)
- Emma C Scott
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA.
| | - Andrea C Baines
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Yutao Gong
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Rodney Moore
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Gulsum E Pamuk
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Haleh Saber
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ashim Subedee
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
- National Cancer Institute, Rockville, MD, USA
| | - Matthew D Thompson
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Wenming Xiao
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Richard Pazdur
- Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - V Ashutosh Rao
- Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Julie Schneider
- Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Julia A Beaver
- Office of Oncologic Diseases, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
- Oncology Center of Excellence, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|