1
|
Captier N, Lerousseau M, Orlhac F, Hovhannisyan-Baghdasarian N, Luporsi M, Woff E, Lagha S, Salamoun Feghali P, Lonjou C, Beaulaton C, Zinovyev A, Salmon H, Walter T, Buvat I, Girard N, Barillot E. Integration of clinical, pathological, radiological, and transcriptomic data improves prediction for first-line immunotherapy outcome in metastatic non-small cell lung cancer. Nat Commun 2025; 16:614. [PMID: 39800784 PMCID: PMC11725576 DOI: 10.1038/s41467-025-55847-5] [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: 02/09/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025] Open
Abstract
Immunotherapy is improving the survival of patients with metastatic non-small cell lung cancer (NSCLC), yet reliable biomarkers are needed to identify responders prospectively and optimize patient care. In this study, we explore the benefits of multimodal approaches to predict immunotherapy outcome using multiple machine learning algorithms and integration strategies. We analyze baseline multimodal data from a cohort of 317 metastatic NSCLC patients treated with first-line immunotherapy, including positron emission tomography images, digitized pathological slides, bulk transcriptomic profiles, and clinical information. Testing multiple integration strategies, most of them yield multimodal models surpassing both the best unimodal models and established univariate biomarkers, such as PD-L1 expression. Additionally, several multimodal combinations demonstrate improved patient risk stratification compared to models built with routine clinical features only. Our study thus provides evidence of the superiority of multimodal over unimodal approaches, advocating for the collection of large multimodal NSCLC datasets to develop and validate robust and powerful immunotherapy biomarkers.
Collapse
Affiliation(s)
- Nicolas Captier
- Laboratoire d'Imagerie Translationnelle en Oncologie, Institut Curie, Inserm U1288, PSL Research University, Orsay, France.
- Bioinformatics and computational systems biology of cancer, Institut Curie, Inserm U900, PSL Research University, Paris, France.
| | - Marvin Lerousseau
- Bioinformatics and computational systems biology of cancer, Institut Curie, Inserm U900, PSL Research University, Paris, France
- CBIO-center for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| | - Fanny Orlhac
- Laboratoire d'Imagerie Translationnelle en Oncologie, Institut Curie, Inserm U1288, PSL Research University, Orsay, France
| | | | - Marie Luporsi
- Laboratoire d'Imagerie Translationnelle en Oncologie, Institut Curie, Inserm U1288, PSL Research University, Orsay, France
- Department of medical imaging, Institut Curie, Paris, France
| | - Erwin Woff
- Laboratoire d'Imagerie Translationnelle en Oncologie, Institut Curie, Inserm U1288, PSL Research University, Orsay, France
- Department of Nuclear Medicine/PET-scan, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sarah Lagha
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | | | - Christine Lonjou
- Bioinformatics and computational systems biology of cancer, Institut Curie, Inserm U900, PSL Research University, Paris, France
| | | | | | - Hélène Salmon
- Immunity and cancer, Institut Curie, Inserm U932, PSL Research University, Paris, France
| | - Thomas Walter
- Bioinformatics and computational systems biology of cancer, Institut Curie, Inserm U900, PSL Research University, Paris, France
- CBIO-center for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| | - Irène Buvat
- Laboratoire d'Imagerie Translationnelle en Oncologie, Institut Curie, Inserm U1288, PSL Research University, Orsay, France
| | - Nicolas Girard
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | - Emmanuel Barillot
- Bioinformatics and computational systems biology of cancer, Institut Curie, Inserm U900, PSL Research University, Paris, France.
| |
Collapse
|
2
|
Ai X, Jia B, He Z, Zhang J, Zhuo M, Zhao J, Wang Z, Zhang J, Fan Z, Zhang X, Li C, Jin F, Li Z, Ma X, Tang H, Yan X, Li W, Xiong Y, Yin H, Chen R, Lu S. Noninvasive early identification of durable clinical benefit from immune checkpoint inhibition: a prospective multicenter study (NCT04566432). Signal Transduct Target Ther 2024; 9:350. [PMID: 39676097 DOI: 10.1038/s41392-024-02060-3] [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: 05/01/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment landscape for patients with non-small cell lung cancer (NSCLC). In spite of durable responses in some patients, many patients develop early disease progression during the ICI treatment. Thus, early identification of patients with no durable benefit would facilitate the clinical decision for these patients. In this prospective, multicenter study, 101 non-EGFR/ALK patients who received ICI treatment were enrolled after screening 328 stage III-IV NSCLC patients. At the date of cutoff, 83 patients were eligible for ICI efficacy evaluation, with 56 patients having progress-free survival (PFS) over 6 months, which was defined as durable clinical benefit (DCB). A multimodal model was established by integrating normalized bTMB, early dynamic of ctDNA and the first RECIST response. This model could robustly predict DCB with area under the curve (AUC) of 0.878, sensitivity of 79.2% at 86.4% specificity (accuracy = 80.0%). This model was further validated in the independent cohort of the DIREct-On study with AUC of 0.887, sensitivity of 94.7% at 85.3% specificity (accuracy = 90.3%). Patients with higher predict scores had substantially longer PFS than those with lower scores (training cohort: median PFS 13.6 vs 4.2 months, P < 0.001, HR = 0.24; validation cohort: median PFS 11.0 vs 2.2 months, P < 0.001, HR = 0.17). Taken together, these results demonstrate that integrating early changes of ctDNA, normalized bTMB, and the first RECIST response can provide accurate, noninvasive, and early prediction of durable benefits for NSCLC patients treated with ICIs. Further prospective studies are warranted to validate these findings and guide clinical decision-making for optimal immunotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Xinghao Ai
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bo Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education / Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhiyi He
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Junping Zhang
- Department of Thoracic Oncology, Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education / Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education / Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhe Wang
- Department of Thoracic Surgery, National Cancer Center / National Clinical Research Center for Cancer / Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jiexia Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zaiwen Fan
- Department of Pulmonary and Critical Care Medicine, Air Force Medical Center, PLA, Beijing, China
| | - Xiaotong Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chong Li
- Department of Pulmonary and Critical Care Medicine, Third Affiliated Hospital of Soochow University, First People's Hospital of Changzhou, Changzhou, China
| | - Feng Jin
- Oncology Department, Qianjiang National Hospital, Chongqing, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hao Tang
- Department of Respiratory and Critical Care Medicine, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiang Yan
- Department of Thoracic Surgery, Peking University People's hospital, Beijing, China
| | - Wei Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | | | - Huan Yin
- GenePlus-Beijing, Beijing, China
| | | | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Field M, Vinod S, Delaney GP, Aherne N, Bailey M, Carolan M, Dekker A, Greenham S, Hau E, Lehmann J, Ludbrook J, Miller A, Rezo A, Selvaraj J, Sykes J, Thwaites D, Holloway L. Federated Learning Survival Model and Potential Radiotherapy Decision Support Impact Assessment for Non-small Cell Lung Cancer Using Real-World Data. Clin Oncol (R Coll Radiol) 2024; 36:e197-e208. [PMID: 38631978 DOI: 10.1016/j.clon.2024.03.008] [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: 03/06/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024]
Abstract
AIMS The objective of this study was to develop a two-year overall survival model for inoperable stage I-III non-small cell lung cancer (NSCLC) patients using routine radiation oncology data over a federated (distributed) learning network and evaluate the potential of decision support for curative versus palliative radiotherapy. METHODS A federated infrastructure of data extraction, de-identification, standardisation, image analysis, and modelling was installed for seven clinics to obtain clinical and imaging features and survival information for patients treated in 2011-2019. A logistic regression model was trained for the 2011-2016 curative patient cohort and validated for the 2017-2019 cohort. Features were selected with univariate and model-based analysis and optimised using bootstrapping. System performance was assessed by the receiver operating characteristic (ROC) and corresponding area under curve (AUC), C-index, calibration metrics and Kaplan-Meier survival curves, with risk groups defined by model probability quartiles. Decision support was evaluated using a case-control analysis using propensity matching between treatment groups. RESULTS 1655 patient datasets were included. The overall model AUC was 0.68. Fifty-eight percent of patients treated with palliative radiotherapy had a low-to-moderate risk prediction according to the model, with survival times not significantly different (p = 0.87 and 0.061) from patients treated with curative radiotherapy classified as high-risk by the model. When survival was simulated by risk group and model-indicated treatment, there was an estimated 11% increase in survival rate at two years (p < 0.01). CONCLUSION Federated learning over multiple institution data can be used to develop and validate decision support systems for lung cancer while quantifying the potential impact of their use in practice. This paves the way for personalised medicine, where decisions can be based more closely on individual patient details from routine care.
Collapse
Affiliation(s)
- M Field
- South Western Sydney Clinical Campus, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia; Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia; South Western Sydney Cancer Services, NSW Health, Sydney, New South Wales, Australia.
| | - S Vinod
- South Western Sydney Clinical Campus, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia; Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia; South Western Sydney Cancer Services, NSW Health, Sydney, New South Wales, Australia
| | - G P Delaney
- South Western Sydney Clinical Campus, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia; Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia; South Western Sydney Cancer Services, NSW Health, Sydney, New South Wales, Australia
| | - N Aherne
- Mid North Coast Cancer Institute, Coffs Harbour, New South Wales, Australia; Rural Clinical School, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - M Bailey
- Illawarra Cancer Care Centre, Wollongong, New South Wales, Australia
| | - M Carolan
- Illawarra Cancer Care Centre, Wollongong, New South Wales, Australia
| | - A Dekker
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - S Greenham
- Mid North Coast Cancer Institute, Coffs Harbour, New South Wales, Australia
| | - E Hau
- Sydney West Radiation Oncology Network, Sydney, Australia; Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - J Lehmann
- School of Mathematical and Physical Sciences, University of Newcastle, Newcastle, New South Wales, Australia; Department of Radiation Oncology, Calvary Mater, Newcastle, New South Wales, Australia; Institute of Medical Physics, School of Physics, University of Sydney, Sydney, New South Wales, Australia
| | - J Ludbrook
- Department of Radiation Oncology, Calvary Mater, Newcastle, New South Wales, Australia
| | - A Miller
- Illawarra Cancer Care Centre, Wollongong, New South Wales, Australia
| | - A Rezo
- Canberra Health Services, Canberra, Australian Capital Territory, Australia
| | - J Selvaraj
- South Western Sydney Clinical Campus, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia; Canberra Health Services, Canberra, Australian Capital Territory, Australia
| | - J Sykes
- Sydney West Radiation Oncology Network, Sydney, Australia; Institute of Medical Physics, School of Physics, University of Sydney, Sydney, New South Wales, Australia
| | - D Thwaites
- Institute of Medical Physics, School of Physics, University of Sydney, Sydney, New South Wales, Australia; Radiotherapy Research Group, Leeds Institute for Medical Research, St James's Hospital and the University of Leeds, Leeds, UK
| | - L Holloway
- South Western Sydney Clinical Campus, School of Clinical Medicine, UNSW, Sydney, New South Wales, Australia; Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia; South Western Sydney Cancer Services, NSW Health, Sydney, New South Wales, Australia; Institute of Medical Physics, School of Physics, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
4
|
Li ZC, Wang J, Liu HB, Zheng YM, Huang JH, Cai JB, Zhang L, Liu X, Du L, Yang XT, Chai XQ, Jiang YH, Ren ZG, Zhou J, Fan J, Yu DC, Sun HC, Huang C, Liu F. Proteomic and metabolomic features in patients with HCC responding to lenvatinib and anti-PD1 therapy. Cell Rep 2024; 43:113877. [PMID: 38421869 DOI: 10.1016/j.celrep.2024.113877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 12/16/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
Combination therapy (lenvatinib/programmed death-1 inhibitor) is effective for treating unresectable hepatocellular carcinoma (uHCC). We reveal that responders have better overall and progression-free survival, as well as high tumor mutation burden and special somatic variants. We analyze the proteome and metabolome of 82 plasma samples from patients with hepatocellular carcinoma (HCC; n = 51) and normal controls (n = 15), revealing that individual differences outweigh treatment differences. Responders exhibit enhanced activity in the alternative/lectin complement pathway and higher levels of lysophosphatidylcholines (LysoPCs), predicting a favorable prognosis. Non-responders are enriched for immunoglobulins, predicting worse outcomes. Compared to normal controls, HCC plasma proteins show acute inflammatory response and platelet activation, while LysoPCs decrease. Combination therapy increases LysoPCs/phosphocholines in responders. Logistic regression/random forest models using metabolomic features achieve good performance in the prediction of responders. Proteomic analysis of cancer tissues unveils molecular features that are associated with side effects in responders receiving combination therapy. In conclusion, our analysis identifies plasma features associated with uHCC responders to combination therapy.
Collapse
Affiliation(s)
- Zhong-Chen Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jie Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Minhang Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical of Sciences, Fudan University, 131 DongAn Road, Shanghai 200032, China
| | - He-Bin Liu
- Shanghai Omicsolution Co., Ltd., 28 Yuanwen Road, Shanghai 201199, China
| | - Yi-Min Zheng
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jian-Hang Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China; Minhang Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical of Sciences, Fudan University, 131 DongAn Road, Shanghai 200032, China
| | - Jia-Bin Cai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Institutes of Biomedical of Sciences, Fudan University, 220 Handan Road, Shanghai 200433, China
| | - Xin Liu
- Department of Central Laboratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Zhijiang Road, Shanghai 200071, China
| | - Ling Du
- Minhang Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical of Sciences, Fudan University, 131 DongAn Road, Shanghai 200032, China
| | - Xue-Ting Yang
- Minhang Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical of Sciences, Fudan University, 131 DongAn Road, Shanghai 200032, China
| | - Xiao-Qiang Chai
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Ying-Hua Jiang
- Minhang Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical of Sciences, Fudan University, 131 DongAn Road, Shanghai 200032, China
| | - Zheng-Gang Ren
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China
| | - De-Cai Yu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China.
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Fudan University, Shanghai 200032, China.
| | - Feng Liu
- Minhang Hospital, Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical of Sciences, Fudan University, 131 DongAn Road, Shanghai 200032, China.
| |
Collapse
|
5
|
Prelaj A, Miskovic V, Zanitti M, Trovo F, Genova C, Viscardi G, Rebuzzi SE, Mazzeo L, Provenzano L, Kosta S, Favali M, Spagnoletti A, Castelo-Branco L, Dolezal J, Pearson AT, Lo Russo G, Proto C, Ganzinelli M, Giani C, Ambrosini E, Turajlic S, Au L, Koopman M, Delaloge S, Kather JN, de Braud F, Garassino MC, Pentheroudakis G, Spencer C, Pedrocchi ALG. Artificial intelligence for predictive biomarker discovery in immuno-oncology: a systematic review. Ann Oncol 2024; 35:29-65. [PMID: 37879443 DOI: 10.1016/j.annonc.2023.10.125] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/31/2023] [Accepted: 10/08/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND The widespread use of immune checkpoint inhibitors (ICIs) has revolutionised treatment of multiple cancer types. However, selecting patients who may benefit from ICI remains challenging. Artificial intelligence (AI) approaches allow exploitation of high-dimension oncological data in research and development of precision immuno-oncology. MATERIALS AND METHODS We conducted a systematic literature review of peer-reviewed original articles studying the ICI efficacy prediction in cancer patients across five data modalities: genomics (including genomics, transcriptomics, and epigenomics), radiomics, digital pathology (pathomics), and real-world and multimodality data. RESULTS A total of 90 studies were included in this systematic review, with 80% published in 2021-2022. Among them, 37 studies included genomic, 20 radiomic, 8 pathomic, 20 real-world, and 5 multimodal data. Standard machine learning (ML) methods were used in 72% of studies, deep learning (DL) methods in 22%, and both in 6%. The most frequently studied cancer type was non-small-cell lung cancer (36%), followed by melanoma (16%), while 25% included pan-cancer studies. No prospective study design incorporated AI-based methodologies from the outset; rather, all implemented AI as a post hoc analysis. Novel biomarkers for ICI in radiomics and pathomics were identified using AI approaches, and molecular biomarkers have expanded past genomics into transcriptomics and epigenomics. Finally, complex algorithms and new types of AI-based markers, such as meta-biomarkers, are emerging by integrating multimodal/multi-omics data. CONCLUSION AI-based methods have expanded the horizon for biomarker discovery, demonstrating the power of integrating multimodal data from existing datasets to discover new meta-biomarkers. While most of the included studies showed promise for AI-based prediction of benefit from immunotherapy, none provided high-level evidence for immediate practice change. A priori planned prospective trial designs are needed to cover all lifecycle steps of these software biomarkers, from development and validation to integration into clinical practice.
Collapse
Affiliation(s)
- A Prelaj
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan; Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy; ESMO Real World Data and Digital Health Working Group, ESMO, Lugano, Switzerland.
| | - V Miskovic
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan; Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy
| | - M Zanitti
- Department of Electronic Systems, Aalborg University Copenhagen, Denmark
| | - F Trovo
- Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy
| | - C Genova
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa; Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa
| | - G Viscardi
- Precision Medicine Department, Università degli Studi della Campania Luigi Vanvitelli, Naples
| | - S E Rebuzzi
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa; Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
| | - L Mazzeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan; Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy
| | - L Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - S Kosta
- Department of Electronic Systems, Aalborg University Copenhagen, Denmark
| | - M Favali
- Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy
| | - A Spagnoletti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - L Castelo-Branco
- ESMO European Society for Medical Oncology, Lugano, Switzerland; NOVA National School of Public Health, Lisboa, Portugal
| | - J Dolezal
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, USA
| | - A T Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, USA
| | - G Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - C Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - M Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - C Giani
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - E Ambrosini
- Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy
| | - S Turajlic
- Cancer Dynamics Laboratory, The Francis Crick Institute, London
| | - L Au
- Renal and Skin Unit, The Royal Marsden NHS Foundation Trust, London, UK; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne; Sir Peter MacCallum Department of Medical Oncology, The University of Melbourne, Melbourne, Australia
| | - M Koopman
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, The Netherlands; ESMO Real World Data and Digital Health Working Group, ESMO, Lugano, Switzerland
| | - S Delaloge
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; ESMO Real World Data and Digital Health Working Group, ESMO, Lugano, Switzerland
| | - J N Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - F de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan
| | - M C Garassino
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, USA
| | | | - C Spencer
- Cancer Dynamics Laboratory, The Francis Crick Institute, London.
| | - A L G Pedrocchi
- Nearlab, Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milano, Italy
| |
Collapse
|
6
|
Çalışkan M, Tazaki K. AI/ML advances in non-small cell lung cancer biomarker discovery. Front Oncol 2023; 13:1260374. [PMID: 38148837 PMCID: PMC10750392 DOI: 10.3389/fonc.2023.1260374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/16/2023] [Indexed: 12/28/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths among both men and women, representing approximately 25% of cancer fatalities each year. The treatment landscape for non-small cell lung cancer (NSCLC) is rapidly evolving due to the progress made in biomarker-driven targeted therapies. While advancements in targeted treatments have improved survival rates for NSCLC patients with actionable biomarkers, long-term survival remains low, with an overall 5-year relative survival rate below 20%. Artificial intelligence/machine learning (AI/ML) algorithms have shown promise in biomarker discovery, yet NSCLC-specific studies capturing the clinical challenges targeted and emerging patterns identified using AI/ML approaches are lacking. Here, we employed a text-mining approach and identified 215 studies that reported potential biomarkers of NSCLC using AI/ML algorithms. We catalogued these studies with respect to BEST (Biomarkers, EndpointS, and other Tools) biomarker sub-types and summarized emerging patterns and trends in AI/ML-driven NSCLC biomarker discovery. We anticipate that our comprehensive review will contribute to the current understanding of AI/ML advances in NSCLC biomarker research and provide an important catalogue that may facilitate clinical adoption of AI/ML-derived biomarkers.
Collapse
Affiliation(s)
- Minal Çalışkan
- Translational Science Department, Precision Medicine Function, Daiichi Sankyo, Inc., Basking Ridge, NJ, United States
| | - Koichi Tazaki
- Translational Science Department I, Precision Medicine Function, Daiichi Sankyo, Tokyo, Japan
| |
Collapse
|
7
|
Hozumi H, Shimizu H. Bayesian network enables interpretable and state-of-the-art prediction of immunotherapy responses in cancer patients. PNAS NEXUS 2023; 2:pgad133. [PMID: 37152678 PMCID: PMC10162686 DOI: 10.1093/pnasnexus/pgad133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 03/07/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023]
Abstract
Immune checkpoint inhibitors, especially PD-1/PD-L1 blockade, have revolutionized cancer treatment and brought tremendous benefits to patients who otherwise would have had a limited prognosis. Nonetheless, only a small fraction of patients respond to immunotherapy, and the costs and side effects of immune checkpoint inhibitors cannot be ignored. With the advent of machine and deep learning, clinical and genetic data have been used to stratify patient responses to immunotherapy. Unfortunately, these approaches have typically been "black-box" methods that are unable to explain their predictions, thereby hindering their responsible clinical application. Herein, we developed a "white-box" Bayesian network model that achieves accurate and interpretable predictions of immunotherapy responses against nonsmall cell lung cancer (NSCLC). This tree-augmented naïve Bayes (TAN) model accurately predicted durable clinical benefits and distinguished two clinically significant subgroups with distinct prognoses. Furthermore, our state-of-the-art white-box TAN approach achieved greater accuracy than previous methods. We hope that our model will guide clinicians in selecting NSCLC patients who truly require immunotherapy and expect our approach to be easily applied to other types of cancer.
Collapse
Affiliation(s)
- Hideki Hozumi
- School of Medicine, Keio University, Tokyo 160-8582, Japan
| | | |
Collapse
|
8
|
Kulshrestha R, Saxena H, Kumar R, Spalgais S, Mrigpuri P, Goel N, Menon B, Rani M, Mahor P, Bhutani I. Subtyping of advanced lung cancer based on PD-L1 expression, tumor histopathology and mutation burden (EGFR and KRAS): a study from North India. Monaldi Arch Chest Dis 2023; 93. [PMID: 36723380 DOI: 10.4081/monaldi.2023.2449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/13/2022] [Indexed: 02/02/2023] Open
Abstract
Immune checkpoint inhibitor (PD-L1) therapy of advanced non-small-cell lung cancer (NSCLC) has variable outcomes. Tumor subtypes based on PD-L1 expression, histopathology, mutation burden is required for patient stratification and formulation of treatment guidelines. Lung cancers (n=57) diagnosed at Pathology department, VPCI (2018-2021) were retrospectively analyzed. PD-L1(SP263) expressed by tumor cells [low (<1%), medium (1-49%), high (≥50%)] was correlated with histopathology, microenvironment, EGFR, KRAS expression. Patients were categorized into high and low risk based on their: i) gender: males (n=47, 30-89 years), females (n=10, 45-80 years); ii) smoking history: males 26/47 (45.61%), females 1/10 (10%); iii) tumor subtyping: squamous cell carcinoma 15/57 (26.32%), adenocarcinoma 6/57 (17.54%), NSCLC-undifferentiated 24/57 (42.10%), adenosquamous carcinoma 5/57 (8.77 %), carcinosarcoma 4/57 (7.02%), small cell carcinoma 1/57 (1.75%); iv) inflammatory tumor microenvironment/TILs 44/57 (77.1%); iv) PD-L1 positivity-31/57 (54.3%); v) concomitant EGFR/KRAS positivity. PD-L1positive cases showed squamous/undifferentiated histopathology, concomitant EGFR+ (9/20, 45%) and KRAS+ (8/15, 53.3%), smoking+ (21/31,67.74%).PD-L1 negative cases (26/57, 45.6%), were EGFR+ (2/14, 14.28%) and KRAS+ (6/19, 31.5%). The high-risk lung cancer subtypes show squamous/undifferentiated histopathology, inflammatory microenvironment, male preponderance, smoking history, higher concomitant PD-L1, KRAS and EGFR positivity. Lung cancer subtyping can predict clinical response/resistance of patients prior to initiation of PD-L1 inhibitor therapies and can be used to guide therapy.
Collapse
Affiliation(s)
- Ritu Kulshrestha
- Department of Pathology, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Himanshi Saxena
- Department of Pathology, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Raj Kumar
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Sonam Spalgais
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Parul Mrigpuri
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Nitin Goel
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Balakrishnan Menon
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Meenu Rani
- Department of Pathology, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Pawan Mahor
- Department of Pathology, Vallabhbhai Patel Chest Institute, University of Delhi.
| | - Ishita Bhutani
- Department of Pathology, Vallabhbhai Patel Chest Institute, University of Delhi.
| |
Collapse
|
9
|
Xu D, Chen R, Jiang Y, Wang S, Liu Z, Chen X, Fan X, Zhu J, Li J. Application of machine learning in the prediction of deficient mismatch repair in patients with colorectal cancer based on routine preoperative characterization. Front Oncol 2022; 12:1049305. [PMID: 36620593 PMCID: PMC9814116 DOI: 10.3389/fonc.2022.1049305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Simple summary Detecting deficient mismatch repair (dMMR) in patients with colorectal cancer is essential for clinical decision-making, including evaluation of prognosis, guidance of adjuvant chemotherapy and immunotherapy, and primary screening for Lynch syndrome. However, outside of tertiary care centers, existing detection methods are not widely disseminated and highly depend on the experienced pathologist. Therefore, it is of great clinical significance to develop a broadly accessible and low-cost tool for dMMR prediction, particularly prior to surgery. In this study, we developed a convenient and reliable model for predicting dMMR status in CRC patients on routine preoperative characterization utilizing multiple machine learning algorithms. This model will work as an automated screening tool for identifying patients suitable for mismatch repair testing and consequently for improving the detection rate of dMMR, while reducing unnecessary labor and cost in patients with proficient mismatch repair. Background Deficient mismatch repair (dMMR) indicates a sustained anti-tumor immune response and has a favorable prognosis in patients with colorectal cancer (CRC). Although all CRC patients are recommended to undergo dMMR testing after surgery, current diagnostic approaches are not available for all country hospitals and patients. Therefore, efficient and low-cost predictive models for dMMR, especially for preoperative evaluations, are warranted. Methods A large scale of 5596 CRC patients who underwent surgical resection and mismatch repair testing were enrolled and randomly divided into training and validation cohorts. The clinical features exploited for predicting dMMR comprised the demographic characteristics, preoperative laboratory data, and tumor burden information. Machine learning (ML) methods involving eight basic algorithms, ensemble learning methods, and fusion algorithms were adopted with 10-fold cross-validation, and their performance was evaluated based on the area under the receiver operating characteristic curve (AUC) and calibration curves. The clinical net benefits were assessed using a decision curve analysis (DCA), and a nomogram was developed to facilitate model clinical practicality. Results All models achieved an AUC of nearly 0.80 in the validation cohort, with the stacking model exhibiting the best performance (AUC = 0.832). Logistical DCA revealed that the stacking model yielded more clinical net benefits than the conventional regression models. In the subgroup analysis, the stacking model also predicted dMMR regardless of the clinical stage. The nomogram showed a favorable consistence with the actual outcome in the calibration curve. Conclusion With the aid of ML algorithms, we developed a novel and robust model for predicting dMMR in CRC patients with satisfactory discriminative performance and designed a user-friendly and convenient nomogram.
Collapse
Affiliation(s)
- Dong Xu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi’an, China,School of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Rujie Chen
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi’an, China,Department of Neurosurgery, Xijing Hospital, Air Force Medical University, Xi’an, China,State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi’an, China
| | - Yu Jiang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi’an, China,School of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Shuai Wang
- Xi’an Institute of Flight of the Air Force, Ming Gang Station Hospital, Minggang, China
| | - Zhiyu Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi’an, China,School of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Xihao Chen
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi’an, China,School of Clinical Medicine, Xi’an Medical University, Xi’an, China
| | - Xiaoyan Fan
- Department of Experiment Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Jun Zhu
- Department of General Surgery, The Southern Theater Air Force Hospital, Guangzhou, China,*Correspondence: Jipeng Li, ; Jun Zhu,
| | - Jipeng Li
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, Air force Medical University, Xi’an, China,State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi’an, China,Department of Experiment Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China,*Correspondence: Jipeng Li, ; Jun Zhu,
| |
Collapse
|
10
|
Humayun M, Sujatha R, Almuayqil SN, Jhanjhi NZ. A Transfer Learning Approach with a Convolutional Neural Network for the Classification of Lung Carcinoma. Healthcare (Basel) 2022; 10:1058. [PMID: 35742109 PMCID: PMC9222675 DOI: 10.3390/healthcare10061058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/04/2022] [Accepted: 06/06/2022] [Indexed: 12/02/2022] Open
Abstract
Lung cancer is among the most hazardous types of cancer in humans. The correct diagnosis of pathogenic lung disease is critical for medication. Traditionally, determining the pathological form of lung cancer involves an expensive and time-consuming process investigation. Lung cancer is a leading cause of mortality worldwide, with lung tissue nodules being the most prevalent way for doctors to identify it. The proposed model is based on robust deep-learning-based lung cancer detection and recognition. This study uses a deep neural network as an extraction of features approach in a computer-aided diagnosing (CAD) system to assist in detecting lung illnesses at high definition. The proposed model is categorized into three phases: first, data augmentation is performed, classification is then performed using the pretrained CNN model, and lastly, localization is completed. The amount of obtained data in medical image assessment is occasionally inadequate to train the learning network. We train the classifier using a technique known as transfer learning (TL) to solve the issue introduced into the process. The proposed methodology offers a non-invasive diagnostic tool for use in the clinical assessment that is effective. The proposed model has a lower number of parameters that are much smaller compared to the state-of-the-art models. We also examined the desired dataset's robustness depending on its size. The standard performance metrics are used to assess the effectiveness of the proposed architecture. In this dataset, all TL techniques perform well, and VGG 16, VGG 19, and Xception for 20 epoch structure are compared. Preprocessing functions as a wonderful bridge to build a dependable model and eventually helps to forecast future scenarios by including the interface at a faster phase for any model. At the 20th epoch, the accuracy of VGG 16, VGG 19, and Xception is 98.83 percent, 98.05 percent, and 97.4 percent.
Collapse
Affiliation(s)
- Mamoona Humayun
- Department of Information Systems, College of Computer and Information Sciences, Jouf University, Sakakah 72312, Saudi Arabia;
| | - R. Sujatha
- School of Information Technology & Engineering, Vellore Institute of Technology, Vellore 632001, India;
| | - Saleh Naif Almuayqil
- Department of Information Systems, College of Computer and Information Sciences, Jouf University, Sakakah 72312, Saudi Arabia;
| | - N. Z. Jhanjhi
- School of Computer Science (SCS), Taylor’s University, Subang Jaya 47500, Malaysia;
| |
Collapse
|
11
|
Effect of Family Participatory Nursing Model Based on WeChat Platform on Psychological Elasticity and Quality of Life of Patients with Lung Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4704107. [PMID: 35578722 PMCID: PMC9107353 DOI: 10.1155/2022/4704107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 11/24/2022]
Abstract
Objective To explore the effect of family participatory nursing model based on WeChat platform on psychological elasticity and life quality of patients with lung cancer. Methods One hundred patients with lung cancer treated in our hospital from February 2019 to April 2021 were enrolled. The patients were randomly assigned into control group and study group. The control group received routine nursing and the study group received family participatory nursing model based on WeChat platform. The scores of nursing satisfaction, self-feeling burden, symptom group assessment scale, family care index, and compliance and life quality were compared. Results First of all, we compared the nursing satisfaction: the study group was very satisfied in 25 cases, satisfactory in 20 cases, general in 5 cases, the satisfaction rate was 100.00%; the control group was very satisfied in 14 cases, satisfactory in 17 cases, general in 13 cases, dissatisfied in 6 cases, and the satisfaction rate was 88.00%. Compared between the two groups, the nursing satisfaction in the study group was higher compared to the control group (P < 0.05). Secondly, we compared the self-perceived burden, the emotional burden, physical burden, and economic burden and total scores of the study group were lower compared to the control group (P < 0.05). Comparing the scores of symptom group evaluation scale, the scores of respiratory symptoms, fatigue symptoms, psychological and emotional symptoms, digestive tract symptoms, and nervous system symptoms in the study group were lower compared to the control group (P < 0.05). In terms of the family care index of the two groups, the scores of fitness, cooperation, length, emotion, and intimacy in the study group were higher compared to the control group (P < 0.05). Comparison of compliance, the study group was very compliant in 42 cases, compliant in 7 cases, noncompliant in 1 case, the compliance rate was 98.00%; the control group was very compliant in 24 cases, compliant in 20 cases, noncompliant in 6 cases, the compliance rate was 88.00%. The compliance rate in the study group was higher compared to the control group (P < 0.05). Finally, we compared the scores of qualities of life. Before nursing, there exhibited no significant difference (P > 0.05). The scores of physiological function, psychological function, social function, and health self-cognition in the study group were lower compared to the control group (P < 0.05). Conclusion The family participatory nursing model based on WeChat platform helps to facilitate the psychological elasticity and quality of life of patients with lung cancer and further promote patients' self-care ability and compliance with lung cancer. It can also effectively enhance family care and nursing satisfaction; the nursing model is worth popularizing and applying in clinic.
Collapse
|
12
|
Wang Y, Cai H, Pu Y, Li J, Yang F, Yang C, Chen L, Hu Z. The value of AI in the Diagnosis, Treatment, and Prognosis of Malignant Lung Cancer. FRONTIERS IN RADIOLOGY 2022; 2:810731. [PMID: 37492685 PMCID: PMC10365105 DOI: 10.3389/fradi.2022.810731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 03/30/2022] [Indexed: 07/27/2023]
Abstract
Malignant tumors is a serious public health threat. Among them, lung cancer, which has the highest fatality rate globally, has significantly endangered human health. With the development of artificial intelligence (AI) and its integration with medicine, AI research in malignant lung tumors has become critical. This article reviews the value of CAD, computer neural network deep learning, radiomics, molecular biomarkers, and digital pathology for the diagnosis, treatment, and prognosis of malignant lung tumors.
Collapse
Affiliation(s)
- Yue Wang
- Department of PET/CT Center, Cancer Center of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haihua Cai
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yongzhu Pu
- Department of PET/CT Center, Cancer Center of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jindan Li
- Department of PET/CT Center, Cancer Center of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fake Yang
- Department of PET/CT Center, Cancer Center of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Conghui Yang
- Department of PET/CT Center, Cancer Center of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Long Chen
- Department of PET/CT Center, Cancer Center of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhanli Hu
- Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
13
|
Gong X, Zheng B, Xu G, Chen H, Chen C. Application of machine learning approaches to predict the 5-year survival status of patients with esophageal cancer. J Thorac Dis 2022; 13:6240-6251. [PMID: 34992804 PMCID: PMC8662490 DOI: 10.21037/jtd-21-1107] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/24/2021] [Indexed: 01/15/2023]
Abstract
Background Accurate prognostic estimation for esophageal cancer (EC) patients plays an important role in the process of clinical decision-making. The objective of this study was to develop an effective model to predict the 5-year survival status of EC patients using machine learning (ML) algorithms. Methods We retrieved the information of patients diagnosed with EC between 2010 and 2015 from the Surveillance, Epidemiology, and End Results (SEER) Program, including 24 features. A total of 8 ML models were applied to the selected dataset to classify the EC patients in terms of 5-year survival status, including 3 newly developed gradient boosting models (GBM), XGBoost, CatBoost, and LightGBM, 2 commonly used tree-based models, gradient boosting decision trees (GBDT) and random forest (RF), and 3 other ML models, artificial neural networks (ANN), naive Bayes (NB), and support vector machines (SVM). A 5-fold cross-validation was used in model performance measurement. Results After excluding records with missing data, the final study population comprised 10,588 patients. Feature selection was conducted based on the χ2 test, however, the experiment results showed that the complete dataset provided better prediction of outcomes than the dataset with removal of non-significant features. Among the 8 models, XGBoost had the best performance [area under the receiver operating characteristic (ROC) curve (AUC): 0.852 for XGBoost, 0.849 for CatBoost, 0.850 for LightGBM, 0.846 for GBDT, 0.838 for RF, 0.844 for ANN, 0.833 for NB, and 0.789 for SVM]. The accuracy and logistic loss of XGBoost were 0.875 and 0.301, respectively, which were also the best performances. In the XGBoost model, the SHapley Additive exPlanations (SHAP) value was calculated and the result indicated that the four features: reason no cancer-directed surgery, Surg Prim Site, age, and stage group had the greatest impact on predicting the outcomes. Conclusions The XGBoost model and the complete dataset can be used to construct an accurate prognostic model for patients diagnosed with EC which may be applicable in clinical practice in the future.
Collapse
Affiliation(s)
- Xian Gong
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Bin Zheng
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Guobing Xu
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Hao Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| | - Chun Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Cardio-Thoracic Surgery (Fujian Medical University), Fujian Province University, Fuzhou, China
| |
Collapse
|
14
|
Machine Learning for Prediction of Immunotherapy Efficacy in Non-Small Cell Lung Cancer from Simple Clinical and Biological Data. Cancers (Basel) 2021; 13:cancers13246210. [PMID: 34944830 PMCID: PMC8699503 DOI: 10.3390/cancers13246210] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/30/2021] [Accepted: 12/03/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are now a therapeutic standard in advanced non-small cell lung cancer (NSCLC), but strong predictive markers for ICIs efficacy are still lacking. We evaluated machine learning models built on simple clinical and biological data to individually predict response to ICIs. METHODS Patients with metastatic NSCLC who received ICI in second line or later were included. We collected clinical and hematological data and studied the association of this data with disease control rate (DCR), progression free survival (PFS) and overall survival (OS). Multiple machine learning (ML) algorithms were assessed for their ability to predict response. RESULTS Overall, 298 patients were enrolled. The overall response rate and DCR were 15.3% and 53%, respectively. Median PFS and OS were 3.3 and 11.4 months, respectively. In multivariable analysis, DCR was significantly associated with performance status (PS) and hemoglobin level (OR 0.58, p < 0.0001; OR 1.8, p < 0.001). These variables were also associated with PFS and OS and ranked top in random forest-based feature importance. Neutrophil-to-lymphocyte ratio was also associated with DCR, PFS and OS. The best ML algorithm was a random forest. It could predict DCR with satisfactory efficacy based on these three variables. Ten-fold cross-validated performances were: accuracy 0.68 ± 0.04, sensitivity 0.58 ± 0.08; specificity 0.78 ± 0.06; positive predictive value 0.70 ± 0.08; negative predictive value 0.68 ± 0.06; AUC 0.74 ± 0.03. CONCLUSION Combination of simple clinical and biological data could accurately predict disease control rate at the individual level.
Collapse
|