1
|
Xie J, Ma RW, Feng YJ, Qiao Y, Zhu HY, Tao XP, Chen WJ, Liu CY, Li T, Liu K, Cheng LM. Machine learning-based risk prediction model for pertussis in children: a multicenter retrospective study. BMC Infect Dis 2025; 25:428. [PMID: 40148755 PMCID: PMC11951648 DOI: 10.1186/s12879-025-10797-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Pertussis is a highly contagious respiratory disease. Even though vaccination has reduced the incidence, cases have resurfaced in certain regions due to immune escape and waning vaccine efficacy. Identifying high-risk patients to mitigate transmission and avert complications promptly is imperative. Nevertheless, the current diagnostic methods, including PCR and bacterial culture, are time-consuming and expensive. Some studies have attempted to develop risk prediction models based on multivariate data, but their performance can be improved. Therefore, this study aims to further optimize and expand the risk assessment tool to more efficiently identify high-risk individuals and compensate for the shortcomings of existing diagnostic methods. OBJECTIVE The aim of this study was to develop a pertussis risk prediction model that is both efficient and has good generalization ability, applicable to different datasets. The model was constructed using machine learning techniques based on multicenter data and screened for key features. The performance and generalization ability of the model were evaluated by deploying it on an online platform. At the same time, this study aims to provide a rapid and accurate auxiliary diagnostic tool for clinical practice to help identify high-risk patients in a timely manner, optimize early intervention strategies, reduce the risk of complications and reduce transmission, thereby improving the efficiency of public health management. METHODS First, data from 1085 suspected pertussis patients from 7 centers were collected, and ten key features were analyzed using the lasso regression and Boruta algorithm: PDW-MPV-RATIO, SII, white blood cells, platelet distribution width, mean platelet volume, lymphocytes, cough duration, vaccination, fever, and lytic lymphocytes.Eight models were then trained and validated to assess their performance and to confirm their generalization ability with external datasets based on these features. Finally, an online platform was constructed for clinicians to use the models in real time. RESULTS The random forest model demonstrated excellent discrimination ability in the validation set, with an AUC of 0.98, and an AUC of 0.97 in the external validation set. Calibration curve and decision curve analysis showed that the model had high accuracy in predicting low-to-medium risk patients, which could help clinicians avoid unnecessary interventions, especially in resource-limited settings. The application of this model can help optimize the early identification and management of high-risk patients and improve clinical decision-making. CONCLUSION The pertussis prediction model devised in this study was validated using multicenter data, exhibited high prediction performance, and was successfully implemented online. Future research should broaden the data sources and incorporate dynamic data to enhance the model's accuracy and applicability.
Collapse
Affiliation(s)
- Juan Xie
- Department of Anesthesiology, Kunming Children'S Hospital, Kunming City, Yunnan Province, China
| | - Run-Wei Ma
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences/Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming City, Yunnan Province, China
| | - Yu-Jing Feng
- Comprehensive Pediatrics, Wenshan Maternal and Child Health Care Hospital, Wenshan City, Yunnan Province, China
| | - Yuan Qiao
- Comprehensive Pediatrics and Neonatology, Chuxiong Yi Autonomous Prefecture People's Hospital, Chuxiong City, Yunnan Province, China
| | - Hong-Yan Zhu
- Pediatric Respiratory Department, Qujing Maternal and Child Health Hospital, Qujing City, Yunnan Province, China
| | - Xing-Ping Tao
- Department of Pediatrics, Kaiyuan People's Hospital, Kaiyuan, China
| | - Wen-Juan Chen
- Department of Pediatrics and Emergency, Yuxi Children'S Hospital, Yuxi City, Yunnan Province, China
| | - Cong-Yun Liu
- Comprehensive Pediatrics & Pulmonary and Critical Care Medicine, Baoshan People's Hospital, Baoshan City, Yunnan Province, China
| | - Tan Li
- Department of Respiratory Medicine Kunming Children'S Hospital, Kunming City, Yunnan Province, China
| | - Kai Liu
- Comprehensive Pediatrics & Pulmonary and Critical Care Medicine, Kunming Children'S Hospital, Yunnan Province, Shulin Street 28, Kunming City, Yunnan Province, 650000, China.
| | - Li-Ming Cheng
- Department of Anesthesiology, Kunming Children'S Hospital, Kunming City, Yunnan Province, China.
| |
Collapse
|
2
|
Gholap AD, Uddin MJ, Faiyazuddin M, Omri A, Gowri S, Khalid M. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Comput Biol Med 2024; 178:108702. [PMID: 38878397 DOI: 10.1016/j.compbiomed.2024.108702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/12/2024] [Accepted: 06/01/2024] [Indexed: 07/24/2024]
Abstract
Artificial intelligence (AI) has emerged as a powerful tool to revolutionize the healthcare sector, including drug delivery and development. This review explores the current and future applications of AI in the pharmaceutical industry, focusing on drug delivery and development. It covers various aspects such as smart drug delivery networks, sensors, drug repurposing, statistical modeling, and simulation of biotechnological and biological systems. The integration of AI with nanotechnologies and nanomedicines is also examined. AI offers significant advancements in drug discovery by efficiently identifying compounds, validating drug targets, streamlining drug structures, and prioritizing response templates. Techniques like data mining, multitask learning, and high-throughput screening contribute to better drug discovery and development innovations. The review discusses AI applications in drug formulation and delivery, clinical trials, drug safety, and pharmacovigilance. It addresses regulatory considerations and challenges associated with AI in pharmaceuticals, including privacy, data security, and interpretability of AI models. The review concludes with future perspectives, highlighting emerging trends, addressing limitations and biases in AI models, and emphasizing the importance of collaboration and knowledge sharing. It provides a comprehensive overview of AI's potential to transform the pharmaceutical industry and improve patient care while identifying further research and development areas.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra, 401404, India.
| | - Md Jasim Uddin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| | - Md Faiyazuddin
- School of Pharmacy, Al-Karim University, Katihar, Bihar, 854106, India; Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Tamil Nadu, India.
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON, P3E 2C6, Canada.
| | - S Gowri
- PG & Research, Department of Physics, Cauvery College for Women, Tiruchirapalli, Tamil Nadu, 620018, India
| | - Mohammad Khalid
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK; Sunway Centre for Electrochemical Energy and Sustainable Technology (SCEEST), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia; University Centre for Research and Development, Chandigarh University, Mohali, Punjab, 140413, India.
| |
Collapse
|
3
|
Sinha T, Khan A, Awan M, Bokhari SFH, Ali K, Amir M, Jadhav AN, Bakht D, Puli ST, Burhanuddin M. Artificial Intelligence and Machine Learning in Predicting the Response to Immunotherapy in Non-small Cell Lung Carcinoma: A Systematic Review. Cureus 2024; 16:e61220. [PMID: 38939246 PMCID: PMC11210434 DOI: 10.7759/cureus.61220] [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] [Accepted: 05/27/2024] [Indexed: 06/29/2024] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is a prevalent and aggressive form of lung cancer, with a poor prognosis for metastatic disease. Immunotherapy, particularly immune checkpoint inhibitors (ICIs), has revolutionized the management of NSCLC, but response rates are highly variable. Identifying reliable predictive biomarkers is crucial to optimize patient selection and treatment outcomes. This systematic review aimed to evaluate the current state of artificial intelligence (AI) and machine learning (ML) applications in predicting the response to immunotherapy in NSCLC. A comprehensive literature search identified 19 studies that met the inclusion criteria. The studies employed diverse AI/ML techniques, including deep learning, artificial neural networks, support vector machines, and gradient boosting methods, applied to various data modalities such as medical imaging, genomic data, clinical variables, and immunohistochemical markers. Several studies demonstrated the ability of AI/ML models to accurately predict immunotherapy response, progression-free survival, and overall survival in NSCLC patients. However, challenges remain in data availability, quality, and interpretability of these models. Efforts have been made to develop interpretable AI/ML techniques, but further research is needed to improve transparency and explainability. Additionally, translating AI/ML models from research settings to clinical practice poses challenges related to regulatory approval, data privacy, and integration into existing healthcare systems. Nonetheless, the successful implementation of AI/ML models could enable personalized treatment strategies, improve treatment outcomes, and reduce unnecessary toxicities and healthcare costs associated with ineffective treatments.
Collapse
Affiliation(s)
- Tanya Sinha
- Internal Medicine, Tribhuvan University, Kathmandu, NPL
| | - Aiman Khan
- Medicine, Liaquat College of Medicine and Dentistry, Karachi, PAK
| | - Manahil Awan
- General Practice, Liaquat National Hospital and Medical College, Karachi, PAK
| | | | - Khawar Ali
- Medicine and Surgery, King Edward Medical University, Lahore, PAK
| | - Maaz Amir
- Medicine and Surgery, King Edward Medical University, Lahore, PAK
| | - Aneesh N Jadhav
- Pediatrics, Bharat Ratna Dr. Babasaheb Ambedkar Memorial Hospital, Mumbai, IND
| | - Danyal Bakht
- Medicine and Surgery, Mayo Hospital, Lahore, PAK
| | - Sai Teja Puli
- Internal Medicine, Bhaskar Medical College, Hyderabad, IND
| | | |
Collapse
|
4
|
Didier AJ, Nigro A, Noori Z, Omballi MA, Pappada SM, Hamouda DM. Application of machine learning for lung cancer survival prognostication-A systematic review and meta-analysis. Front Artif Intell 2024; 7:1365777. [PMID: 38646415 PMCID: PMC11026647 DOI: 10.3389/frai.2024.1365777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/18/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction Machine learning (ML) techniques have gained increasing attention in the field of healthcare, including predicting outcomes in patients with lung cancer. ML has the potential to enhance prognostication in lung cancer patients and improve clinical decision-making. In this systematic review and meta-analysis, we aimed to evaluate the performance of ML models compared to logistic regression (LR) models in predicting overall survival in patients with lung cancer. Methods We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) statement. A comprehensive search was conducted in Medline, Embase, and Cochrane databases using a predefined search query. Two independent reviewers screened abstracts and conflicts were resolved by a third reviewer. Inclusion and exclusion criteria were applied to select eligible studies. Risk of bias assessment was performed using predefined criteria. Data extraction was conducted using the Critical Appraisal and Data Extraction for Systematic Reviews of Prediction Modeling Studies (CHARMS) checklist. Meta-analytic analysis was performed to compare the discriminative ability of ML and LR models. Results The literature search resulted in 3,635 studies, and 12 studies with a total of 211,068 patients were included in the analysis. Six studies reported confidence intervals and were included in the meta-analysis. The performance of ML models varied across studies, with C-statistics ranging from 0.60 to 0.85. The pooled analysis showed that ML models had higher discriminative ability compared to LR models, with a weighted average C-statistic of 0.78 for ML models compared to 0.70 for LR models. Conclusion Machine learning models show promise in predicting overall survival in patients with lung cancer, with superior discriminative ability compared to logistic regression models. However, further validation and standardization of ML models are needed before their widespread implementation in clinical practice. Future research should focus on addressing the limitations of the current literature, such as potential bias and heterogeneity among studies, to improve the accuracy and generalizability of ML models for predicting outcomes in patients with lung cancer. Further research and development of ML models in this field may lead to improved patient outcomes and personalized treatment strategies.
Collapse
Affiliation(s)
- Alexander J. Didier
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Anthony Nigro
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Zaid Noori
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Mohamed A. Omballi
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Scott M. Pappada
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
- Department of Anesthesiology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Danae M. Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
- Division of Hematology and Oncology, Department of Medicine, The University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| |
Collapse
|
5
|
Sathish G, Monavarshini LK, Sundaram K, Subramanian S, Kannayiram G. Immunotherapy for lung cancer. Pathol Res Pract 2024; 254:155104. [PMID: 38244436 DOI: 10.1016/j.prp.2024.155104] [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: 12/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/22/2024]
Abstract
Immune checkpoint blockers have transformed non-small-cell lung cancer treatment, but they can lead to autoimmune and inflammatory side effects, leading to the concurrent use of immunosuppressive treatments. In this analysis, we delve into the potential of antibodies checkpoint blockade, focusing on CTLA-4 inhibition using ipilimumab, as a groundbreaking cancer immunotherapy. We also concentrate on the role of biomarkers, particularly PD-L1 activity and mutation significance, in predicting the response to programmed cell death protein 1 blockage and the prevalence of side effects associated with immune-related side effects. In describing the patterns of cancer response to immunotherapy, we underline the limitations of response assessment criteria like RECIST and World Health Organization. We also stress the necessity of ongoing studies and clinical trials, standardized guidelines, and additional research to improve response assessment in the era of immunotherapy.
Collapse
Affiliation(s)
- Girshani Sathish
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - L K Monavarshini
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Keerthi Sundaram
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Sendilvelan Subramanian
- Deparment of Mechanical Engineering, Dr.MGR Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Gomathi Kannayiram
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India.
| |
Collapse
|
6
|
Yeramosu T, Ahmad W, Bashir A, Wait J, Bassett J, Domson G. Predicting five-year mortality in soft-tissue sarcoma patients. Bone Joint J 2023; 105-B:702-710. [PMID: 37257862 DOI: 10.1302/0301-620x.105b6.bjj-2022-0998.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Aims The aim of this study was to identify factors associated with five-year cancer-related mortality in patients with limb and trunk soft-tissue sarcoma (STS) and develop and validate machine learning algorithms in order to predict five-year cancer-related mortality in these patients. Methods Demographic, clinicopathological, and treatment variables of limb and trunk STS patients in the Surveillance, Epidemiology, and End Results Program (SEER) database from 2004 to 2017 were analyzed. Multivariable logistic regression was used to determine factors significantly associated with five-year cancer-related mortality. Various machine learning models were developed and compared using area under the curve (AUC), calibration, and decision curve analysis. The model that performed best on the SEER testing data was further assessed to determine the variables most important in its predictive capacity. This model was externally validated using our institutional dataset. Results A total of 13,646 patients with STS from the SEER database were included, of whom 35.9% experienced five-year cancer-related mortality. The random forest model performed the best overall and identified tumour size as the most important variable when predicting mortality in patients with STS, followed by M stage, histological subtype, age, and surgical excision. Each variable was significant in logistic regression. External validation yielded an AUC of 0.752. Conclusion This study identified clinically important variables associated with five-year cancer-related mortality in patients with limb and trunk STS, and developed a predictive model that demonstrated good accuracy and predictability. Orthopaedic oncologists may use these findings to further risk-stratify their patients and recommend an optimal course of treatment.
Collapse
Affiliation(s)
- Teja Yeramosu
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Waleed Ahmad
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Azhar Bashir
- Department of Orthopaedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jacob Wait
- School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - James Bassett
- College of Medicine and Life Sciences, University of Toledo, Toledo, Ohio, USA
| | - Gregory Domson
- Department of Orthopaedic Surgery, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
7
|
Prelaj A, Galli EG, Miskovic V, Pesenti M, Viscardi G, Pedica B, Mazzeo L, Bottiglieri A, Provenzano L, Spagnoletti A, Marinacci R, De Toma A, Proto C, Ferrara R, Brambilla M, Occhipinti M, Manglaviti S, Galli G, Signorelli D, Giani C, Beninato T, Pircher CC, Rametta A, Kosta S, Zanitti M, Di Mauro MR, Rinaldi A, Di Gregorio S, Antonia M, Garassino MC, de Braud FGM, Restelli M, Lo Russo G, Ganzinelli M, Trovò F, Pedrocchi ALG. Real-world data to build explainable trustworthy artificial intelligence models for prediction of immunotherapy efficacy in NSCLC patients. Front Oncol 2023; 12:1078822. [PMID: 36755856 PMCID: PMC9899835 DOI: 10.3389/fonc.2022.1078822] [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: 10/24/2022] [Accepted: 12/14/2022] [Indexed: 01/24/2023] Open
Abstract
Introduction Artificial Intelligence (AI) methods are being increasingly investigated as a means to generate predictive models applicable in the clinical practice. In this study, we developed a model to predict the efficacy of immunotherapy (IO) in patients with advanced non-small cell lung cancer (NSCLC) using eXplainable AI (XAI) Machine Learning (ML) methods. Methods We prospectively collected real-world data from patients with an advanced NSCLC condition receiving immune-checkpoint inhibitors (ICIs) either as a single agent or in combination with chemotherapy. With regards to six different outcomes - Disease Control Rate (DCR), Objective Response Rate (ORR), 6 and 24-month Overall Survival (OS6 and OS24), 3-months Progression-Free Survival (PFS3) and Time to Treatment Failure (TTF3) - we evaluated five different classification ML models: CatBoost (CB), Logistic Regression (LR), Neural Network (NN), Random Forest (RF) and Support Vector Machine (SVM). We used the Shapley Additive Explanation (SHAP) values to explain model predictions. Results Of 480 patients included in the study 407 received immunotherapy and 73 chemo- and immunotherapy. From all the ML models, CB performed the best for OS6 and TTF3, (accuracy 0.83 and 0.81, respectively). CB and LR reached accuracy of 0.75 and 0.73 for the outcome DCR. SHAP for CB demonstrated that the feature that strongly influences models' prediction for all three outcomes was Neutrophil to Lymphocyte Ratio (NLR). Performance Status (ECOG-PS) was an important feature for the outcomes OS6 and TTF3, while PD-L1, Line of IO and chemo-immunotherapy appeared to be more important in predicting DCR. Conclusions In this study we developed a ML algorithm based on real-world data, explained by SHAP techniques, and able to accurately predict the efficacy of immunotherapy in sets of NSCLC patients.
Collapse
Affiliation(s)
- Arsela Prelaj
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy,*Correspondence: Arsela Prelaj,
| | - Edoardo Gregorio Galli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Vanja Miskovic
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Mattia Pesenti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giuseppe Viscardi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Medical Oncology Unit, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Benedetta Pedica
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Laura Mazzeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Achille Bottiglieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Leonardo Provenzano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Andrea Spagnoletti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Roberto Marinacci
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alessandro De Toma
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Roberto Ferrara
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marta Brambilla
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Mario Occhipinti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Sara Manglaviti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Giulia Galli
- Medical Oncology Unit, Policlinico San Matteo Fondazione IRCCS, Pavia, Italy
| | - Diego Signorelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Claudia Giani
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Teresa Beninato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Chiara Carlotta Pircher
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Alessandro Rametta
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Sokol Kosta
- Department of Electronic System, Aalborg University, Copenhagen, Aalborg, Denmark
| | - Michele Zanitti
- Department of Electronic System, Aalborg University, Copenhagen, Aalborg, Denmark
| | - Maria Rosa Di Mauro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Arturo Rinaldi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Settimio Di Gregorio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Martinetti Antonia
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marina Chiara Garassino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Thoracic Oncology Program, Section of Hematology/Oncology, University of Chicago, Chicago, IL, United States
| | - Filippo G. M. de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy,Oncology Department, University of Milan, Milan, Italy
| | - Marcello Restelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giuseppe Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Francesco Trovò
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | | |
Collapse
|
8
|
Artificial Intelligence Applied to clinical trials: opportunities and challenges. HEALTH AND TECHNOLOGY 2023; 13:203-213. [PMID: 36923325 PMCID: PMC9974218 DOI: 10.1007/s12553-023-00738-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023]
Abstract
Background Clinical Trials (CTs) remain the foundation of safe and effective drug development. Given the evolving data-driven and personalized medicine approach in healthcare, it is imperative for companies and regulators to utilize tailored Artificial Intelligence (AI) solutions that enable expeditious and streamlined clinical research. In this paper, we identified opportunities, challenges, and potential implications of AI in CTs. Methods Following an extensive search in relevant databases and websites, we gathered publications tackling the use of AI and Machine Learning (ML) in CTs from the past 5 years in the US and Europe, including Regulatory Authorities' documents. Results Documented applications of AI commonly concern the oncology field and are mostly being applied in the area of recruitment. Main opportunities discussed aim to create efficiencies across CT activities, including the ability to reduce sample sizes, improve enrollment and conduct faster, more optimized adaptive CTs. While AI is an area of enthusiastic development, the identified challenges are ethical in nature and relate to data availability, standards, and most importantly, lack of regulatory guidance hindering the acceptance of AI tools in drug development. However, future implications are significant and are anticipated to improve the probability of success, reduce trial burden and overall, speed up research and regulatory approval. Conclusion The use of AI in CTs is in its relative infancy; however, it is a fast-evolving field. As regulators provide more guidance on the acceptability of AI in specific areas, we anticipate the scope of use to broaden and the volume of implementation to increase rapidly.
Collapse
|
9
|
Development and Validation of Novel Deep-Learning Models Using Multiple Data Types for Lung Cancer Survival. Cancers (Basel) 2022; 14:cancers14225562. [PMID: 36428655 PMCID: PMC9688689 DOI: 10.3390/cancers14225562] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
A well-established lung-cancer-survival-prediction model that relies on multiple data types, multiple novel machine-learning algorithms, and external testing is absent in the literature. This study aims to address this gap and determine the critical factors of lung cancer survival. We selected non-small-cell lung cancer patients from a retrospective dataset of the Taipei Medical University Clinical Research Database and Taiwan Cancer Registry between January 2008 and December 2018. All patients were monitored from the index date of cancer diagnosis until the event of death. Variables, including demographics, comorbidities, medications, laboratories, and patient gene tests, were used. Nine machine-learning algorithms with various modes were used. The performance of the algorithms was measured by the area under the receiver operating characteristic curve (AUC). In total, 3714 patients were included. The best performance of the artificial neural network (ANN) model was achieved when integrating all variables with the AUC, accuracy, precision, recall, and F1-score of 0.89, 0.82, 0.91, 0.75, and 0.65, respectively. The most important features were cancer stage, cancer size, age of diagnosis, smoking, drinking status, EGFR gene, and body mass index. Overall, the ANN model improved predictive performance when integrating different data types.
Collapse
|
10
|
Damrauer JS, Beckabir W, Klomp J, Zhou M, Plimack ER, Galsky MD, Grivas P, Hahn NM, O'Donnell PH, Iyer G, Quinn DI, Vincent BG, Quale DZ, Wobker SE, Hoadley KA, Kim WY, Milowsky MI. Collaborative study from the Bladder Cancer Advocacy Network for the genomic analysis of metastatic urothelial cancer. Nat Commun 2022; 13:6658. [PMID: 36333289 PMCID: PMC9636269 DOI: 10.1038/s41467-022-33980-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
Urothelial Cancer - Genomic Analysis to Improve Patient Outcomes and Research (NCT02643043), UC-GENOME, is a genomic analysis and biospecimen repository study in 218 patients with metastatic urothelial carcinoma. Here we report on the primary outcome of the UC-GENOME-the proportion of subjects who received next generation sequencing (NGS) with treatment options-and present the initial genomic analyses and clinical correlates. 69.3% of subjects had potential treatment options, however only 5.0% received therapy based on NGS. We found an increased frequency of TP53E285K mutations as compared to non-metastatic cohorts and identified features associated with benefit to chemotherapy and immune checkpoint inhibition, including: Ba/Sq and Stroma-rich subtypes, APOBEC mutational signature (SBS13), and inflamed tumor immune phenotype. Finally, we derive a computational model incorporating both genomic and clinical features predictive of immune checkpoint inhibitor response. Future work will utilize the biospecimens alongside these foundational analyses toward a better understanding of urothelial carcinoma biology.
Collapse
Affiliation(s)
- Jeffrey S Damrauer
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Wolfgang Beckabir
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Jeff Klomp
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA
| | - Mi Zhou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Elizabeth R Plimack
- Department of Hematology and Oncology, Fox Chase Cancer Center, Temple Health, Philadelphia, PA, USA
| | - Matthew D Galsky
- Division of Hematology and Medical Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Petros Grivas
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, USA
| | - Noah M Hahn
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter H O'Donnell
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Gopa Iyer
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David I Quinn
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
- Division of Hematology, University of North Carolina, Chapel Hill, NC, USA
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Bioinformatics and Computational Biology, Computational Medicine Program, University of North Carolina, Chapel Hill, USA
| | | | - Sara E Wobker
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC, USA.
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Division of Oncology, University of North Carolina, Chapel Hill, NC, USA.
| | - Matthew I Milowsky
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA.
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA.
- Division of Oncology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Multi-Omics Approaches for the Prediction of Clinical Endpoints after Immunotherapy in Non-Small Cell Lung Cancer: A Comprehensive Review. Biomedicines 2022; 10:biomedicines10061237. [PMID: 35740259 PMCID: PMC9219996 DOI: 10.3390/biomedicines10061237] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/04/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized the management of locally advanced and advanced non-small lung cancer (NSCLC). With an improvement in the overall survival (OS) as both first- and second-line treatments, ICIs, and especially programmed-death 1 (PD-1) and programmed-death ligands 1 (PD-L1), changed the landscape of thoracic oncology. The PD-L1 level of expression is commonly accepted as the most used biomarker, with both prognostic and predictive values. However, even in a low expression level of PD-L1, response rates remain significant while a significant number of patients will experience hyperprogression or adverse events. The dentification of such subtypes is thus of paramount importance. While several studies focused mainly on the prediction of the PD-L1 expression status, others aimed directly at the development of prediction/prognostic models. The response to ICIs depends on a complex physiopathological cascade, intricating multiple mechanisms from the molecular to the macroscopic level. With the high-throughput extraction of features, omics approaches aim for the most comprehensive assessment of each patient. In this article, we will review the place of the different biomarkers (clinical, biological, genomics, transcriptomics, proteomics and radiomics), their clinical implementation and discuss the most recent trends projecting on the future steps in prediction modeling in NSCLC patients treated with ICI.
Collapse
|
12
|
Hindocha S, Charlton TG, Linton-Reid K, Hunter B, Chan C, Ahmed M, Robinson EJ, Orton M, Ahmad S, McDonald F, Locke I, Power D, Blackledge M, Lee RW, Aboagye EO. A comparison of machine learning methods for predicting recurrence and death after curative-intent radiotherapy for non-small cell lung cancer: Development and validation of multivariable clinical prediction models. EBioMedicine 2022; 77:103911. [PMID: 35248997 PMCID: PMC8897583 DOI: 10.1016/j.ebiom.2022.103911] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Surveillance is universally recommended for non-small cell lung cancer (NSCLC) patients treated with curative-intent radiotherapy. High-quality evidence to inform optimal surveillance strategies is lacking. Machine learning demonstrates promise in accurate outcome prediction for a variety of health conditions. The purpose of this study was to utilise readily available patient, tumour, and treatment data to develop, validate and externally test machine learning models for predicting recurrence, recurrence-free survival (RFS) and overall survival (OS) at 2 years from treatment. METHODS A retrospective, multicentre study of patients receiving curative-intent radiotherapy for NSCLC was undertaken. A total of 657 patients from 5 hospitals were eligible for inclusion. Data pre-processing derived 34 features for predictive modelling. Combinations of 8 feature reduction methods and 10 machine learning classification algorithms were compared, producing risk-stratification models for predicting recurrence, RFS and OS. Models were compared with 10-fold cross validation and an external test set and benchmarked against TNM-stage and performance status. Youden Index was derived from validation set ROC curves to distinguish high and low risk groups and Kaplan-Meier analyses performed. FINDINGS Median follow-up time was 852 days. Parameters were well matched across training-validation and external test sets: Mean age was 73 and 71 respectively, and recurrence, RFS and OS rates at 2 years were 43% vs 34%, 54% vs 47% and 54% vs 47% respectively. The respective validation and test set AUCs were as follows: 1) RFS: 0·682 (0·575-0·788) and 0·681 (0·597-0·766), 2) Recurrence: 0·687 (0·582-0·793) and 0·722 (0·635-0·81), and 3) OS: 0·759 (0·663-0·855) and 0·717 (0·634-0·8). Our models were superior to TNM stage and performance status in predicting recurrence and OS. INTERPRETATION This robust and ready to use machine learning method, validated and externally tested, sets the stage for future clinical trials entailing quantitative personalised risk-stratification and surveillance following curative-intent radiotherapy for NSCLC. FUNDING A full list of funding bodies that contributed to this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Sumeet Hindocha
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London SW36JJ, UK; AI for Healthcare Centre for Doctoral Training, Imperial College London, Exhibition Road, London SW7 2BX, UK; Department of Clinical Oncology, Institute of Cancer Research NIHR Biomedical Research Centre, London, UK; Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK; Early Diagnosis and Detection Centre, National Institute for Health Research (NIHR) Biomedical Research Centre at The Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, London
| | - Thomas G Charlton
- Guy's Cancer Centre, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London SE19RT UK
| | - Kristofer Linton-Reid
- Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Benjamin Hunter
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London SW36JJ, UK; Department of Clinical Oncology, Institute of Cancer Research NIHR Biomedical Research Centre, London, UK; Cancer Imaging Centre, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London W12 0NN, UK; Early Diagnosis and Detection Centre, National Institute for Health Research (NIHR) Biomedical Research Centre at The Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, London
| | - Charleen Chan
- Department of Clinical Oncology, Institute of Cancer Research NIHR Biomedical Research Centre, London, UK
| | - Merina Ahmed
- Lung Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM25PT, UK
| | - Emily J Robinson
- Clinical Trials Unit, Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM25PT, UK
| | - Matthew Orton
- Artificial Intelligence Imaging Hub, Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM25PT, UK
| | - Shahreen Ahmad
- Guy's Cancer Centre, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London SE19RT UK
| | - Fiona McDonald
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London SW36JJ, UK; Department of Clinical Oncology, Institute of Cancer Research NIHR Biomedical Research Centre, London, UK
| | - Imogen Locke
- Lung Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton SM25PT, UK
| | - Danielle Power
- Department of Clinical Oncology, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK
| | - Matthew Blackledge
- Radiotherapy and Imaging, Institute of Cancer Research, 123 Old Brompton Road, London SW7 3RP, UK
| | - Richard W Lee
- Lung Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London SW36JJ, UK; Early Diagnosis and Detection Centre, National Institute for Health Research (NIHR) Biomedical Research Centre at The Royal Marsden NHS Foundation Trust and the Institute of Cancer Research, London; National Heart and Lung Institute, Imperial College, London, UK.
| | - Eric O Aboagye
- Department of Clinical Oncology, Institute of Cancer Research NIHR Biomedical Research Centre, London, UK.
| |
Collapse
|
13
|
DePhillipo NN, Aman ZS, Dekker TJ, Moatshe G, Chahla J, LaPrade RF. Preventative and Disease-Modifying Investigations for Osteoarthritis Management Are Significantly Under-represented in the Clinical Trial Pipeline: A 2020 Review. Arthroscopy 2021; 37:2627-2639. [PMID: 33812028 DOI: 10.1016/j.arthro.2021.03.050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/10/2021] [Accepted: 03/23/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE To conduct a review of active United States-based clinical trials investigating preventative, symptom resolution, and disease-modifying therapies for osteoarthritis (OA). METHODS We conducted a review of currently active clinical trials for OA using data obtained from the ClinicalTrials.gov database as of August 2020. The inclusion criteria were active studies registered in the United States that involved the prevention, symptom resolution, or disease modification of OA. Descriptive statistics were recorded and summarized. RESULTS A total of 3,859 clinical trials were identified, and 310 were included in the final analysis. Of the currently active trials, 89% (n = 275) targeted symptom resolution in patients with existing OA, 6% (n = 19) targeted OA disease-modifying therapeutics, and 5% (n = 16) targeted the prevention of OA in high-risk patients (P < .001). Primary interventions included medical devices (44%, n = 137), pharmaceutical drugs (14%, n = 42), surgical procedures (14%, n = 42), cellular biologics (13%, n = 41), and behavioral therapies (13%, n = 41). There was a significantly higher number of disease-modifying therapeutics for cellular biologics than pharmaceutical drugs (30% vs 14%) (P = .015). Most trials targeted the knee joint (63%, P = .042), with 38% of all trials evaluating joint arthroplasty. There were no significant differences between private sector and government funding sources (43% and 49%, respectively) (P = .288), yet there was a significantly lower rate of funding from industry (8%) (P = .026). CONCLUSIONS There was a significantly higher number of clinical trials investigating symptomatic resolution therapy (89%) for existing OA in comparison to preventative (5%) and disease-modifying (6%) therapies. The most common interventions involved medical devices and joint replacement surgery, with the knee joint accounting for more than 60% of the current clinical trials for OA. There was a significantly higher number of disease-modifying therapeutics for cellular biologics than pharmaceutical drugs. Funding of clinical trials was split between the private sector and government, with a low rate of reported funding from industry partners. CLINICAL RELEVANCE Identifying existing needs in the current market may help increase rates of research funding or optimize current funding pathways, in this study, specifically for targeting unaddressed focus areas in OA research. Our systematic review highlights the potential need for additional research and development regarding OA preventative and disease-modifying therapies.
Collapse
Affiliation(s)
| | - Zachary S Aman
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, U.S.A
| | | | - Gilbert Moatshe
- Oslo Sports Trauma Research Center, Oslo, Norway; Ulleval University Hospital, Oslo, Norway
| | - Jorge Chahla
- Midwest Orthopedics at Rush, Chicago, Illinois, U.S.A
| | | |
Collapse
|
14
|
Sundrani S, Lu J. Computing the Hazard Ratios Associated With Explanatory Variables Using Machine Learning Models of Survival Data. JCO Clin Cancer Inform 2021; 5:364-378. [PMID: 33797958 DOI: 10.1200/cci.20.00172] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE The application of Cox proportional hazards (CoxPH) models to survival data and the derivation of hazard ratio (HR) are well established. Although nonlinear, tree-based machine learning (ML) models have been developed and applied to the survival analysis, no methodology exists for computing HRs associated with explanatory variables from such models. We describe a novel way to compute HRs from tree-based ML models using the SHapley Additive exPlanation values, which is a locally accurate and consistent methodology to quantify explanatory variables' contribution to predictions. METHODS We used three sets of publicly available survival data consisting of patients with colon, breast, or pan cancer and compared the performance of CoxPH with the state-of-the-art ML model, XGBoost. To compute the HR for explanatory variables from the XGBoost model, the SHapley Additive exPlanation values were exponentiated and the ratio of the means over the two subgroups was calculated. The CI was computed via bootstrapping the training data and generating the ML model 1,000 times. Across the three data sets, we systematically compared HRs for all explanatory variables. Open-source libraries in Python and R were used in the analyses. RESULTS For the colon and breast cancer data sets, the performance of CoxPH and XGBoost was comparable, and we showed good consistency in the computed HRs. In the pan-cancer data set, we showed agreement in most variables but also an opposite finding in two of the explanatory variables between the CoxPH and XGBoost result. Subsequent Kaplan-Meier plots supported the finding of the XGBoost model. CONCLUSION Enabling the derivation of HR from ML models can help to improve the identification of risk factors from complex survival data sets and to enhance the prediction of clinical trial outcomes.
Collapse
Affiliation(s)
- Sameer Sundrani
- Modeling and Simulation/Clinical Pharmacology, Genentech, South San Francisco, CA.,Biomedical Computation, Schools of Engineering and Medicine, Stanford University, Stanford, CA
| | - James Lu
- Modeling and Simulation/Clinical Pharmacology, Genentech, South San Francisco, CA
| |
Collapse
|
15
|
Wang Y, Zou S, Zhao Z, Liu P, Ke C, Xu S. New insights into small-cell lung cancer development and therapy. Cell Biol Int 2020; 44:1564-1576. [PMID: 32281704 PMCID: PMC7496722 DOI: 10.1002/cbin.11359] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/24/2020] [Accepted: 04/11/2020] [Indexed: 12/24/2022]
Abstract
Small‐cell lung cancer (SCLC) accounts for approximately 15% of lung cancer cases; however, it is characterized by easy relapse and low survival rate, leading to one of the most intractable diseases in clinical practice. Despite decades of basic and clinical research, little progress has been made in the management of SCLC. The current standard first‐line regimens of SCLC still remain to be cisplatin or carboplatin combined with etoposide, and the adverse events of chemotherapy are by no means negligible. Besides, the immunotherapy on SCLC is still in an early stage and novel studies are urgently needed. In this review, we describe SCLC development and current therapy, aiming at providing useful advices on basic research and clinical strategy.
Collapse
Affiliation(s)
- Yuwen Wang
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Songyun Zou
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Zhuyun Zhao
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Po Liu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Changneng Ke
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Shi Xu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China.,Division of Respiratory Medicine, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| |
Collapse
|