See Lung Cancer with an AI
Abstract
:Simple Summary
Abstract
1. Introduction
2. Proof of Concept and Important Aspects of Lung Cancer Screening Implementation
2.1. Proof of Concept
2.2. Country-Specific Differences
Country | Status |
---|---|
Albania, Bosnia and Herzegovina, Bulgaria, Cyprus, Iceland, Latvia, Lithuania, Malta, Moldova, Montenegro, North Macedonia, Portugal, Slovenia | Unknown status. No further information available. |
Belarus, Norway, Switzerland | Implementation research. No further information available. |
Austria | Implementation research. The Austrian Society of Pneumology and the Austrian Radiological Society recommended the implementation of LDCT screening and formed a taskforce focused on this in 2018. In February 2021, it announced plans to implement future pilots looking at the feasibility of LDCT screening in Austria. No further information is available. |
Belgium | Implementation research. Belgium participated in a large clinical effectiveness RCT (NELSON), and results were reported in 2020. The decision to implement LCS is governed by the Flemish, Walloon, or Brussels’ Ministers of Health. Funding was recently approved for a pilot in the Flemish region. |
Croatia | Implementation roll-out. Croatia launched a national LDCT LCS in October 2020, as the first country in Europe. It is financed by the Croatian Health Insurance Fund. First term will conclude in 2023 before renewal. |
Czech Republic | Implementation roll-out. In January 2022, a five-year national LCS was initiated by the Czech Ministry of Health. It is fully covered by public health insurance. The anticipated completion is 2026. It is expected to be renewed. |
Denmark | Unknown status. Denmark previously conducted a large clinical effectiveness RCT (the DLCST), which ended in 2015. In January 2021, a proposal was submitted to the Danish National Board of Health (NBH) to introduce a screening pilot in Denmark. Currently, the NBH and Ministry of Health are considering the proposal and possible funding of a pilot as part of a HTA evaluation, which would serve as the first step towards national implementation. |
Estonia | Implementation research. An ongoing regional pilot in Tartu aims to assess the feasibility of introducing a national LCS. The pilot builds on earlier implementation research, including a study conducted by the National Institute for Health Development (TAI). |
Finland | Unknown status. In November 2021, a working group appointed by the Finnish Medical Association Duodecim, the Finnish Lung Doctors’ Association, and the Finnish Society for Oncology reviewed its clinical guidelines for lung cancer. It concluded that further research was required around the optimal eligibility criteria, interval, and cost-effectiveness of LDCT LCS before its implementation. |
France | Implementation research. Pilots ongoing in France, with funding from public and private sources. In February 2022, the High Authority for Health (HAS) announced it will carry out an update to the 2016 evaluation of the evidence for LDCT LCS, and nationally-funded implementation pilots will begin. |
Germany | Implementation research. Several clinical effectiveness trials and economic evaluations have previously taken place in Germany. Two prospective evaluations performed by the Federal Office for Radiation Protection and the Institute for Quality and Efficiency in Health Care (IQWiG) were supportive of LCS in 2020, and this must be approved by The Federal Joint Committee (G-BA). Several implementation trials are ongoing (HANSE and the 4-IN-THE-LUNG-RUN). |
Greece | Unknown status. As of 2021, there was no official recommendation from the Greek Ministry of Health regarding LDCT LCS. The development of a National Lung Cancer Control Strategy which includes early detection was announced. On a private level, there are implementation studies around screening programmes in a select few hospitals in Athens, Thessaloniki, and Crete. |
Hungary | Implementation research. The first phase of a national LDCT lung cancer screening pilot program (HUNCHEST) was completed in 2018. The second phase is currently ongoing and expected to end in 2022. |
Ireland | Unknown status. There are a few small-scale opportunistic screening programmes for lung cancer, but these are exclusive to private hospitals and not endorsed by the NCCP or National Screening Service. |
Italy | Formal commitment. In Italy, the Ministry of Health has committed to a national programme of implementation, with a national pilot study being conducted in several centers. Italy also led several large RCTs investigating the clinical effectiveness of LDCT screening. |
Luxembourg | Unknown status. In 2020, it was reported that preliminary discussions for the implementation of an LDCT LCS had begun with the National healthcare system and National Health Insurance Fund. |
Netherlands | Implementation research. The Netherlands participated in a large clinical effectiveness RCT (NELSON); results were reported in 2020. Recruitment recently began for the Dutch arm of the 4-IN-THE-LUNG-RUN trial, which is due to end in 2024. The government has yet to support the implementation of LDCT screening. |
United Kingdom | Formal commitment. The UK National Screening Committee (UK NSC) recently reviewed its recommendations for lung cancer screening, the outcome of which was announced in September 2022. The UK NSC now recommends LDCT screening for current and former smokers aged 55–74. England and Scotland are currently engaged in implementation research (e.g., LUNGSCOT study and the Targeted Lung Health Check pilot programme). |
Poland | Implementation roll-out. Poland initiated pilot studies for the early detection of lung cancer via LDCT screening in 2008. These pilots were introduced independently in Szczecin, Gdańsk, Poznań, and Warsaw. In 2020, a national programme (the WWRP) began. The three-year centrally administered programme is being implemented. The country is divided in six macroregions with Centers of Excellence and cooperation sites. |
Romania | Formal commitment. The latest NCCP has pledged that the Ministry of Health and the National Institute of Public Health will develop a national pilot programme between 2023 and 2025. The proposed target population is current and former smokers aged 50–80 who will be offered annual LDCT screening. |
Serbia | Implementation research. The first screening pilot programme in Serbia is currently ongoing in the Autonomous Province of Vojvodina (APV). Initially planned as a region-wide pilot, due to the COVID-19 pandemic, it is only taking place in the South Bačka district. |
Slovakia | Implementation research. A Ministry of Health taskforce is currently developing a plan for a national LDCT lung cancer screening programme in Slovakia. |
Spain | Implementation research. There is no national LCS program in Spain. There have been some implementation studies. Currently, there are plans for a national pilot (CASSANDRA) led by the Spanish Society of Pneumology and Thoracic Surgery (SEPAR). This is due to begin in 2022. |
Sweden | Implementation research. A regional cancer center was commissioned to deliver a regional lung cancer screening pilot that targets women in Stockholm. If successful, findings will be used to inform the development of a national organized LDCT screening programme, in which men would also be eligible to participate. |
2.3. Obstacles to Overcome
2.3.1. False Positive Results
2.3.2. Political Level and Cost-Effectiveness
2.4. Hope or Hype from the European Commission?
3. What about Tobacco?
3.1. Horrific Statistics
3.2. Beyond the Tobacco
4. The Need for Immediate Action
- Will this put a strain on healthcare systems with staff and equipment shortages in some countries?
- Could an artificial intelligence application in healthcare solve some of these problems?
5. Artificial Intelligence in Healthcare with a Focus on Oncology
6. Radiomics in Lung Cancer Screening, Diagnostics, and Prognostication
6.1. Basic Concept
6.2. Radiomics in Lung Cancer Screening
- Veye Lung Nodules—CE-certified automated lung nodule management assistant integrated into the radiological workflow. It can be used for automated lung nodule detection and quantification on chest CT scans.
- Veye Reporting—the interactive solution for lung nodule reporting allows for the generation of standardized quality reporting.
6.3. Detection, Diagnosis, Staging
6.4. Prognosis/Prediction
7. Future or Current Directions?
8. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
- European Lung White Book. Available online: https://www.erswhitebook.org/chapters/lung-cancer/ (accessed on 13 January 2023).
- Wilson, J.; Junger, G. Principles and Practice of Screening for Disease. Geneva: World Health Organization. 1968. Available online: https://apps.who.int/iris/bitstream/handle/10665/37650/WHO_PHP_34.pdf?sequence=17 (accessed on 10 December 2022).
- Strengthening Europe in the Fight against Cancer. Policy Department for Economic, Scientific and Quality of Life Policies Directorate-General for Internal Policies. Authors: Norbert COUESPEL et al. PE 642.388–2020. Available online: https://www.europarl.europa.eu/RegData/etudes/STUD/2020/642388/IPOL_STU(2020)642388_EN.pdf (accessed on 10 December 2022).
- Cancer Control: Prevention. WHO Guide for Effective Programmes. Geneva: World Health Organization. 2007. Available online: https://www.google.com/url?sa=t&rct=j&q=&esrc=s&source=web&cd=&ved=2ahUKEwj4i8WHisL8AhVtl4sKHVbyCGoQFnoECA8QAQ&url=https%3A%2F%2Fapps.who.int%2Firis%2Fbitstream%2F10665%2F43467%2F1%2F9241546999_eng.pdf&usg=AOvVaw0Bv_jde4J66kapZgBmQwcE (accessed on 10 December 2022).
- Tindle, H.A.; Duncan, M.S.; Greevy, R.A.; Vasan, R.S.; Kundu, S.; Massion, P.P.; Freiberg, M.S. Lifetime Smoking History and Risk of Lung Cancer: Results from the Framingham Heart Study. J. Natl. Cancer Inst. 2018, 110, 1201–1207. [Google Scholar] [CrossRef]
- The National Lung Screening Trial Research Team. Reduced Lung-Cancer Mortality with Low-Dose Computed Tomographic Screening. N. Engl. J. Med. 2011, 365, 395–409. [Google Scholar] [CrossRef] [Green Version]
- Pastorino, U.; Silva, M.; Sestini, S.; Sabia, S.; Boeri, M.; Cantarutti, A.; Sverzellati, N.; Sozzi, G.; Marchiano, A. Prolonged lung cancer screening reduced 10-year mortality in the MILD trial: New confirmation of lung cancer screening efficacy. Ann. Oncol. 2019, 30, 1162–1169. [Google Scholar] [CrossRef]
- Becker, N.; Motsch, E.; Trotter, A.; Claus, P.H.; Dienemann, H.; Schnabel, P.A.; Kauczor, H.U.; González Maldonado, S.; Miller, A.B.; Kaaks, R.; et al. Lung cancer mortality reduction by LDCT screening-Results from the randomized German LUSI trial. Int. J. Cancer 2020, 146, 1503–1513. [Google Scholar] [CrossRef]
- De Koning, H.J.; van der Aalst, C.M.; de Jong, P.A.; Scholten, E.T.; Nackaerts, K.; Heuvelmans, M.A.; Lammers, J.-W.J.; Weenink, C.; Yousaf-Khan, U.; Horeweg, N.; et al. Reduced Lung-Cancer Mortality with Volume CT Screening in a Randomized Trial. N. Engl. J. Med. 2020, 382, 503–513. [Google Scholar] [CrossRef]
- Sadate, A.; Occean, B.V.; Beregi, J.P.; Hamard, A.; Addala, T.; de Forges, H.; Fabbro-Peray, P.; Frandon, J. Systematic review and meta-analysis on the impact of lung cancer screening by low-dose computed tomography. Eur. J. Cancer 2020, 134, 107–114. [Google Scholar] [CrossRef]
- New Medicare Lung Cancer Screening Guidelines a Huge Step Forward–More Lives to be Saved. Available online: https://www.acr.org/Media-Center/ACR-News-Releases/2022/New-Medicare-Lung-Cancer-Screening-Guidelines-a-Huge-Step-Forward (accessed on 10 December 2022).
- Lung cancer in urban China. Available online: http://www.cancercontrol.info/wp-content/uploads/2015/07/87-93-Qiao_cc2015.pdf (accessed on 10 December 2022).
- Financing Lung Cancer Screening in China Financial Innovations Lab® Report. Available online: https://milkeninstitute.org/sites/default/files/reports-pdf/No-3900-LungCancerScreeningChina-MR19-WEB_2.pdf (accessed on 10 December 2022).
- Zhang, Y.; Chen, H. Lung cancer screening: Who pays? who receives?—The Chinese perspective. Transl. Lung Cancer Res. 2021, 10, 2389–2394. [Google Scholar] [CrossRef]
- Van Meerbeeck, J.P.; Franck, C. Lung cancer screening in Europe: Where are we in 2021? Transl. Lung Cancer Res. 2021, 10, 2407–2417. [Google Scholar] [CrossRef]
- UK National Screening Committee. Available online: https://view-health-screening-recommendations.service.gov.uk/lung-cancer/ (accessed on 10 December 2022).
- The Lung Cancer Policy Network. Interactive Map of Lung Cancer Screening Database (First Edition). 2022. Available online: https://www.lungcancerpolicynetwork.com/interactive-map/ (accessed on 15 January 2023).
- Hammer, M.M.; Byrne, S.C.; Kong, C.Y. Factors Influencing the False Positive Rate in CT Lung Cancer Screening. Acad. Radiol. 2022, 29 (Suppl. S2), S18–S22. [Google Scholar] [CrossRef]
- Rzyman, W.; Szurowska, E.; Adamek, M. Implementation of lung cancer screening at the national level: Polish example. Transl. Lung Cancer Res. 2019, 8, S95–S105. [Google Scholar] [CrossRef]
- Dziadziuszko, K.; Szurowska, E. Pulmonary nodule radiological diagnostic algorithm in lung cancer screening. Transl. Lung Cancer Res. 2021, 10, 1124–1135. [Google Scholar] [CrossRef]
- Ten Haaf, K.; Tammemagi, M.C.; Bondy, S.J.; van der Aalst, C.M.; Gu, S.; McGregor, S.E.; Nicholas, G.; de Koning, H.J.; Paszat, L.F. Performance and cost-effectiveness of computed tomography lung cancer screening scenarios in a population-based setting: A microsimulation modeling analysis in Ontario, Canada. PLoS Med. 2017, 14, e1002225. [Google Scholar] [CrossRef]
- Goffin, J.R.; Flanagan, W.M.; Miller, A.B.; Fitzgerald, N.R.; Memon, S.; Wolfson, M.C.; Evans, W.K. Costeffectiveness of Lung Cancer Screening in Canada. JAMA Oncol. 2015, 1, 807–813. [Google Scholar] [CrossRef]
- Pyenson, B.S.; Sander, M.S.; Jiang, Y.; Kahn, H.; Mulshine, J.L. An actuarial analysis shows that offering lung cancer screening as an insurance benefit would save lives at relatively low cost. Health Aff. 2012, 31, 770–779. [Google Scholar] [CrossRef]
- Villanti, A.C.; Jiang, Y.; Abrams, D.B.; Pyenson, B.S. A cost-utility analysis of lung cancer screening and the additional benefits of incorporating smoking cessation interventions. PLoS ONE 2013, 8, e71379. [Google Scholar] [CrossRef] [Green Version]
- Field, J.K.; Duffy, S.W.; Baldwin, D.R.; Brain, K.E.; Devaraj, A.; Eisen, T.; Green, B.A.; Holemans, J.A.; Kavanagh, T.; Kerr, K.M.; et al. The UK Lung Cancer Screening Trial: A pilot randomised controlled trial of low-dose computed tomography screening for the early detection of lung cancer. Health Technol. Assess. 2016, 20, 1–146. [Google Scholar] [CrossRef] [Green Version]
- Field, J.K.; Mulshine, J.L. Lung Cancer Risk Reduction & Prevention. Available online: https://www.ilcn.org/lung-cancer-screening-is-cost-effective/ (accessed on 10 December 2022).
- Du, Y.; Sidorenkov, G.; Heuvelmans, M.A.; Groen, H.J.M.; Vermeulen, K.M.; Greuter, M.J.W.; de Bock, G.H. Cost-effectiveness of lung cancer screening with low-dose computed tomography in heavy smokers: A microsimulation modelling study. Eur. J. Cancer 2020, 135, 121–129. [Google Scholar] [CrossRef]
- Vergnenègre, A.; Chouaid, C. Review of economic analyses of treatment for non-small-cell lung cancer (NSCLC). Expert Rev. Pharm. Outcomes Res. 2018, 18, 519–528. [Google Scholar] [CrossRef]
- Sivignon, M.; Monnier, R.; Tehard, B.; Roze, S. Cost-effectiveness of alectinib compared to crizotinib for the treatment of first-line ALK+ advanced non-small-cell lung cancer in France. PLoS ONE 2020, 15, e0226196. [Google Scholar] [CrossRef] [Green Version]
- Oudkerk, M.; Devaraj, A.; Vliegenthart, R.; Henzler, T.; Prosch, H.; Heussel, C.P.; Bastarrika, G.; Sverzellati, N.; Mascalchi, M.; Delorme, S.; et al. European position statement on lung cancer screening. Lancet Oncol. 2017, 8, e754–e766. [Google Scholar] [CrossRef]
- Rapporteur: Véronique Trillet-Lenoir. REPORT on strengthening Europe in the Fight against Cancer–Towards a Comprehensive and Coordinated Strategy (2020/2267(INI)), Special Committee on Beating Cancer Plenary sitting, European Parliament 2019–2024, A9-9999/2021. Available online: https://www.europarl.europa.eu/cmsdata/243836/BECA%20final%20report_tabled_PE693752v02-00EN.pdf (accessed on 10 December 2022).
- Europe’s Beating Cancer Plan: New Actions to Increase access to Cancer Prevention, Early Detection, Treatment, and Care. Brussels. 2022. Available online: https://ec.europa.eu/commission/presscorner/detail/en/ip_22_702 (accessed on 10 December 2022).
- Cancer Plan Europe. Available online: https://ec.europa.eu/info/strategy/priorities-2019-2024/promoting-our-european-way-life/european-health-union/cancer-plan-europe_en (accessed on 10 December 2022).
- Improving cancer screening in the European Union. 2022. Available online: https://sapea.info/topic/cancer-screening (accessed on 10 December 2022).
- Proposal for a Council Recommendation on Strengthening Prevention through Early Detection: A New EU Approach on Cancer Screening Replacing Council Recommendation 2003/878/EC {SWD(2022) 296 final}, Brussels, 20.9.2022 COM(2022) 474 Final 2022/0290 (NLE). Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/HTML/?uri=COM:2022:474:FIN (accessed on 10 December 2022).
- Lung Cancer Policy Network. Available online: https://www.healthpolicypartnership.com/project/lung-cancer-policy-network/ (accessed on 10 December 2022).
- Lung Cancer Policy Network. Available online: https://www.lungcancerpolicynetwork.com/network-resources/ (accessed on 10 December 2022).
- Proposed Amendments to the EU Commission draft Recommendation on Cancer Screening Submission from The Lung Cancer Policy Network. Available online: https://www.lungcancerpolicynetwork.com/app/uploads/Proposed-amendments-to-the-EU-Commission-draft-recommendation-on-cancer-screening.pdf (accessed on 10 December 2022).
- Palenie Tytoniu w Europie. Available online: https://recigar.pl/palenie-tytoniu/#W_ktorym_kraju_europejskim_pali_sie_najwiecej_papierosow_dziennie (accessed on 10 December 2022).
- Tobbaco Fact Sheets. Available online: https://www.who.int/news-room/fact-sheets/detail/tobacco (accessed on 10 December 2022).
- Lourenco, J.; Clark, O. Clinical Radiology Census Report, the Royal College of Radiologist. Available online: https://www.rcr.ac.uk/sites/default/files/clinical_radiology_census_report_2021.pdf (accessed on 10 December 2022).
- Choi, R.Y.; Coyner, A.S.; Kalpathy-Cramer, J.; Chiang, M.F.; Campbell, J.P. Introduction to Machine Learning, Neural Networks, and Deep Learning. Transl. Vis. Sci. Technol. 2020, 9, 14. [Google Scholar] [CrossRef]
- Binczyk, F.; Prazuch, W.; Bozek, P.; Polanska, J. Radiomics and artificial intelligence in lung cancer screening. Transl. Lung Cancer Res. 2021, 10, 1186–1199. [Google Scholar] [CrossRef]
- Liability Rules for Artificial Intelligence. Available online: https://ec.europa.eu/info/business-economy-euro/doing-business-eu/contract-rules/digital-contracts/liability-rules-artificial-intelligence_en (accessed on 10 December 2022).
- Report from the Commission to the European Parliament, the Council and the European Economic and Social Committee Report on the Safety and Liability Implications of Artificial Intelligence, the Internet of Things and Robotics. COM/2020/64 Final. Brussels, 19.2.2020 COM (2020) 64 Final Document 52020DC0064. Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A52020DC0064 (accessed on 10 December 2022).
- P9_TA (2020)0276 Civil Liability Regime for Artificial Intelligence. European Parliament Resolution of 20 October 2020 with Recommendations to the Commission on a Civil Liability Regime for Artificial Intelligence (2020/2014(INL)), (2021/C 404/05). Available online: https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX:52020IP0276 (accessed on 10 December 2022).
- Cancer Imaging Program, National Cancer Institute. Available online: https://imaging.cancer.gov/default.htm (accessed on 10 December 2022).
- Armato, S.G.; McLennan, G.; Bidaut, L.; McNitt-Gray, M.F.; Meyer, C.R.; Reeves, A.P.; Zhao, B.; Aberle, D.R.; Henschke, C.I.; Hoffman, E.A.; et al. The Lung Image Database Consortium (LIDC) and Image Database Resource Initiative (IDRI): A completed reference database of lung nodules on CT scans. Med. Phys. 2011, 38, 915–931. [Google Scholar] [CrossRef] [Green Version]
- The Cancer Genome Atlas (TCGA) was a joint effort of the National Cancer Institute (NCI) and the National Human Genome Research Institute (NHGRI), Which Are Both Part of the National Institutes of Health, U.S. Department of Health and Human Services. Available online: https://www.genome.gov/Funded-Programs-Projects/Cancer-Genome-Atlas (accessed on 10 December 2022).
- Kann, B.H.; Hosny, A.; Aerts, H.J.W.L. Artificial intelligence for clinical oncology. Cancer Cell. 2021, 39, 916–927. [Google Scholar] [CrossRef]
- Chen, T.; Li, X.; Mao, Q.; Wang, Y.; Li, H.; Wang, C.; Shen, Y.; Guo, E.; He, Q.; Tian, J.; et al. An artificial intelligence method to assess the tumor microenvironment with treatment outcomes for gastric cancer patients after gastrectomy. J. Transl. Med. 2022, 20, 100. [Google Scholar] [CrossRef]
- Mohammadi, A.; Mirza-Aghazadeh-Attari, M.; Faeghi, F.; Homayoun, H.; Abolghasemi, J.; Vogl, T.J.; Bureau, N.J.; Bakhshandeh, M.; Acharya, R.U.; Ardakani, A.A. Tumor Microenvironment, Radiology, and Artificial Intelligence: Should We Consider Tumor Periphery? J. Ultrasound Med. 2022, 41, 2919–3165. [Google Scholar] [CrossRef]
- Adams, S.J.; Stone, E.; Baldwin, D.R.; Vliegenthart, R.; Lee, P.; Fintelmann, F.J. Lung cancer screening. Lancet 2022, 401, 390–408. [Google Scholar] [CrossRef]
- Luchini, C.; Pea, A.; Scarpa, A. Artificial intelligence in oncology: Current applications and future perspectives. Br. J. Cancer. 2022, 126, 4–9. [Google Scholar] [CrossRef]
- Lambin, P.; Rios-Velazquez, E.; Leijenaar, R.; Carvalho, S.; van Stiphout, R.G.; Granton, P.; Zegers, C.M.; Gillies, R.; Boellard, R.; Dekker, A.; et al. Radiomics: Extracting more information from medical images using advanced feature analysis. Eur. J. Cancer 2012, 48, 441–446. [Google Scholar] [CrossRef] [Green Version]
- Radiomics: Decoding the Tumor Phenotype with Non-Invasive Imaging ALPHA GRID. Available online: https://www.youtube.com/watch?v=Tq980GEVP0Y (accessed on 10 December 2022).
- Liu, Z.; Wang, S.; Dong, D.; Wei, J.; Fang, C.; Zhou, X.; Sun, K.; Li, L.; Li, B.; Wang, M.; et al. The Applications of Radiomics in Precision Diagnosis and Treatment of Oncology: Opportunities and Challenges. Theranostics 2019, 9, 1303–1322. [Google Scholar] [CrossRef]
- Aerts, H.J.W.L. The Potential of Radiomic-Based Phenotyping in Precision Medicine: A Review. JAMA Oncol. 2016, 1, 1636–1642. [Google Scholar] [CrossRef]
- Yip, S.S.F.; Aerts, H.J.W.L. Applications and limitations of radiomics. Phys. Med. Biol. 2016, 61, 150–166. [Google Scholar] [CrossRef] [Green Version]
- Fornacon-Wooda, I.; Faivre-Finna, C.; O’Connora, J.P.B.; Pricea, G.J. Radiomics as a personalized medicine tool in lung cancer: Separating the hope from the hype. Lung Cancer 2020, 146, 197–208. [Google Scholar] [CrossRef]
- Lu, L.; Ehmke, R.C.; Schwartz, L.H.; Zhao, B. Assessing agreement between radiomic features computed for multiple CT imaging settings. PLoS ONE 2016, 11, e0166550. [Google Scholar] [CrossRef] [Green Version]
- Varghese, B.A.; Cen, S.Y.; Hwang, D.H.; Duddalwar, V.A. Texture Analysis of Imaging Radiologists Need to Know Cardiopulmonar Imaging. Am. J. Roentgenol. 2019, 212, 520–528. [Google Scholar] [CrossRef]
- Ger, R.B.; Zhou, S.; Chi, P.C.M.; Lee, H.J.; Layman, R.R.; Jones, A.K.; Goff, D.L.; Fuller, C.D.; Howell, R.M.; Li, H.; et al. Comprehensive investigation on controlling or CT imaging variabilities in radiomics studies. Sci. Rep. 2018, 8, 13047. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.-J.; Wu, F.-Z.; Yang, S.-C.; Tang, E.-K.; Liang, C.-H. Radiomics in Early Lung Cancer Diagnosis: From Diagnosis to Clinical Decision Support and Education. Diagnostics 2022, 12, 1064. [Google Scholar] [CrossRef]
- Calhoun, M.E.; Hofmanninger, J.; Wood, C.; Langs, G.; Makropoulos, A. Combining automated malignancy risk estimation with lung nodule detection may reduce physician effort and increase diagnostic accuracy. J. Thorac. Oncol. 2022, 17, S523. [Google Scholar] [CrossRef]
- AI-Rad Companion. Available online: https://www.siemens-healthineers.com/digital-health-solutions/digital-solutions-overview/clinical-decision-support/ai-rad-companion (accessed on 19 January 2023).
- AI-Rad Companion. Available online: https://grand-challenge.org/aiforradiology/product/siemens-rad-companion-chest-ct/ (accessed on 19 January 2023).
- Aidence. Available online: https://www.aidence.com (accessed on 19 January 2023).
- Aidence. AI in Lung Cancer. Papers from BJR. Available online: https://www.bir.org.uk/media/477315/lung_cancer_and_ai_final_updated_v2_150622.pdf (accessed on 19 January 2023).
- Radnet. Available online: https://www.radnet.com (accessed on 19 January 2023).
- Pulmonary Nodules QC. Available online: https://grand-challenge.org/aiforradiology/product/aidoc-pulmonary-nodules/ (accessed on 19 January 2023).
- Aidoc. Available online: https://www.aidoc.com/radiology-ai/ (accessed on 19 January 2023).
- Veolity MeVis. Available online: https://grand-challenge.org/aiforradiology/product/mevis-veolity/ (accessed on 19 January 2023).
- Veolity. Available online: https://www.veolity.com (accessed on 19 January 2023).
- InferRead CT Lung. Available online: https://grand-challenge.org/aiforradiology/product/infervision-ct-lung/ (accessed on 19 January 2023).
- InferVision. Available online: https://global.infervision.com (accessed on 19 January 2023).
- JLD-01K. Available online: https://grand-challenge.org/aiforradiology/product/jlk-inc-jld-01k/ (accessed on 19 January 2023).
- Lancaster, H.L.; Zheng, S.; Aleshina, O.O.; Yu, D.; Chernina, V.; Heuvelmans, M.A.; de Bock, G.H.; Dorrius, M.D.; Willem Gratama, J.; Morozov, S.P.; et al. Outstanding negative prediction performance of solid pulmonary nodule volume AI for ultra-LDCT baseline lung cancer screening risk stratification. Lung Cancer 2022, 165, 133–140. [Google Scholar] [CrossRef]
- Hwang, E.J.; Goo, J.M.; Kim, H.Y.; Yoon, S.H.; Jin, G.Y.; Yi, J.; Kim, Y. Variability in interpretation of low-dose chest CT using computerized assessment in a nationwide lung cancer screening program: Comparison of prospective reading at individual institutions and retrospective central reading. Eur. Radiol. 2021, 31, 2845–2855. [Google Scholar] [CrossRef]
- Hwang, E.J.; Goo, J.M.; Kim, H.Y.; Yoon, S.H.; Jin, G.Y.; Yi, J.; Kim, Y. Implementation of the cloud-based computerized interpretation system in a nationwide lung cancer screening with low-dose CT: Comparison with the conventional reading system. Eur. Radiol. 2021, 31, 475–485. [Google Scholar] [CrossRef]
- Aview LCS+. Available online: https://grand-challenge.org/aiforradiology/product/coreline-soft-aview-lcs/ (accessed on 19 January 2023).
- Aview LCS. Available online: https://www.corelinesoft.com/en/lcs/ (accessed on 19 January 2023).
- Contexflow Demo. Available online: https://calendly.com/contextflow-js/contextflow-demo?month=2023-02 (accessed on 19 January 2023).
- Forte, G.C.; Altmayer, S.; Silva, R.F.; Stefani, M.T.; Libermann, L.L.; Cavion, C.C.; Youssef, A.; Forghani, R.; King, J.; Mohamed, T.-L.; et al. Deep Learning Algorithms for Diagnosis of Lung Cancer: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 3856. [Google Scholar] [CrossRef]
- McNitt-Gray, M.F.; Hart, E.M.; Wyckoff, N.; Sayre, J.W.; Goldin, J.G.; Aberle, D.R. A pattern classification approach to characterizing solitary pulmonary nodules imaged on high resolution CT: Preliminary results. Med. Phys. 1999, 26, 880–888. [Google Scholar] [CrossRef]
- Brunetti, A.; Altini, N.; Buongiorno, D.; Garolla, E.; Corallo, F.; Gravina, M.; Bevilacqua, V.; Prencipe, B. A Machine Learning and Radiomics Approach in Lung Cancer for Predicting Histological Subtype. Appl. Sci. 2022, 12, 5829. [Google Scholar] [CrossRef]
- Naqi, S.M.; Sharif, M.; Yasmin, M.; Fernandes, S.L. Lung nodule detection using polygon approximation and hybrid features from CT images. Curr. Med. Imaging 2018, 14, 108–117. [Google Scholar] [CrossRef]
- Huidrom, R.; Chanu, Y.J.; Singh, K.M. Pulmonary nodule detection on computed tomography using the neuro-evolutionary scheme. Signal Image Video Process. 2019, 13, 53–60. [Google Scholar] [CrossRef]
- Gong, J.; Liu, J.Y.; Wang, L.J.; Sun, X.W.; Zheng, B.; Nie, S.D. Automatic detection of pulmonary nodules in CT images by incorporating 3D tensor filtering with local image feature analysis. Phys. Med. 2018, 46, 124–133. [Google Scholar] [CrossRef]
- Sun, L.; Wang, Z.; Pu, H.; Yuan, G.; Guo, L.; Pu, T.; Peng, Z. Spectral analysis for pulmonary nodule detection using the optimal fractional S-Transform. Comput. Biol. Med. 2020, 119, 103675. [Google Scholar] [CrossRef]
- Zhao, Y.; Wang, Z.; Liu, X.; Chen, Q.; Li, C.; Zhao, H.; Wang, Z. Pulmonary Nodule Detection Based on Multiscale Feature Fusion. Comput. Math. Methods Med. 2022, 2022, 8903037. [Google Scholar] [CrossRef]
- Lin, J.; She, Q.; Chen, Y. Pulmonary nodule detection based on IR-UNet + + . Med. Biol. Eng. Comput. 2023, 61, 485–495. [Google Scholar] [CrossRef]
- Sethy, P.K.; Geetha Devi, A.; Padhan, B.; Behera, S.K.; Sreedhar, S.; Das, K. Lung cancer histopathological image classification using wavelets and AlexNet. J. Xray Sci. Technol. 2023, 31, 211–221. [Google Scholar] [CrossRef]
- Gugulothu, V.K.; Balaji, S. An automatic classification of pulmonary nodules for lung cancer diagnosis using novel LLXcepNN classifier. J. Cancer Res. Clin. Oncol. 2023, 1–9. [Google Scholar] [CrossRef]
- Thattaamuriyil Padmakumari, L.; Guido, G.; Caruso, D.; Nacci, I.; Del Gaudio, A.; Zerunian, M.; Polici, M.; Gopalakrishnan, R.; Sayed Mohamed, A.K.; De Santis, D.; et al. The Role of Chest CT Radiomics in Diagnosis of Lung Cancer or Tuberculosis: A Pilot Study. Diagnostics 2022, 12, 739. [Google Scholar] [CrossRef]
- Chang, R.; Qi, S.; Zuo, Y.; Yue, Y.; Zhang, X.; Guan, Y.; Qian, W. Predicting chemotherapy response in non-small-cell lung cancer via computed tomography radiomic features: Peritumoral, intratumoral, or combined? Front. Oncol. 2022, 12, 915835. [Google Scholar] [CrossRef]
- Aerts, H.J.; Velazquez, E.R.; Leijenaar, R.T.; Parmar, C.; Grossmann, P.; Carvalho, S.; Bussink, J.; Monshouwer, R.; Haibe-Kains, B.; Rietveld, D.; et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat. Commun. 2014, 5, 4006, Erratum in: Nat. Commun. 2014, 5, 4644. [Google Scholar] [CrossRef] [Green Version]
- Ferreira Junior, J.R.; Koenigkam-Santos, M.; Cipriano, F.E.G.; Fabro, A.T.; Azevedo-Marques, P.M. Radiomics-based features for pattern recognition of lung cancer histopathology and metastases. Comput. Methods Programs Biomed. 2018, 159, 23–30. [Google Scholar] [CrossRef]
- Ardila, D.; Kiraly, A.P.; Bharadwaj, S.; Choi, B.; Reicher, J.J.; Peng, L.; Tse, D.; Etemadi, M.; Ye, W.; Corrado, G.; et al. End-to-end lung cancer screening with three-dimensional deep learning on low-dose chest computed tomography. Nat. Med. 2019, 25, 954–961. [Google Scholar] [CrossRef]
- Zhou, Y.; Dong, Y.; Sun, Q.; Fang, C. Diagnosis and prognosis of non-small cell lung cancer based on machine learning algorithms. Comb. Chem. High Throughput Screen. 2023. [Google Scholar] [CrossRef]
- Lu, C.F.; Liao, C.Y.; Chao, H.S.; Chiu, H.Y.; Wang, T.W.; Lee, Y.; Chen, J.R.; Shiao, T.H.; Chen, Y.M.; Wu, Y.T. A radiomics-based deep learning approach to predict progression free-survival after tyrosine kinase inhibitor therapy in non-small cell lung cancer. Cancer Imaging 2023, 23, 9. [Google Scholar] [CrossRef]
- Shimada, Y.; Kudo, Y.; Maehara, S.; Fukuta, K.; Masuno, R.; Park, J.; Ikeda, N. Artificial intelligence-based radiomics for the prediction of nodal metastasis in early-stage lung cancer. Sci. Rep. 2023, 13, 1028. [Google Scholar] [CrossRef]
- Terada, Y.; Isaka, M.; Kawata, T.; Mizuno, K.; Muramatsu, K.; Katsumata, S.; Konno, H.; Nagata, T.; Mizuno, T.; Serizawa, M.; et al. The efficacy of a machine learning algorithm for assessing tumour components as a prognostic marker of surgically resected stage IA lung adenocarcinoma. Jpn. J. Clin Oncol. 2023, 53, 161–167. [Google Scholar] [CrossRef]
- Pan, Z.; Zhang, R.; Shen, S.; Lin, Y.; Zhang, L.; Wang, X.; Ye, Q.; Wang, X.; Chen, J.; Zhao, Y.; et al. OWL: An optimized and independently validated machine learning prediction model for lung cancer screening based on the UK Biobank, PLCO, and NLST populations. EBioMedicine 2023, 88, 104443. [Google Scholar] [CrossRef]
- Onozato, Y.; Iwata, T.; Uematsu, Y.; Shimizu, D.; Yamamoto, T.; Matsui, Y.; Ogawa, K.; Kuyama, J.; Sakairi, Y.; Kawakami, E.; et al. Predicting pathological highly invasive lung cancer from preoperative [18F]FDG PET/CT with multiple machine learning models. Eur. J. Nucl. Med. Mol. Imaging 2023, 50, 715–726. [Google Scholar] [CrossRef]
- Ge, G.; Zhang, J. Feature selection methods and predictive models in CT lung cancer radiomics. J. Appl. Clin. Med. Phys. 2023, 24, e13869. [Google Scholar] [CrossRef]
- Klang, E. Deep learning and medical imaging. J. Thorac. Dis. 2018, 10, 1325–1328. [Google Scholar] [CrossRef]
- Lawson, C.E.; Marti, J.M.; Radivojevic, T.; Jonnalagadda, S.V.R.; Gentz, R.; Hillson, N.J.; Peisert, S.; Kim, J.; Simmons, B.A.; Petzold, C.J.; et al. Machine learning for metabolic engineering: A review. Metab. Eng. 2021, 63, 34–60. [Google Scholar] [CrossRef]
- Šarić, M.; Russo, M.; Stella, M.; Sikora, M. CNN-based method for lung cancer detection in whole slide histopathology images. In Proceedings of the 2019 4th International Conference on Smart and Sustainable Technologies (SpliTech), Split, Croatia, 18–21 June 2019; pp. 1–4. [Google Scholar]
- Aprupe, L.; Litjens, G.; Brinker, T.J.; van der Laak, J.; Grabe, N. Robust and accurate quantification of biomarkers of immune cells in lung cancer micro-environment using deep convolutional neural networks. Peer J. 2019, 7, e6335. [Google Scholar] [CrossRef]
- Evans, A.J.; Bauer, T.W.; Bui, M.M.; Cornish, T.C.; Duncan, H.; Glassy, E.F.; Hipp, J.; McGee, R.S.; Murphy, D.; Myers, C. US Food and Drug Administration approval of whole slide imaging for primary diagnosis: A key milestone is reached, and new questions are raised. Arch. Pathol. Lab. Med. 2018, 142, 1383–1387. [Google Scholar] [CrossRef] [Green Version]
- Chiu, H.-Y.; Chao, H.-S.; Chen, Y.-M. Application of Artificial Intelligence in Lung Cancer. Cancers 2022, 14, 1370. [Google Scholar] [CrossRef]
- Wei, J.W.; Tafe, L.J.; Linnik, Y.A.; Vaickus, L.J.; Tomita, N.; Hassanpour, S. Pathologist-level classification of histologic patterns on resected lung adenocarcinoma slides with deep neural networks. Sci. Rep. 2019, 9, 3358. [Google Scholar] [CrossRef] [Green Version]
- Sha, L.; Osinski, B.L.; Ho, I.Y.; Tan, T.L.; Willis, C.; Weiss, H.; Beaubier, N.; Mahon, B.M.; Taxter, T.J.; Yip, S.S. Multi-field-of-view deep learning model predicts nonsmall cell lung cancer programmed death-ligand 1 status from whole-slide hematoxylin and eosin images. J. Pathol. Inform. 2019, 10, 24. [Google Scholar] [CrossRef]
- Hondelink, L.M.; Hüyük, M.; Postmus, P.E.; Smit, V.T.; Blom, S.; von der Thüsen, J.H.; Cohen, D. Development and validation of a supervised deep learning algorithm for automated whole-slide programmed death-ligand 1 tumour proportion score assessment in non-small cell lung cancer. Histopathology 2021, 80, 635–647. [Google Scholar] [CrossRef]
- Wu, J.; Liu, C.; Liu, X.; Sun, W.; Li, L.; Gao, N.; Zhang, Y.; Yang, X.; Zhang, J.; Wang, H.; et al. Artificial intelligence-assisted system for precision diagnosis of PD-L1 expression in non-small cell lung cancer. Mod. Pathol. 2022, 35, 403–411. [Google Scholar] [CrossRef]
- Viswanathan, V.S.; Toro, P.; Corredor, G.; Mukhopadhyay, S.; Madabhushi, A. The state of the art for artificial intelligence in lung digital pathology. J. Pathol. 2022, 257, 413–429. [Google Scholar] [CrossRef]
- Xie, X.; Fu, C.C.; Lv, L.; Ye, Q.; Yu, Y.; Fang, Q.; Zhang, L.; Hou, L.; Wu, C. Deep convolutional neural network-based classification of cancer cells on cytological pleural effusion images. Mod. Pathol. 2022, 35, 609–614. [Google Scholar] [CrossRef]
- Dilbar, A.; Qin, H.; Yen-Cheng, C.; Chi-Cheng, F.; Wei, Y.; Lei, L.; Dexian, Y.; Chun, L.; Maosong, Y.; Yong, Z.; et al. Artificial intelligence-based rapid on-site cytopathological evaluation for bronchoscopy examinations. Intell. Based Med. 2022, 6, 100069. [Google Scholar] [CrossRef]
- Wang, S.; Shi, J.; Ye, Z.; Dong, D.; Yu, D.; Zhou, M.; Liu, Y.; Gevaert, O.; Wang, K.; Zhu, Y. Predicting EGFR mutation status in lung adenocarcinoma on computed tomography image using deep learning. Eur. Respir. J. 2019, 53, 1800986. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Ding, C.; Zhang, H.; Song, J.; Wu, L. Radiomics for the prediction of EGFR mutation subtypes in non-small cell lung cancer. Med. Phys. 2019, 46, 4545–4552. [Google Scholar] [CrossRef]
- Echle, A.; Rindtorff, N.T.; Brinker, T.J.; Luedde, T.; Pearson, A.T.; Kather, J.N. Deep learning in cancer pathology: A new generation of clinical biomarkers. Br. J. Cancer 2021, 124, 686–696. [Google Scholar] [CrossRef]
- Predicting Response to Immunotherapy Using Computer Extracted Featuresof Cancer Nuclei from Hematoxylin and Eosin (H&E) Stained Images of Non-Small Cell Lung Cancer (NSCLC). Available online: https://patents.google.com/patent/US11055844B2/en (accessed on 19 January 2023).
- Qiu, Z.B.; Zhang, C.; Chu, X.P.; Cai, F.Y.; Yang, X.N.; Wu, Y.L.; Zhong, W.Z. Quantifying invasiveness of clinical stage IA lung adenocarcinoma with computed tomography texture features. J. Thorac. Cardiovasc. Surg. 2022, 163, 805–815.e3. [Google Scholar] [CrossRef]
- Jones, G.D.; Brandt, W.S.; Shen, R.; Sanchez-Vega, F.; Tan, K.S.; Martin, A.; Zhou, J.; Berger, M.; Solit, D.B.; Schultz, N. A genomic-pathologic annotated risk model to predict recurrence in early-stage lung adenocarcinoma. JAMA Surg. 2021, 156, e205601. [Google Scholar] [CrossRef]
- Lewis, J.E.; Kemp, M.L. Integration of machine learning and genome-scale metabolic modeling identifies multi-omics biomarkers for radiation resistance. Nat. Commun. 2021, 12, 2700. [Google Scholar] [CrossRef]
- EUCANIMAGE. Available online: https://eucanimage.eu (accessed on 19 January 2023).
- EUCAIM. Available online: https://www.eibir.org/projects/eucaim/ (accessed on 19 January 2023).
- Adamek, M.; Biernat, W.; Chorostowska-Wynimko, J.; Didkowska, J.A.; Dziadziuszko, K.; Grodzki, T.; Jassem, J.; Kępka, L.; Kowalski, D.; Krawczyk, P.; et al. Lung Cancer in Poland. J. Thorac. Oncol. 2020, 15, 1271–1276. [Google Scholar] [CrossRef]
- Ostrowski, M.; Bińczyk, F.; Marjański, T.; Dziedzic, R.; Pisiak, S.; Małgorzewicz, S.; Adamek, M.; Polańska, J.; Rzyman, W. Performance of various risk prediction models in a large lung cancer screening cohort in Gdańsk, Poland-a comparative study. Transl. Lung Cancer Res. 2021, 10, 1083–1090. [Google Scholar] [CrossRef]
- Widłak, P.; Jelonek, K.; Kurczyk, A.; Zyla, J.; Sitkiewicz, M.; Bottoni, E.; Veronesi, G.; Polanska, J.; Rzyman, W. Serum metabolite profiles in participants of lung cancer screening study; comparison of two independent cohorts. Cancers 2021, 13, 2714. [Google Scholar] [CrossRef]
- Smolarz, M.; Kurczyk, A.; Jelonek, K.; Żyła, J.; Mielańczyk, Ł.; Sitkiewicz, M.; Pietrowska, M.; Polańska, J.; Rzyman, W.; Widłak, P. The Lipid Composition of Serum-Derived Small Extracellular Vesicles in Participants of a Lung Cancer Screening Study. Cancers 2021, 13, 3414. [Google Scholar] [CrossRef]
- Metabolic and radiomic signature of early lung cancer. 2017 NCN. Available online: https://projekty.ncn.gov.pl/index.php?projekt_id=392614 (accessed on 19 January 2023).
- Smolarz, M.; Widlak, P. Serum Exosomes and Their miRNA Load-A Potential Biomarker of Lung Cancer. Cancers 2021, 13, 1373. [Google Scholar] [CrossRef]
- Ros-Mazurczyk, M.; Wojakowska, A.; Marczak, L.; Polanski, K.; Pietrowska, M.; Polanska, J.; Dziadziuszko, R.; Jassem, J.; Rzyman, W.; Widlak, P. Panel of serum metabolites discriminates cancer patients and healthy participants of lung cancer screening—A pilot study. Acta Biochim. Pol. 2017, 64, 513–518. [Google Scholar] [CrossRef]
- Mikhael, P.G.; Wohlwend, J.; Yala, A.; Karstens, L.; Xiang, J.; Takigami, A.K.; Bourgouin, P.P.; Chan, P.; Mrah, S.; Amayri, W.; et al. Sybil: A Validated Deep Learning Model to Predict Future Lung Cancer Risk From a Single Low-Dose Chest Computed Tomography. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2023, JCO2201345. [Google Scholar] [CrossRef]
- Clinical Trials. Available online: https://clinicaltrials.gov/ct2/results?cond=&term=artificial+intelligence&cntry=&state=&city=&dist= (accessed on 19 January 2023).
- Flores, R.; Henschke, C.; Taioli, E.; Yankelevitz, D. P2.06-045 Initiative for Early Lung Cancer Research on Treatment (IELCART). Journal of thoracic oncology: Official publication of the International Association for the Study of Lung Cancer. Poster Sess. 2017, 12, S1100. [Google Scholar] [CrossRef] [Green Version]
- Hey Google. Is This Lung Cancer? Available online: https://www.genengnews.com/insights/hey-google-is-this-lung-cancer/ (accessed on 15 January 2023).
- Van Leeuwen, K.G.; Schalekamp, S.; Rutten, M.J.C.M.; van Ginneken, B.; de Rooij, M. Artificial intelligence in radiology: 100 commercially available products and their scientific evidence. Eur. Radiol. 2021, 31, 3797–3804. [Google Scholar] [CrossRef]
- The global AI in the Medical Imaging Market Is Expected to Grow at a CAGR of 45.68% during the Forecast Period 2022–2027. Available online: https://finance.yahoo.com/news/global-ai-medical-imaging-market-084600155.html?guccounter=1&guce_referrer=aHR0cHM6Ly93d3cuZ29vZ2xlLmNvbS8&guce_referrer_sig=AQAAAM01n1pEJdgVCHCMdG9uOqA9i41Vmzh4eQ7YYgQ0UlS6baFXlYfleKOumFCXcvuahxBBt0JtZvKwWcYw4FEN4gv-3IEgPqDLKe4IpLmZT8yuIsAhJjWFN4pmRArfhKwFCBVpoMEWEo9rA7nDKjwrXLZmzOJ9dZYd5stRResaNXZmReportLinker,2022 (accessed on 10 December 2022).
- AI in Medical Imaging Market–Global Outlook & Forecast 2022–2027 ReportLinker. 2022. Available online: https://www.reportlinker.com/p06288135/?utm_source=GNW (accessed on 10 December 2022).
Image Processing | Technical Factors |
---|---|
acquisition | scanner reproducibility mode |
matrix size respiratory motion artefacts | |
reconstruction | parameters/protocol algorithm slice thickness [62] plane pixel dimension [62] soft/sharp kernel [63] |
segmentation | threshold discretization |
resampling filters | |
pre-processing | method |
feature extraction | shape texture size volume heterogeneity intensity histogram |
feature correlation | voxels |
test/data analysis | method |
clinical outcome modeling | statistical model |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Bidzińska, J.; Szurowska, E. See Lung Cancer with an AI. Cancers 2023, 15, 1321. https://doi.org/10.3390/cancers15041321
Bidzińska J, Szurowska E. See Lung Cancer with an AI. Cancers. 2023; 15(4):1321. https://doi.org/10.3390/cancers15041321
Chicago/Turabian StyleBidzińska, Joanna, and Edyta Szurowska. 2023. "See Lung Cancer with an AI" Cancers 15, no. 4: 1321. https://doi.org/10.3390/cancers15041321
APA StyleBidzińska, J., & Szurowska, E. (2023). See Lung Cancer with an AI. Cancers, 15(4), 1321. https://doi.org/10.3390/cancers15041321