CC BY-NC-ND 4.0 · Indian J Med Paediatr Oncol 2021; 42(06): 511-517
DOI: 10.1055/s-0041-1735577
Review Article

Artificial Intelligence: A New Tool in Oncologist's Armamentarium

Vineet Talwar
1   Department of Medical Oncology, Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
,
Kundan Singh Chufal
2   Department of Radiation Oncology, Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
,
Srujana Joga
1   Department of Medical Oncology, Rajiv Gandhi Cancer Institute & Research Centre, New Delhi, India
› Author Affiliations

Abstract

Artificial intelligence (AI) has become an essential tool in human life because of its pivotal role in communications, transportation, media, and social networking. Inspired by the complex neuronal network and its functions in human beings, AI, using computer-based algorithms and training, had been explored since the 1950s. To tackle the enormous amount of patients' clinical data, imaging, histopathological data, and the increasing pace of research on new treatments and clinical trials, and ever-changing guidelines for treatment with the advent of novel drugs and evidence, AI is the need of the hour. There are numerous publications and active work on AI's role in the field of oncology. In this review, we discuss the fundamental terminology of AI, its applications in oncology on the whole, and its limitations. There is an inter-relationship between AI, machine learning and, deep learning. The virtual branch of AI deals with machine learning. While the physical branch of AI deals with the delivery of different forms of treatment—surgery, targeted drug delivery, and elderly care. The applications of AI in oncology include cancer screening, diagnosis (clinical, imaging, and histopathological), radiation therapy (image acquisition, tumor and organs at risk segmentation, image registration, planning, and delivery), prediction of treatment outcomes and toxicities, prediction of cancer cell sensitivity to therapeutics and clinical decision-making. A specific area of interest is in the development of effective drug combinations tailored to every patient and tumor with the help of AI. Radiomics, the new kid on the block, deals with the planning and administration of radiotherapy. As with any new invention, AI has its fallacies. The limitations include lack of external validation and proof of generalizability, difficulty in data access for rare diseases, ethical and legal issues, no precise logic behind the prediction, and last but not the least, lack of education and expertise among medical professionals. A collaboration between departments of clinical oncology, bioinformatics, and data sciences can help overcome these problems in the near future.



Publication History

Article published online:
13 December 2021

© 2021. Indian Society of Medical and Paediatric Oncology. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Thieme Medical and Scientific Publishers Pvt. Ltd.
A-12, 2nd Floor, Sector 2, Noida-201301 UP, India

 
  • References

  • 1 Hamet P, Tremblay J. Artificial intelligence in medicine. Metabolism 2017; 69: S36-S40
  • 2 Data Science Bowl. 2017 Accessed July 3, 2020 at: https://www.kaggle.com/c/data-science-bowl-2017
  • 3 The digital mammography DREAM challenge. Accessed July 11, 2020 at: https://www.ibm.com/blogs/research/2017/06/dream-challenge-results/
  • 4 McKinney SM, Sieniek M, Godbole V. et al. International evaluation of an AI system for breast cancer screening. Nature 2020; 577 (7788): 89-94
  • 5 Webster DE, Suver C, Doerr M. et al. The Mole Mapper Study, mobile phone skin imaging and melanoma risk data collected using ResearchKit. Sci Data 2017; 4: 170005 https://doi.org/10.1038/sdata.2017.5
  • 6 Wang P, Xiao X, Brown JRG. et al. Development and validation of a deep-learning algorithm for the detection of polyps during colonoscopy. Nat Biomed Eng 2018; 2: 741-748
  • 7 Miotto R, Li L, Kidd BA, Dudley JT. Deep patient: an unsupervised representation to predict the future of patients from the electronic health records. Sci Rep 2016; 6: 26094 https://doi.org/10.1038/srep26094
  • 8 Kann BH, Aneja S, Loganadane GV. et al. Pretreatment identification of head and neck cancer nodal metastasis and extranodal extension using deep learning neural networks. Sci Rep 2018; 8: 14036. https://doi.org/10.1038/s41598-018-32441-y
  • 9 Chang K, Bai HX, Zhou H. et al. Residual convolutional neural network for determination of IDH status in low- and high-grade gliomas from MR imaging. Clin Cancer Res 2018; 24: 1073-1081
  • 10 Chang P, Grinband J, Weinberg BD. et al. Deep-learning convolutional neural networks accurately classify genetic mutations in gliomas. Am J Neuroradiol 2018; 39: 1201-1207
  • 11 Bibault JE, Giraud P, Durdux C. et al. Deep learning and radiomics predict complete response after neo-adjuvant chemoradiation for locally advanced rectal cancer. Sci Rep 2018; 8: 12611 DOI: 10.1038/s41598-018-30657-6.
  • 12 Sun R, Limkin EJ, Vakalopoulou M. et al. A radiomics approach to assess tumour-infiltrating CD8 cells and response to anti-PD-1 or anti-PD-L1 immunotherapy: an imaging biomarker, retrospective multicohort study. Lancet Oncol 2018; 19: 1180-1191
  • 13 Bejnordi BE, Veta M, van Diest PJ. et al. Diagnostic assessment of deep learning algorithms for detection of lymph node metastases in women with breast cancer. JAMA 2017; 318: 2199-2210
  • 14 Arvaniti E, Fricker KS, Moret M. et al. Automated Gleason grading of prostate cancer tissue microarrays via deep learning. Sci Rep 2018; 8: 12054, https://doi.org/10.1038/s41598-018-30535-1
  • 15 Coudray N, Ocampo PS, Sakellaropoulos T. et al. Classification and mutation prediction from non–small cell lung cancer histopathology images using deep learning. Nat Med 2018; 24: 1559-1567
  • 16 Arabi H, Dowling JA, Burgos N. et al. Comparative study of algorithms for synthetic CT generation from MRI: consequences for MRI-guided radiation planning in the pelvic region. Med Phys 2018; 45: 5218-5233
  • 17 Siversson C, Nordström F, Nilsson T. et al. Technical note: MRI only prostate radiotherapy planning using the statistical decomposition algorithm. Med Phys 2015; 42: 6090-6097
  • 18 Vinod SK, Jameson MG, Min M, Holloway LC. Uncertainties in volume delineation in radiation oncology: a systematic review and recommendations for future studies. Radiother Oncol 2016; 121: 169-179
  • 19 Hoang Duc AK, Eminowicz G, Mendes R. et al. Validation of clinical acceptability of An atlas-based segmentation algorithm for the delineation of organs at risk in head and neck cancer. Med Phys 2015; 42: 5027-5034
  • 20 Lustberg T, van Soest J, Gooding M. et al. Clinical evaluation of atlas and deep learning based automatic contouring for lung cancer. Radiother Oncol 2018; 126 (02) 312-317
  • 21 Men K, Dai J, Li Y. Automatic segmentation of the clinical target volume and organs at risk in the planning CT for rectal cancer using deep dilated convolutional neural networks. Med Phys 2017; 44: 6377-6389
  • 22 Viergever MA, Maintz JBA, Klein S, Murphy K, Staring M, Pluim JPW. A survey of medical image registration - under review. Med Image Anal 2016; 33: 140-144
  • 23 Yang X, Kwitt R, Styner M, Niethammer M. Quicksilver: Fast predictive image registration-a deep learning approach. NeuroImage 2017; Sep 1; 158: 378-396
  • 24 Miao S, Wang ZJ, Zheng Y, Liao R. Real-time 2D/3D registration via CNN regression. Paper presented at: Biomedical Imaging ISBI 2016 IEEE 13th International Symposium, IEEE 2016: 1430-1434
  • 25 McIntosh C, Welch M, McNiven A, Jaffray DA, Purdie TG. Fully automated treatment planning for head and neck radiotherapy using a voxel-based dose prediction and dose mimicking method. Phys Med Biol 2017; 62: 5926-5944
  • 26 Kim KH, Lee S, Shim JB. et al. A text-based data mining and toxicity prediction modeling system for a clinical decision support in radiation oncology: a preliminary study. J Korean Phys Soc 2017; 71: 231-237
  • 27 Kang J, Rancati T, Lee S. et al. Machine learning and radiogenomics: lessons learned and future directions. Front Oncol 2018; 8: 228 DOI: 10.3389/fonc.2018.00228.
  • 28 Ogunmolu OP, Gu X, Jiang S, Gans NR. A real-time, soft robotic patient positioning system for mask-less head-and-neck cancer radiotherapy: an initial investigation. Paper presented at: IEEE International Conference Automation Science Engineering CASE; August 24–28; 2015; Gothenburg, Sweden; 2015: 1539-1545
  • 29 Ogunmolu OP, Gu X, Jiang S, Gans NR. Vision-based control of a soft robot for maskless head and neck cancer radiotherapy. Paper presented at: IEEE International Conference Automation Science EngIneering CASE; August 21–24; 2016; Fort Worth, United States 2016: 180-187
  • 30 Park S, Lee SJ, Weiss E, Motai Y. Intra-. and inter-fractional variation prediction of lung tumors using fuzzy deep learning. IEEE J Transl Eng Health Med 2016; 4: 1-12
  • 31 Hanai T, Yatabe Y, Nakayama Y. et al. Prognostic models in patients with non-small-cell lung cancer using artificial neural networks in comparison with logistic regression. Cancer Sci 2003; 94 (05) 473-477
  • 32 Pella A, Cambria R, Riboldi M. et al. Use of machine learning methods for prediction of acute toxicity in organs at risk following prostate radiotherapy. Med Phys 2011; 38: 2859-2867
  • 33 Carrara M, Massari E, Cicchetti A. et al. Development of a ready-to-use graphical tool based on artificial neural network classification: application for the prediction of late fecal incontinence after prostate cancer radiation therapy. Int J Radiat Oncol Biol Phys 2018; 102 (05) 1533-1542
  • 34 Lee S, Kerns S, Ostrer H. et al. Machine learning on a genome-wide association study to predict late genitourinary toxicity after prostate radiation therapy. Int J Radiat Oncol Biol Phys 2018; 101: 128-135
  • 35 Ibragimov B, Toesca D, Chang D. et al. Development of deep neural network for individualized hepatobiliary toxicity prediction after liver SBRT. Med Phys 2018; 45: 4763-4774
  • 36 Zhen X, Chen J, Zhong Z. et al. Deep convolutional neural network with transfer learning for rectum toxicity prediction in cervical cancer radiotherapy: a feasibility study. Phys Med Biol 2017; 62: 8246-8263
  • 37 Wang J, Cao H, Zhang JZH, Qi Y. Computational protein design with deep learning neural networks. Sci Rep 2018; 8: 6349 DOI: 10.1038/s41598-018-24760-x.
  • 38 Zitnik M, Agrawal M, Leskovec J. Modeling polypharmacy side effects with graph convolutional networks. Bioinformatics 2018; 34: i457-i466
  • 39 Eulenberg P, Köhler N, Blasi T. et al. Reconstructing cell cycle and disease progression using deep learning. Nat Commun 2017; 8: 463 DOI: 10.1038/s41467-017-00623-3.
  • 40 Buggenthin F, Buettner F, Hoppe PS. et al. Prospective identification of hematopoietic lineage choice by deep learning. Nat Methods 2017; 14: 403-406
  • 41 Artemov AV, Putin E, Vanhaelen Q. et al. Integrated deep learned transcriptomic and structure- based predictor of clinical trials outcomes. Accessed December 29, 2016. at: https://www.biorxiv.org/content/10.1101/095653v2
  • 42 Menden MP, Iorio F, Garnett M. et al. Machine learning prediction of cancer cell sensitivity to drugs based on genomic and chemical properties. PLoS One 2013; 8: e61318 DOI: 10.1371/journal.pone.0061318.
  • 43 Han Y, Kim D. Deep convolutional neural networks for pan-specific peptide-MHC class I binding prediction. BMC Bioinformatics 2017; 18: 585 https://doi.org/10.1186/s12859-017-1997-x
  • 44 Sennaar K. AI and machine learning for clinical trials: examining 3 current applications. Emerj - Artificial Intelligence Research and Insight. Accessed January 18, 2019 at: https://emerj.com/ai-sector-overviews/ai-machine-learning-clinical-trials-examining-x-current-applications/
  • 45 Somashekhar SP, Sepúlveda MJ, Puglielli S. et al. Watson for Oncology and breast cancer treatment recommendations: agreement with an expert multidisciplinary tumor board. Ann Oncol 2018; 29(2): 418-423
  • 46 Liu C, Liu X, Wu F. et al. Using artificial intelligence (Watson for Oncology) for treatment recommendations amongst Chinese patients with lung cancer: feasibility study. J Med Internet Res 2018; 20: e11087 DOI: 10.2196/11087.
  • 47 Hegde RB, Prasad K, Hebbar H, Singh BMK. Comparison of traditional image processing and deep learning approaches for classification of white blood cells in peripheral blood smear images. Biocybern Biomed Eng 2019; 39 (02) 382-392
  • 48 Chandradevan R, Aljudi AA, Drumheller BR. et al. Machine- based detection and classification for bone marrow aspirate differential counts: initial development focusing on nonneoplastic cells. Lab Invest 2020; 100 (01) 98-109
  • 49 Milgrom SA, Elhalawani H, Lee J. et al. A PET radiomics model to predict refractory mediastinal Hodgkin lymphoma. Sci Rep 2019; 9 (01) 1322 DOI: 10.1038/s41598-018-37197-z.
  • 50 Nazha A, Komrokji RS, Meggendorfer M. et al. A personalized prediction model to risk stratify patients with myelodysplastic syndromes. Blood 2018; 132 (Suppl. 01) 793-793
  • 51 Ni W, Hu B, Zheng C. et al. Automated analysis of acute myeloid leukemia minimal residual disease using a support vector machine. Oncotarget 2016; 7 (44) 71915-71921
  • 52 Nazha A, Sekeres MA, Bejar R. et al. Genomic biomarkers to predict resistance to hypomethylating agents in patients with myelodysplastic syndromes using artificial intelligence. JCO Precis Oncol 2019; 3: 1-11
  • 53 Wilkinson MD, Dumontier M, Aalbersberg IJ. et al. The FAIR guiding principles for scientific data management and stewardship. Sci Data 2016; 3: 160018 https://doi.org/10.1038/sdata.2016.18
  • 54 Chavan V, Penev L. The data paper: a mechanism to incentivize data publishing in biodiversity science. BMC Bioinformatics 2011; 12 (Suppl. 15) S2 https://doi.org/10.1186/1471-2105-12-S15-S2
  • 55 Castelvecchi D. Can we open the black box of AI?. Nature 2016; 538(7623): 20-23
  • 56 Key Changes with the General Data Protection Regulation – EUGDPR. Accessed December 3, 2018 at: https://ec.europa.eu/info/law/law-topic/data-protection/data-protection-eu_en
  • 57 Yu KH, Beam AL, Kohane IS. Artificial intelligence in healthcare. Nat Biomed Eng 2018; 2: 719-731