Skip to main content
Log in

Radiomics in nuclear medicine: robustness, reproducibility, standardization, and how to avoid data analysis traps and replication crisis

  • Original Article
  • Published:
European Journal of Nuclear Medicine and Molecular Imaging Aims and scope Submit manuscript

Abstract

Radiomics in nuclear medicine is rapidly expanding. Reproducibility of radiomics studies in multicentre settings is an important criterion for clinical translation. We therefore performed a meta-analysis to investigate reproducibility of radiomics biomarkers in PET imaging and to obtain quantitative information regarding their sensitivity to variations in various imaging and radiomics-related factors as well as their inherent sensitivity. Additionally, we identify and describe data analysis pitfalls that affect the reproducibility and generalizability of radiomics studies. After a systematic literature search, 42 studies were included in the qualitative synthesis, and data from 21 were used for the quantitative meta-analysis. Data concerning measurement agreement and reliability were collected for 21 of 38 different factors associated with image acquisition, reconstruction, segmentation and radiomics-specific processing steps. Variations in voxel size, segmentation and several reconstruction parameters strongly affected reproducibility, but the level of evidence remained weak. Based on the meta-analysis, we also assessed inherent sensitivity to variations of 110 PET image biomarkers. SUVmean and SUVmax were found to be reliable, whereas image biomarkers based on the neighbourhood grey tone difference matrix and most biomarkers based on the size zone matrix were found to be highly sensitive to variations, and should be used with care in multicentre settings. Lastly, we identify 11 data analysis pitfalls. These pitfalls concern model validation and information leakage during model development, but also relate to reporting and the software used for data analysis. Avoiding such pitfalls is essential for minimizing bias in the results and to enable reproduction and validation of radiomics studies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Kessler LG, Barnhart HX, Buckler AJ, Choudhury KR, Kondratovich MV, Toledano A, et al. The emerging science of quantitative imaging biomarkers terminology and definitions for scientific studies and regulatory submissions. Stat Methods Med Res. 2015;24:9–26.

    Article  PubMed  Google Scholar 

  2. Vallières M, Zwanenburg A, Badic B, Cheze Le Rest C, Visvikis D, Hatt M. Responsible radiomics research for faster clinical translation. J Nucl Med. 2018;59:189–93.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Lodge MA. Repeatability of SUV in oncologic 18F-FDG PET. J Nucl Med. 2017;58:523–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Boellaard R. Standards for PET image acquisition and quantitative data analysis. J Nucl Med. 2009;50(Suppl 1):11S–20S.

    Article  CAS  PubMed  Google Scholar 

  5. Boellaard R, Delgado-Bolton R, Oyen WJG, Giammarile F, Tatsch K, Eschner W, et al. FDG PET/CT: EANM procedure guidelines for tumour imaging: version 2.0. Eur J Nucl Med Mol Imaging. 2015;42:328–54.

    Article  CAS  PubMed  Google Scholar 

  6. Hatt M, Tixier F, Pierce L, Kinahan PE, Le Rest CC, Visvikis D. Characterization of PET/CT images using texture analysis: the past, the present… any future? Eur J Nucl Med Mol Imaging. 2017;44:151–65.

    Article  PubMed  Google Scholar 

  7. Lovinfosse P, Visvikis D, Hustinx R, Hatt M. FDG PET radiomics: a review of the methodological aspects. Clin Transl Imaging. 2018;6:379–91.

    Article  Google Scholar 

  8. Reuzé S, Schernberg A, Orlhac F, Sun R, Chargari C, Dercle L, et al. Radiomics in nuclear medicine applied to radiation therapy: methods, pitfalls, and challenges. Int J Radiat Oncol Biol Phys. 2018;102:1117–42.

    Article  PubMed  Google Scholar 

  9. Traverso A, Wee L, Dekker A, Gillies R. Repeatability and reproducibility of radiomic features: a systematic review. Int J Radiat Oncol Biol Phys. 2018;102:1143–58.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Moher D, Liberati A, Tetzlaff J, Altman DG, PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009;6:e1000097.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Med. 2009;6:e1000100.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Bland JM, Altman DG. Statistical methods for assessing agreement between two methods of clinical measurement. Lancet. 1986;1:307–10.

    Article  CAS  PubMed  Google Scholar 

  13. McGraw KO, Wong SP. Forming inferences about some intraclass correlation coefficients. Psychol Methods. 1996;1:30–46.

    Article  Google Scholar 

  14. de Vet HCW, Terwee CB, Knol DL, Bouter LM. When to use agreement versus reliability measures. J Clin Epidemiol. 2006;59:1033–9.

    Article  PubMed  Google Scholar 

  15. Kottner J, Audige L, Brorson S, Donner A, Gajewski BJ, Hróbjartsson A, et al. Guidelines for Reporting Reliability and Agreement Studies (GRRAS) were proposed. Int J Nurs Stud. 2011;48:661–71.

    Article  PubMed  Google Scholar 

  16. Nyflot M, Bowen SR, Yang F, Byrd D, Sandison GA, Kinahan PE. Quantitative radiomics: effects of stochastic variability on PET textural features and implications for clinical trials. Int J Radiat Oncol Biol Phys. 2015;93:E566–7.

    Article  Google Scholar 

  17. Carles M, Torres-Espallardo I, Alberich-Bayarri A, Olivas C, Bello P, Nestle U, et al. Evaluation of PET texture features with heterogeneous phantoms: complementarity and effect of motion and segmentation method. Phys Med Biol. 2017;62:652–68.

    Article  CAS  PubMed  Google Scholar 

  18. Yip S, McCall K, Aristophanous M, Chen AB, Aerts HJWL, Berbeco R. Comparison of texture features derived from static and respiratory-gated PET images in non-small cell lung cancer. PLoS One. 2014;9:e115510.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Lovat E, Siddique M, Goh V, Ferner RE, Cook GJR, Warbey VS. The effect of post-injection 18F-FDG PET scanning time on texture analysis of peripheral nerve sheath tumours in neurofibromatosis-1. EJNMMI Res. 2017;7:35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Manabe O, Ohira H, Hirata K, Hayashi S, Naya M, Tsujino I, et al. Use of 18F-FDG PET/CT texture analysis to diagnose cardiac sarcoidosis. Eur J Nucl Med Mol Imaging. 2019;46:1240–7.

    Article  CAS  PubMed  Google Scholar 

  21. Bailly C, Bodet-Milin C, Couespel S, Necib H, Kraeber-Bodéré F, Ansquer C, et al. Revisiting the robustness of PET-based textural features in the context of multi-centric trials. PLoS One. 2016;11:e0159984.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Grootjans W, Tixier F, van der Vos CS, Vriens D, Le Rest CC, Bussink J, et al. The impact of optimal respiratory gating and image noise on evaluation of intratumor heterogeneity on 18F-FDG PET imaging of lung cancer. J Nucl Med. 2016;57:1692–8.

    Article  PubMed  Google Scholar 

  23. Presotto L, Bettinardi V, De Bernardi E, Belli ML, Cattaneo GM, Broggi S, et al. PET textural features stability and pattern discrimination power for radiomics analysis: an “ad-hoc” phantoms study. Phys Med. 2018;50:66–74.

    Article  CAS  PubMed  Google Scholar 

  24. Shiri I, Rahmim A, Ghaffarian P, Geramifar P, Abdollahi H, Bitarafan-Rajabi A. The impact of image reconstruction settings on 18F-FDG PET radiomic features: multi-scanner phantom and patient studies. Eur Radiol. 2017;27:4498–509.

    Article  PubMed  Google Scholar 

  25. Carles M, Bach T, Torres-Espallardo I, Baltas D, Nestle U, Martí-Bonmatí L. Significance of the impact of motion compensation on the variability of PET image features. Phys Med Biol. 2018;63:065013.

    Article  CAS  PubMed  Google Scholar 

  26. Oliver JA, Budzevich M, Zhang GG, Dilling TJ, Latifi K, Moros EG. Variability of image features computed from conventional and respiratory-gated PET/CT images of lung cancer. Transl Oncol. 2015;8:524–34.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Tixier F, Vriens D, Cheze-Le Rest C, Hatt M, Disselhorst JA, Oyen WJG, et al. Comparison of tumor uptake heterogeneity characterization between static and parametric 18F-FDG PET images in non-small cell lung cancer. J Nucl Med. 2016;57:1033–9.

    Article  CAS  PubMed  Google Scholar 

  28. Reuzé S, Orlhac F, Chargari C, Nioche C, Limkin E, Riet F, et al. Prediction of cervical cancer recurrence using textural features extracted from 18F-FDG PET images acquired with different scanners. Oncotarget. 2017;8:43169–79.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Desseroit M-C, Tixier F, Weber WA, Siegel BA, Cheze Le Rest C, Visvikis D, et al. Reliability of PET/CT shape and heterogeneity features in functional and morphologic components of non-small cell lung cancer tumors: a repeatability analysis in a prospective multicenter cohort. J Nucl Med. 2017;58:406–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Forgacs A, Pall Jonsson H, Dahlbom M, Daver F, DiFranco M, Opposits G, et al. A study on the basic criteria for selecting heterogeneity parameters of F18-FDG PET images. PLoS One. 2016;11:e0164113.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Gallivanone F, Interlenghi M, D’Ambrosio D, Trifirò G, Castiglioni I. Parameters influencing PET imaging features: a phantom study with irregular and heterogeneous synthetic lesions. Contrast Media Mol Imaging. 2018;2018:5324517.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Leijenaar RTH, Carvalho S, Velazquez ER, van Elmpt WJC, Parmar C, Hoekstra OS, et al. Stability of FDG-PET radiomics features: an integrated analysis of test-retest and inter-observer variability. Acta Oncol. 2013;52:1391–7.

    Article  CAS  PubMed  Google Scholar 

  33. Tixier F, Hatt M, Le Rest CC, Le Pogam A, Corcos L, Visvikis D. Reproducibility of tumor uptake heterogeneity characterization through textural feature analysis in 18F-FDG PET. J Nucl Med. 2012;53:693–700.

    Article  PubMed  Google Scholar 

  34. van Velden FHP, Nissen IA, Jongsma F, Velasquez LM, Hayes W, Lammertsma AA, et al. Test-retest variability of various quantitative measures to characterize tracer uptake and/or tracer uptake heterogeneity in metastasized liver for patients with colorectal carcinoma. Mol Imaging Biol. 2014;16:13–8.

    Article  PubMed  Google Scholar 

  35. van Velden FHP, Kramer GM, Frings V, Nissen IA, Mulder ER, de Langen AJ, et al. Repeatability of radiomic features in non-small-cell lung cancer [(18)F]FDG-PET/CT studies: impact of reconstruction and delineation. Mol Imaging Biol. 2016;18:788–95.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Willaime JMY, Turkheimer FE, Kenny LM, Aboagye EO. Quantification of intra-tumour cell proliferation heterogeneity using imaging descriptors of 18F fluorothymidine-positron emission tomography. Phys Med Biol. 2013;58:187–203.

    Article  CAS  PubMed  Google Scholar 

  37. Altazi BA, Zhang GG, Fernandez DC, Montejo ME, Hunt D, Werner J, et al. Reproducibility of F18-FDG PET radiomic features for different cervical tumor segmentation methods, gray-level discretization, and reconstruction algorithms. J Appl Clin Med Phys. 2017;18:32–48.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Galavis PE, Hollensen C, Jallow N, Paliwal B, Jeraj R. Variability of textural features in FDG PET images due to different acquisition modes and reconstruction parameters. Acta Oncol. 2010;49:1012–6.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Lasnon C, Majdoub M, Lavigne B, Do P, Madelaine J, Visvikis D, et al. 18F-FDG PET/CT heterogeneity quantification through textural features in the era of harmonisation programs: a focus on lung cancer. Eur J Nucl Med Mol Imaging. 2016;43:2324–35.

    Article  PubMed  Google Scholar 

  40. Yan J, Chu-Shern JL, Loi HY, Khor LK, Sinha AK, Quek ST, et al. Impact of image reconstruction settings on texture features 18F-FDG PET. J Nucl Med. 2015;56:1667–73.

    Article  CAS  PubMed  Google Scholar 

  41. Doumou G, Siddique M, Tsoumpas C, Goh V, Cook GJ. The precision of textural analysis in (18)F-FDG-PET scans of oesophageal cancer. Eur Radiol. 2015;25:2805–12.

    Article  PubMed  Google Scholar 

  42. Hatt M, Tixier F, Cheze Le Rest C, Pradier O, Visvikis D. Robustness of intratumour 18F-FDG PET uptake heterogeneity quantification for therapy response prediction in oesophageal carcinoma. Eur J Nucl Med Mol Imaging. 2013;40:1662–71.

    Article  PubMed  Google Scholar 

  43. Orlhac F, Nioche C, Soussan M, Buvat I. Understanding changes in tumor textural indices in PET: a comparison between visual assessment and index values in simulated and patient data. J Nucl Med. 2017;58:387–92.

    Article  PubMed  Google Scholar 

  44. Bashir U, Azad G, Siddique MM, Dhillon S, Patel N, Bassett P, et al. The effects of segmentation algorithms on the measurement of 18F-FDG PET texture parameters in non-small cell lung cancer. EJNMMI Res. 2017;7:60.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Belli ML, Mori M, Broggi S, Cattaneo GM, Bettinardi V, Dell’Oca I, et al. Quantifying the robustness of [18F]FDG-PET/CT radiomic features with respect to tumor delineation in head and neck and pancreatic cancer patients. Phys Med. 2018;49:105–11.

    Article  PubMed  Google Scholar 

  46. Takeda K, Takanami K, Shirata Y, Yamamoto T, Takahashi N, Ito K, et al. Clinical utility of texture analysis of 18F-FDG PET/CT in patients with stage I lung cancer treated with stereotactic body radiotherapy. J Radiat Res. 2017;58:862–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lu L, Lv W, Jiang J, Ma J, Feng Q, Rahmim A, et al. Robustness of radiomic features in [11C]choline and [18F]FDG PET/CT imaging of nasopharyngeal carcinoma: impact of segmentation and discretization. Mol Imaging Biol. 2016;18:935–45.

    Article  CAS  PubMed  Google Scholar 

  48. Mu W, Chen Z, Liang Y, Shen W, Yang F, Dai R, et al. Staging of cervical cancer based on tumor heterogeneity characterized by texture features on (18)F-FDG PET images. Phys Med Biol. 2015;60:5123–39.

    Article  PubMed  Google Scholar 

  49. Orlhac F, Soussan M, Maisonobe J-A, Garcia CA, Vanderlinden B, Buvat I. Tumor texture analysis in 18F-FDG PET: relationships between texture parameters, histogram indices, standardized uptake values, metabolic volumes, and total lesion glycolysis. J Nucl Med. 2014;55:414–22.

    Article  CAS  PubMed  Google Scholar 

  50. Wu J, Aguilera T, Shultz D, Gudur M, Rubin DL, Loo BW Jr, et al. Early-stage non-small cell lung cancer: quantitative imaging characteristics of (18)F fluorodeoxyglucose PET/CT allow prediction of distant metastasis. Radiology. 2016;281:270–8.

    Article  PubMed  Google Scholar 

  51. Yip SSF, Parmar C, Kim J, Huynh E, Mak RH, Aerts HJWL. Impact of experimental design on PET radiomics in predicting somatic mutation status. Eur J Radiol. 2017;97:8–15.

    Article  PubMed  Google Scholar 

  52. Leijenaar RTH, Nalbantov G, Carvalho S, van Elmpt WJC, Troost EGC, Boellaard R, et al. The effect of SUV discretization in quantitative FDG-PET radiomics: the need for standardized methodology in tumor texture analysis. Sci Rep. 2015;5:11075.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Orlhac F, Soussan M, Chouahnia K, Martinod E, Buvat I. 18F-FDG PET-derived textural indices reflect tissue-specific uptake pattern in non-small cell lung cancer. PLoS One. 2015;10:e0145063.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Hatt M, Majdoub M, Vallières M, Tixier F, Le Rest CC, Groheux D, et al. 18F-FDG PET uptake characterization through texture analysis: investigating the complementary nature of heterogeneity and functional tumor volume in a multi-cancer site patient cohort. J Nucl Med. 2015;56:38–44.

    Article  CAS  PubMed  Google Scholar 

  55. Oliver JA, Budzevich M, Hunt D, Moros EG, Latifi K, Dilling TJ, et al. Sensitivity of image features to noise in conventional and respiratory-gated PET/CT images of lung cancer: uncorrelated noise effects. Technol Cancer Res Treat. 2017;16:595–608.

    Article  PubMed  Google Scholar 

  56. Lv W, Yuan Q, Wang Q, Ma J, Jiang J, Yang W, et al. Robustness versus disease differentiation when varying parameter settings in radiomics features: application to nasopharyngeal PET/CT. Eur Radiol. 2018;28:3245–54.

    Article  PubMed  Google Scholar 

  57. Bogowicz M, Leijenaar RTH, Tanadini-Lang S, Riesterer O, Pruschy M, Studer G, et al. Post-radiochemotherapy PET radiomics in head and neck cancer – the influence of radiomics implementation on the reproducibility of local control tumor models. Radiother Oncol. 2017;125:385–91.

    Article  PubMed  Google Scholar 

  58. Zwanenburg A, Leger S, Vallières M, Löck S, for the Image Biomarker Standardisation Initiative. Image biomarker standardisation initiative [Internet]. arXiv:1612.07003 [cs.CV]. 2016. http://arxiv.org/abs/1612.07003.

  59. Hatt M, Lee JA, Schmidtlein CR, Naqa IE, Caldwell C, De Bernardi E, et al. Classification and evaluation strategies of auto-segmentation approaches for PET: report of AAPM task group no. 211. Med Phys. 2017;44:e1–42.

    Article  CAS  PubMed  Google Scholar 

  60. Nestle U, Kremp S, Schaefer-Schuler A, Sebastian-Welsch C, Hellwig D, Rübe C, et al. Comparison of different methods for delineation of 18F-FDG PET-positive tissue for target volume definition in radiotherapy of patients with non-small cell lung cancer. J Nucl Med. 2005;46:1342–8.

    PubMed  Google Scholar 

  61. Schinagl DAX, Vogel WV, Hoffmann AL, van Dalen JA, Oyen WJ, Kaanders JHAM. Comparison of five segmentation tools for 18F-fluoro-deoxy-glucose-positron emission tomography-based target volume definition in head and neck cancer. Int J Radiat Oncol Biol Phys. 2007;69:1282–9.

    Article  CAS  PubMed  Google Scholar 

  62. Zaidi H, El Naqa I. PET-guided delineation of radiation therapy treatment volumes: a survey of image segmentation techniques. Eur J Nucl Med Mol Imaging. 2010;37:2165–87.

    Article  PubMed  Google Scholar 

  63. Cheebsumon P, Yaqub M, van Velden FHP, Hoekstra OS, Lammertsma AA, Boellaard R. Impact of [18F]FDG PET imaging parameters on automatic tumour delineation: need for improved tumour delineation methodology. Eur J Nucl Med Mol Imaging. 2011;38:2136–44.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Hatt M, Laurent B, Ouahabi A, Fayad H, Tan S, Li L, et al. The first MICCAI challenge on PET tumor segmentation. Med Image Anal. 2018;44:177–95.

    Article  PubMed  Google Scholar 

  65. Shafiq-Ul-Hassan M, Zhang GG, Latifi K, Ullah G, Hunt DC, Balagurunathan Y, et al. Intrinsic dependencies of CT radiomic features on voxel size and number of gray levels. Med Phys. 2017;44:1050–62.

    Article  CAS  PubMed  Google Scholar 

  66. Mackin D, Fave X, Zhang L, Yang J, Jones AK, Ng CS, et al. Harmonizing the pixel size in retrospective computed tomography radiomics studies. PLoS One. 2017;12:e0178524.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Larue RTHM, van Timmeren JE, de Jong EEC, Feliciani G, Leijenaar RTH, Schreurs WMJ, et al. Influence of gray level discretization on radiomic feature stability for different CT scanners, tube currents and slice thicknesses: a comprehensive phantom study. Acta Oncol. 2017;56:1544–53.

    Article  PubMed  Google Scholar 

  68. Foy JJ, Robinson KR, Li H, Giger ML, Al-Hallaq H, Armato SG. Variation in algorithm implementation across radiomics software. J Med Imaging. 2018;5:044505.

    Article  Google Scholar 

  69. Zwanenburg A, Abdalah MA, Apte A, Ashrafinia S, Beukinga J, Bogowicz M, et al. PO-0981: results from the Image Biomarker Standardisation Initiative. Radiother Oncol. 2018;127:S543–4.

    Article  Google Scholar 

  70. Hatt M, Vallieres M, Visvikis D, Zwanenburg A. IBSI: an international community radiomics standardization initiative. J Nucl Med. 2018;59(Suppl 1):287–7.

  71. Domingos P. A few useful things to know about machine learning. Commun ACM. 2012;55:78–87.

    Article  Google Scholar 

  72. Moons KGM, Altman DG, Reitsma JB, Ioannidis JPA, Macaskill P, Steyerberg EW, et al. Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD): explanation and elaboration. Ann Intern Med. 2015;162:W1–73.

    Article  PubMed  Google Scholar 

  73. James G, Witten D, Hastie T, Tibshirani R. An Introduction to Statistical Learning: with Applications in R. New York: Springer; 2013.

    Book  Google Scholar 

  74. Hastie T, Tibshirani R, Friedman J. The Elements of Statistical Learning: Data Mining, Inference, and Prediction. Second ed. New York: Springer Science+Business Media; 2009.

  75. García S, Luengo J, Herrera F. Data Preprocessing in Data Mining. New York: Springer; 2015.

    Book  Google Scholar 

  76. Box GEP, Cox DR. An analysis of transformations. J R Stat Soc Ser B Stat Methodol. 1964;26:211–52.

    Google Scholar 

  77. Yeo I, Johnson RA. A new family of power transformations to improve normality or symmetry. Biometrika. 2000;87:954–9.

    Article  Google Scholar 

  78. Greenland S, Finkle WD. A critical look at methods for handling missing covariates in epidemiologic regression analyses. Am J Epidemiol. 1995;142:1255–64.

    Article  CAS  PubMed  Google Scholar 

  79. Donders ART, van der Heijden GJMG, Stijnen T, Moons KGM. Review: a gentle introduction to imputation of missing values. J Clin Epidemiol. 2006;59:1087–91.

    Article  PubMed  Google Scholar 

  80. Luengo J, García S, Herrera F. On the choice of the best imputation methods for missing values considering three groups of classification methods. Knowl Inf Syst. 2012;32:77–108.

    Article  Google Scholar 

  81. Orlhac F, Boughdad S, Philippe C, Stalla-Bourdillon H, Nioche C, Champion L, et al. A postreconstruction harmonization method for multicenter radiomic studies in PET. J Nucl Med. 2018;59:1321–8.

    Article  CAS  PubMed  Google Scholar 

  82. Orlhac F, Frouin F, Nioche C, Ayache N, Buvat I. Validation of a method to compensate multicenter effects affecting CT radiomics. Radiology. 2019;291:53–9.

    Article  PubMed  Google Scholar 

  83. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics. 2007;8:118–27.

    Article  PubMed  Google Scholar 

  84. Lucia F, Visvikis D, Vallières M, Desseroit M-C, Miranda O, Robin P, et al. External validation of a combined PET and MRI radiomics model for prediction of recurrence in cervical cancer patients treated with chemoradiotherapy. Eur J Nucl Med Mol Imaging. 2019;46:864–77.

    Article  PubMed  Google Scholar 

  85. Foley KG, Shi Z, Whybra P, Kalendralis P, Larue R, Berbee M, et al. External validation of a prognostic model incorporating quantitative PET image features in oesophageal cancer. Radiother Oncol. 2019;133:205–12.

    Article  PubMed  Google Scholar 

  86. Chatterjee A, Vallières M, Dohan A, Levesque IR, Ueno Y, Saif S, et al. Creating robust predictive radiomic models for data from independent institutions using normalization. IEEE TRPMS. 2019;3:210–5.

    Google Scholar 

  87. He H, Garcia EA. Learning from imbalanced data. IEEE Trans Knowl Data Eng. 2009;21:1263–84.

  88. Krawczyk B. Learning from imbalanced data: open challenges and future directions. Prog Artif Intell. 2016;5:221–32.

    Article  Google Scholar 

  89. Chawla NV, Bowyer KW, Hall LO, Kegelmeyer WP. SMOTE: synthetic minority over-sampling technique. J Artif Intell Res. 2002;16:321–57.

    Article  Google Scholar 

  90. Haibo He, Yang Bai, Garcia EA, Shutao Li. ADASYN: Adaptive synthetic sampling approach for imbalanced learning. 2008 IEEE International Joint Conference on Neural Networks (IEEE World Congress on Computational Intelligence). 2008. p. 1322–8.

  91. Cunningham JP, Ghahramani Z. Linear dimensionality reduction: survey, insights, and generalizations. J Mach Learn Res. 2015;16:2859–900.

    Google Scholar 

  92. John GH, Kohavi R, Pfleger K. Irrelevant features and the subset selection problem. In: Cohen WW, Hirsh H, editors. Machine Learning Proceedings 1994. San Francisco: Morgan Kaufmann; 1994. p. 121–9.

    Chapter  Google Scholar 

  93. Tolosi L, Lengauer T. Classification with correlated features: unreliability of feature ranking and solutions. Bioinformatics. 2011;27:1986–94.

    Article  CAS  PubMed  Google Scholar 

  94. Park MY, Hastie T, Tibshirani R. Averaged gene expressions for regression. Biostatistics. 2007;8:212–27.

    Article  PubMed  Google Scholar 

  95. Leger S, Zwanenburg A, Pilz K, Lohaus F, Linge A, Zöphel K, et al. A comparative study of machine learning methods for time-to-event survival data for radiomics risk modelling. Sci Rep. 2017;7:13206.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  96. Lambin P, Leijenaar RTH, Deist TM, Peerlings J, de Jong EEC, van Timmeren J, et al. Radiomics: the bridge between medical imaging and personalized medicine. Nat Rev Clin Oncol. 2017;14:749–62.

    Article  PubMed  Google Scholar 

  97. Zwanenburg A, Leger S, Agolli L, Pilz K, Troost EGC, Richter C, et al. Assessing robustness of radiomic features by image perturbation. Sci Rep. 2019;9:614.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Guyon I, Elisseeff A. An introduction to variable and feature selection. J Mach Learn Res. 2003;3:1157–82.

    Google Scholar 

  99. Saeys Y, Inza I, Larrañaga P. A review of feature selection techniques in bioinformatics. Bioinformatics. 2007;23:2507–17.

    Article  CAS  PubMed  Google Scholar 

  100. Li J, Cheng K, Wang S, Morstatter F, Trevino RP, Tang J, et al. Feature selection: a data perspective. ACM Computing Surveys. 2018;50:94.

    Google Scholar 

  101. Parmar C, Grossmann P, Bussink J, Lambin P, Aerts HJWL. Machine learning methods for quantitative radiomic biomarkers. Sci Rep. 2015;5:13087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Parmar C, Grossmann P, Rietveld D, Rietbergen MM, Lambin P, Aerts HJWL. Radiomic machine-learning classifiers for prognostic biomarkers of head and neck cancer. Front Oncol. 2015;5:272.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Zhang B, He X, Ouyang F, Gu D, Dong Y, Zhang L, et al. Radiomic machine-learning classifiers for prognostic biomarkers of advanced nasopharyngeal carcinoma. Cancer Lett. 2017;403:21–7.

    Article  CAS  PubMed  Google Scholar 

  104. Sun W, Jiang M, Dang J, Chang P, Yin F-F. Effect of machine learning methods on predicting NSCLC overall survival time based on Radiomics analysis. Radiat Oncol. 2018;13:197.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Kalousis A, Prados J, Hilario M. Stability of feature selection algorithms: a study on high-dimensional spaces. Knowl Inf Syst. 2007;12:95–116.

    Article  Google Scholar 

  106. Haury A-C, Gestraud P, Vert J-P. The influence of feature selection methods on accuracy, stability and interpretability of molecular signatures. PLoS One. 2011;6:e28210.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Saeys Y, Abeel T, Van de Peer Y. Robust feature selection using ensemble feature selection techniques. In: Daelemans W, Goethals B, Morik K, editors. Machine Learning and Knowledge Discovery in Databases. Berlin: Springer; 2008. p. 313–25.

    Chapter  Google Scholar 

  108. Abeel T, Helleputte T, Van de Peer Y, Dupont P, Saeys Y. Robust biomarker identification for cancer diagnosis with ensemble feature selection methods. Bioinformatics. 2010;26:392–8.

    Article  CAS  PubMed  Google Scholar 

  109. Meinshausen N, Bühlmann P. Stability selection. J R Stat Soc Ser B Stat Methodol. 2010;72:417–73.

    Article  Google Scholar 

  110. Wald R, Khoshgoftaar TM, Dittman D, Awada W, Napolitano A. An extensive comparison of feature ranking aggregation techniques in bioinformatics. 2012 IEEE 13th International Conference on Information Reuse Integration (IRI). 2012; p. 377–84.

  111. Bühlmann P, Hothorn T. Boosting algorithms: regularization, prediction and model fitting. Stat Sci. 2007;22:477–505.

    Google Scholar 

  112. Hofner B, Boccuto L, Göker M. Controlling false discoveries in high-dimensional situations: boosting with stability selection. BMC Bioinformatics. 2015;16:144.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Fernández-Delgado M, Cernadas E, Barro S, Amorim D. Do we need hundreds of classifiers to solve real world classification problems? J Mach Learn Res. 2014;15:3133–81.

    Google Scholar 

  114. Nelder JA, Wedderburn RWM. Generalized linear models. J R Stat Soc Ser A. 1972;135:370–84.

    Article  Google Scholar 

  115. Tibshirani R. Regression shrinkage and selection via the lasso. J R Stat Soc Ser B Stat Methodol. 1996;58:267–88.

    Google Scholar 

  116. Breiman L. Random forests. Mach Learn. 2001;45:5–32.

    Article  Google Scholar 

  117. Chen T, Guestrin C. XGBoost: a scalable tree boosting system. Proceedings of the 22nd ACM SIGKDD International Conference on Knowledge Discovery and Data Mining. 2016. p. 785–94.

  118. Bergstra J, Bengio Y. Random search for hyper-parameter optimization. J Mach Learn Res. 2012;13:281–305.

    Google Scholar 

  119. Hutter F, Hoos HH, Leyton-Brown K. Sequential model-based optimization for general algorithm configuration. In: Coello CAC, editor. Learning and Intelligent Optimization. Berlin: Springer; 2011. p. 507–23.

    Chapter  Google Scholar 

  120. Hand DJ, Till RJ. A simple generalisation of the area under the ROC curve for multiple class classification problems. Mach Learn. 2001;45:171–86.

  121. Japkowicz N, Stephen S. The class imbalance problem: a systematic study. Intell Data Anal. 2002;6:429–49.

    Article  Google Scholar 

  122. Brodersen KH, Ong CS, Stephan KE, Buhmann JM. The balanced accuracy and its posterior distribution. 20th International Conference on Pattern Recognition. 2010; p. 3121–4.

  123. Baldi P, Brunak S, Chauvin Y, Andersen CA, Nielsen H. Assessing the accuracy of prediction algorithms for classification: an overview. Bioinformatics. 2000;16:412–24.

    Article  CAS  PubMed  Google Scholar 

  124. Harrell FE Jr, Lee KL, Mark DB. Multivariable prognostic models: issues in developing models, evaluating assumptions and adequacy, and measuring and reducing errors. Stat Med. 1996;15:361–87.

    Article  PubMed  Google Scholar 

  125. Uno H, Cai T, Pencina MJ, D’Agostino RB, Wei LJ. On the C-statistics for evaluating overall adequacy of risk prediction procedures with censored survival data. Stat Med. 2011;30:1105–17.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Royston P, Altman DG. External validation of a Cox prognostic model: principles and methods. BMC Med Res Methodol. 2013;13:33.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Miller ME, Langefeld CD, Tierney WM, Hui SL, McDonald CJ. Validation of probabilistic predictions. Med Decis Mak. 1993;13:49–58.

    Article  CAS  Google Scholar 

  128. Steyerberg EW, Vickers AJ, Cook NR, Gerds T, Gonen M, Obuchowski N, et al. Assessing the performance of prediction models: a framework for traditional and novel measures. Epidemiology. 2010;21:128–38.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Hosmer DW Jr, Lemeshow S, Sturdivant RX. Applied Logistic Regression. Hoboken: Wiley; 2013.

    Book  Google Scholar 

  130. D’Agostino RB, Nam B-H. Evaluation of the performance of survival analysis models: discrimination and calibration measures. In: Balakrishnan N, Rao CR, editors. Handbook of Statistics. Amsterdam: Elsevier; 2003; p. 1–25.

    Google Scholar 

  131. Demler OV, Paynter NP, Cook NR. Tests of calibration and goodness-of-fit in the survival setting. Stat Med. 2015;34:1659–80.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Dupuy A, Simon RM. Critical review of published microarray studies for cancer outcome and guidelines on statistical analysis and reporting. J Natl Cancer Inst. 2007;99:147–57.

    Article  PubMed  Google Scholar 

  133. Shi L, Campbell G, Jones WD, Campagne F, Wen Z, Walker SJ, et al. The MicroArray Quality Control (MAQC)-II study of common practices for the development and validation of microarray-based predictive models. Nat Biotechnol. 2010;28:827–38.

    Article  CAS  PubMed  Google Scholar 

  134. Chalkidou A, O’Doherty MJ, Marsden PK. False discovery rates in PET and CT studies with texture features: a systematic review. PLoS One. 2015;10:e0124165.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  135. Zwanenburg A, Löck S. Why validation of prognostic models matters? Radiother Oncol. 2018;127:370–3.

    Article  PubMed  Google Scholar 

  136. Binder H, Porzelius C, Schumacher M. An overview of techniques for linking high-dimensional molecular data to time-to-event endpoints by risk prediction models. Biom J. 2011;53:170–89.

    Article  PubMed  Google Scholar 

  137. Chen H-C, Kodell RL, Cheng KF, Chen JJ. Assessment of performance of survival prediction models for cancer prognosis. BMC Med Res Methodol. 2012;12:102.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Vallières M, Kay-Rivest E, Perrin LJ, Liem X, Furstoss C, Aerts HJWL, et al. Radiomics strategies for risk assessment of tumour failure in head-and-neck cancer. Sci Rep. 2017;7:10117.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  139. Sun R, Limkin EJ, Vakalopoulou M, Dercle L, Champiat S, Han SR, 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;19:1180–91.

    Article  CAS  PubMed  Google Scholar 

  140. Aerts HJWL, Velazquez ER, Leijenaar RTH, Parmar C, Grossmann P, Carvalho S, et al. Decoding tumour phenotype by noninvasive imaging using a quantitative radiomics approach. Nat Commun. 2014;5:4006.

    Article  CAS  PubMed  Google Scholar 

  141. Leger S, Zwanenburg A, Pilz K, Zschaeck S, Zöphel K, Kotzerke J, et al. CT imaging during treatment improves radiomic models for patients with locally advanced head and neck cancer. Radiother Oncol. 2019;130:10–7.

    Article  PubMed  Google Scholar 

  142. Welch ML, McIntosh C, Haibe-Kains B, Milosevic MF, Wee L, Dekker A, et al. Vulnerabilities of radiomic signature development: the need for safeguards. Radiother Oncol. 2019;130:2–9.

    Article  PubMed  Google Scholar 

  143. Sanduleanu S, Woodruff HC, de Jong EEC, van Timmeren JE, Jochems A, Dubois L, et al. Tracking tumor biology with radiomics: a systematic review utilizing a radiomics quality score. Radiother Oncol. 2018;127:349–60.

    Article  PubMed  Google Scholar 

  144. Collins GS, Reitsma JB, Altman DG, Moons KGM. Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD): the TRIPOD statement. Br J Surg. 2015;102:148–58.

    Article  CAS  PubMed  Google Scholar 

  145. Simera I, Moher D, Hirst A, Hoey J, Schulz KF, Altman DG. Transparent and accurate reporting increases reliability, utility, and impact of your research: reporting guidelines and the EQUATOR Network. BMC Med. 2010;8:24.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Wilkinson MD, Dumontier M, Aalbersberg IJJ, Appleton G, Axton M, Baak A, et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci Data. 2016;3:160018.

    Article  PubMed  PubMed Central  Google Scholar 

  147. R Core Team. R: A Language and Environment for Statistical Computing. Vienna, Austria: R Foundation for Statistical Computing; 2018. https://www.R-project.org/

  148. Pedregosa F, Varoquaux G, Gramfort A, Michel V, Thirion B, Grisel O, et al. Scikit-learn: machine learning in Python. J Mach Learn Res. 2011;12:2825–30.

    Google Scholar 

  149. McKinney W. Data structures for statistical computing in Python. Austin: Proceedings of the 9th Python in Science Conference; 2010. p. 51–6.

    Google Scholar 

  150. Feurer M, Klein A, Eggensperger K, Springenberg J, Blum M, Hutter F. Efficient and robust automated machine learning. In: Cortes C, Lawrence ND, Lee DD, Sugiyama M, Garnett R, editors. Advances in Neural Information Processing Systems 28. New York: Curran Associates; 2015. p. 2962–70.

    Google Scholar 

  151. Kuhn M. Building predictive models in R using the caret package. J Stat Softw. 2008;28:1–26.

    Article  Google Scholar 

  152. Bischl B, Lang M, Kotthoff L, Schiffner J, Richter J, Studerus E, et al. mlr: machine learning in R. J Mach Learn Res. 2016;17:5938–42.

    Google Scholar 

  153. van Griethuysen JJM, Fedorov A, Parmar C, Hosny A, Aucoin N, Narayan V, et al. Computational radiomics system to decode the radiographic phenotype. Cancer Res. 2017;77:e104–7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  154. Deasy JO, Blanco AI, Clark VH. CERR: a computational environment for radiotherapy research. Med Phys. 2003;30:979–85.

    Article  PubMed  Google Scholar 

  155. Apte AP, Iyer A, Crispin-Ortuzar M, Pandya R, van Dijk LV, Spezi E, et al. Technical note: extension of CERR for computational radiomics: a comprehensive MATLAB platform for reproducible radiomics research. Med Phys. 2018;45:3713–20.

    Article  Google Scholar 

  156. Nioche C, Orlhac F, Boughdad S, Reuzé S, Goya-Outi J, Robert C, et al. LIFEx: a freeware for radiomic feature calculation in multimodality imaging to accelerate advances in the characterization of tumor heterogeneity. Cancer Res. 2018;78:4786–9.

    Article  CAS  PubMed  Google Scholar 

  157. Davatzikos C, Rathore S, Bakas S, Pati S, Bergman M, Kalarot R, et al. Cancer imaging phenomics toolkit: quantitative imaging analytics for precision diagnostics and predictive modeling of clinical outcome. J Med Imaging. 2018;5:011018.

    Article  Google Scholar 

  158. Rathore S, Bakas S, Pati S, Akbari H, Kalarot R, Sridharan P, et al. Brain cancer imaging phenomics toolkit (brain-CaPTk): an interactive platform for quantitative analysis of glioblastoma. In: Crimi A, Bakas S, Kuijf H, Menze B, Reyes M, editors. Brainlesion: Glioma, Multiple Sclerosis, Stroke and Traumatic Brain Injuries. Cham: Springer; 2018. p. 133–45.

    Chapter  Google Scholar 

  159. Götz M, Nolden M, Maier-Hein K. MITK phenotyping: an open-source toolchain for image-based personalized medicine with radiomics. Radiother Oncol. 2019;131:108–11.

    Article  PubMed  Google Scholar 

  160. Fendler WP, Eiber M, Beheshti M, Bomanji J, Ceci F, Cho S, et al. 68Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: version 1.0. Eur J Nucl Med Mol Imaging. 2017;44:1014–24.

    Article  PubMed  Google Scholar 

  161. LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015;521:436–44.

    Article  CAS  PubMed  Google Scholar 

  162. Sahiner B, Pezeshk A, Hadjiiski LM, Wang X, Drukker K, Cha KH, et al. Deep learning in medical imaging and radiation therapy. Med Phys. 2019;46:e1–36.

    Article  PubMed  Google Scholar 

  163. Ronneberger O, Fischer P, Brox T. U-Net: convolutional networks for biomedical image segmentation. Medical Image Computing and Computer-Assisted Intervention (MICCAI). Springer, LNCS, vol. 9351. 2015. p. 234–41.

  164. Milletari F, Navab N, Ahmadi S. V-Net: Fully Convolutional Neural Networks for Volumetric Medical Image Segmentation. 2016 Fourth International Conference on 3D Vision (3DV). IEEE; 2016. p. 565–71.

  165. Isensee F, Petersen J, Klein A, Zimmerer D, Jaeger PF, Kohl S, et al. nnU-Net: Self-adapting Framework for U-Net-Based Medical Image Segmentation. arXiv [cs.CV]. 2018. http://arxiv.org/abs/1809.10486.

  166. Zhu B, Liu JZ, Cauley SF, Rosen BR, Rosen MS. Image reconstruction by domain-transform manifold learning. Nature. 2018;555:487–92.

    Article  CAS  PubMed  Google Scholar 

  167. Zhang C, Bengio S, Hardt M, Recht B, Vinyals O. Understanding deep learning requires rethinking generalization. arXiv [cs.LG]. 2016. http://arxiv.org/abs/1611.03530.

  168. Litjens G, Kooi T, Bejnordi BE, Setio AAA, Ciompi F, Ghafoorian M, et al. A survey on deep learning in medical image analysis. Med Image Anal. 2017;42:60–88.

    Article  PubMed  Google Scholar 

Download references

Acknowledgments

The author thanks Dr Jianhua Yan, Dr Matteo Interlenghi, Dr Francesca Gallivanone, Dr Isabella Castiglioni and Dr Lijun Lu for providing data from their studies for use in the meta-analysis.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alex Zwanenburg.

Ethics declarations

Conflicts of interest

None.

Ethical approval

This article does not describe any studies with human participants performed by the author.

Informed consent

This article describes a meta-analysis on completely anonymous, population-level metrics, and no informed consent was required.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Advanced Image Analyses (Radiomics and Artificial Intelligence).

Electronic supplementary material

ESM 1

(PDF 622 kb)

ESM 2

(CSV 371 kb)

ESM 3

(CSV 1 kb)

ESM 4

(CSV 3 kb)

ESM 5

(CSV 9 kb)

ESM 6

(CSV 21 kb)

ESM 7

(CSV 2 kb)

ESM 8

® 5 kb)

ESM 9

® 2 kb)

ESM 10

(STAN 1 kb)

ESM 11

® 12 kb)

ESM 12

(CSV 4 kb)

ESM 13

(CSV 721 bytes)

ESM 14

(CSV 516 bytes)

ESM 15

(CSV 6 kb)

ESM 16

(CSV 5 kb)

ESM 17

(CSV 4 kb)

ESM 18

(CSV 683 bytes)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zwanenburg, A. Radiomics in nuclear medicine: robustness, reproducibility, standardization, and how to avoid data analysis traps and replication crisis. Eur J Nucl Med Mol Imaging 46, 2638–2655 (2019). https://doi.org/10.1007/s00259-019-04391-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00259-019-04391-8

Keywords

Navigation