Skip to main content
Log in

Automatic extraction of lightweight and efficient neural network architecture of heavy convolutional architectures to predict microsatellite instability from hematoxylin and eosin histology in gastric cancer

  • Original Article
  • Published:
Neural Computing and Applications Aims and scope Submit manuscript

Abstract

Cancers have emerged as a significant concern due to their impact on public health and society. The examination and interpretation of tissue sections stained with Hematoxylin and Eosin (H&E) play a crucial role in disease assessment, particularly in cases like gastric cancer. Microsatellite instability (MSI) is suggested to contribute to the carcinogenesis of specific gastrointestinal tumors. However, due to the nonspecific morphology observed in H&E-stained tissue sections, MSI determination often requires costly evaluations through various molecular studies and immunohistochemistry methods in specialized molecular pathology laboratories. Despite the high cost, international guidelines recommend MSI testing for gastrointestinal cancers. Thus, there is a pressing need for a new diagnostic modality with lower costs and widespread applicability for MSI detection. This study aims to detect MSI directly from H&E histology slides in gastric cancer, providing a cost-effective alternative. The performance of well-known deep convolutional neural networks (DCNNs) and a proposed architecture are compared. Medical image datasets are typically smaller than benchmark datasets like ImageNet, necessitating the use of off-the-shelf DCNN architectures developed for large datasets through techniques such as transfer learning. Designing an architecture proportional to a custom dataset can be tedious and may not yield desirable results. In this work, we propose an automatic method to extract a lightweight and efficient architecture from a given heavy architecture (e.g., well-known off-the-shelf DCNNs) proportional to a specific dataset. To predict MSI instability, we extracted the MicroNet architecture from the Xception network using the proposed method and compared its performance with other well-known architectures. The models were trained using tiles extracted from whole-slide images, and two evaluation strategies, tile-based and whole-slide image (WSI)-based, were employed and compared. Additionally, a visual explanation of the best convolutional neural network model is presented to validate numerical results. The MicroNet architecture achieved the best accuracy (0.85) and area under the curve-receiver operating characteristic curve (0.93), outperforming previous works for the study dataset. The proposed method can be utilized by developers to design lightweight and efficient problem-based neural network architectures, such as MicroNet, for MSI prediction.

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
Algorithm 1
Fig. 3
Fig. 4
Algorithm 2
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12
Fig. 13
Fig. 14
Fig. 15
Fig. 16
Fig. 17
Fig. 18
Fig. 19
Fig. 20
Fig. 21
Fig. 22
Fig. 23
Fig. 24
Fig. 25
Fig. 26
Fig. 27
Fig. 28
Fig. 29

Similar content being viewed by others

Data availability

The datasets analyzed during the current study are available in the zenodo repository, https://zenodo.org/record/2530835#.Y9zqn3YzZPY.

Code availability

Source codes of Micronet and utility methods are available at: https://github.com/habibrostami/stad.

References

  1. Cancer (2022). https://www.who.int/news-room/fact-sheets/detail/cancer. Accessed 02 Feb 2022

  2. Demir C, Yener B (2005) Automated cancer diagnosis based on histopathological images: a systematic survey. Rensselaer Polytech Inst Tech Rep

  3. Ahmedt-Aristizabal D, Armin MA, Denman S, Fookes C, Petersson L (2021) A survey on graph-based deep learning for computational histopathology. Comput Med Imaging Graph 95:102027

    Article  Google Scholar 

  4. van der Laak J, Litjens G, Ciompi F (2021) Deep learning in histopathology: the path to the clinic. Nat Med 27(5):775–784

    Article  Google Scholar 

  5. Cho KO, Lee SH, Jang HJ (2020) Feasibility of fully automated classification of whole slide images based on deep learning. Korean J Physiol Pharmacol 24(1):89–99

    Article  Google Scholar 

  6. Arvaniti E et al (2018) Automated Gleason grading of prostate cancer tissue microarrays via deep learning. Sci Rep 8(1):1–11

    Article  Google Scholar 

  7. Steiner DF et al (2018) Impact of deep learning assistance on the histopathologic review of lymph nodes for metastatic breast cancer. Am J Surg Pathol 42(12):1636

    Article  Google Scholar 

  8. Jang H-J, Song IH, Lee SH (2021) Generalizability of deep learning system for the pathologic diagnosis of various cancers. Appl Sci 11(2):808

    Article  Google Scholar 

  9. Echle A et al (2020) Clinical-grade detection of microsatellite instability in colorectal tumors by deep learning. Gastroenterology 159(4):1406–1416

    Article  Google Scholar 

  10. Li K, Luo H, Huang L, Luo H, Zhu X (2020) Microsatellite instability: a review of what the oncologist should know. Cancer Cell Int 20(1):1–13

    Google Scholar 

  11. Kim YB et al (2016) Microsatellite instability of gastric and colorectal cancers as a predictor of synchronous gastric or colorectal neoplasms. Gut Liver 10(2):220

    Article  Google Scholar 

  12. Russakovsky O et al (2015) Imagenet large scale visual recognition challenge. Int J Comput Vis 115(3):211–252

    Article  MathSciNet  Google Scholar 

  13. Liu W, Wang Z, Liu X, Zeng N, Liu Y, Alsaadi FE (2017) A survey of deep neural network architectures and their applications. Neurocomputing 234:11–26

    Article  Google Scholar 

  14. Rajpurkar P et al. (2017) Chexnet: radiologist-level pneumonia detection on chest x-rays with deep learning. ArXiv Prepr. ArXiv171105225

  15. Shen D, Wu G, Suk H-I (2017) Deep learning in medical image analysis. Annu Rev Biomed Eng 19:221–248

    Article  Google Scholar 

  16. Chen JH, Asch SM (2017) Machine learning and prediction in medicine—beyond the peak of inflated expectations. N Engl J Med 376(26):2507

    Article  Google Scholar 

  17. Liu F et al (2018) Deep learning approach for evaluating knee MR images: achieving high diagnostic performance for cartilage lesion detection. Radiology 289(1):160–169

    Article  MathSciNet  Google Scholar 

  18. A Survey on Deep Reinforcement Learning--«Chinese Journal of Computers» 2018年01期 (2022). http://en.cnki.com.cn/Article_en/CJFDTOTAL-JSJX201801001.htm. Accessed 18 Apr 2022

  19. Hornik K, Stinchcombe M, White H (1989) Multilayer feedforward networks are universal approximators. Neural Netw 2(5):359–366

    Article  Google Scholar 

  20. Zhou J, Xiao D, Zhang M (2019) Feature correlation loss in convolutional neural networks for image classification. In: 2019 IEEE 3rd information technology, networking, electronic and automation control conference (ITNEC), pp 219–223. https://doi.org/10.1109/ITNEC.2019.8729534

  21. Chollet F (2017) Xception: deep learning with depthwise separable convolutions. In: 2017 proceedings of the IEEE conference on computer vision and pattern recognition. CVPR, pp 1800–1807. https://doi.org/10.1109/CVPR.2017.195

  22. Sabour S, Frosst N, Hinton GE (2017) Dynamic routing between capsules. Neural Inf Process Syst. https://doi.org/10.1371/journal.pone.0035195

    Article  Google Scholar 

  23. Alber M et al (2019) iNNvestigate neural networks! J Mach Learn Res 20(93):1–8

    MathSciNet  Google Scholar 

  24. Montavon G, Samek W, Müller K-R (2018) Methods for interpreting and understanding deep neural networks. Digit Signal Process 73:1–15

    Article  MathSciNet  Google Scholar 

  25. Kindermans PJ et al. (2017) Learning how to explain neural networks: patternnet and patternattribution. ArXiv Prepr. ArXiv170505598

  26. Camburu OM (2020) Explaining deep neural networks. ArXiv Prepr. ArXiv201001496

  27. Hildebrand LA, Pierce CJ, Dennis M, Paracha M, Maoz A (2021) Artificial intelligence for histology-based detection of microsatellite instability and prediction of response to immunotherapy in colorectal cancer. Cancers 13(3):391

    Article  Google Scholar 

  28. Pressman IS, Xu H, Kang J, Cha YJ, Lee SH, Hwang TH (2020) Deep learning can predict microsatellite instability from histology in colorectal cancer across different ethnic groups. Cancer Res 80:2100

    Article  Google Scholar 

  29. Yamashita R et al (2021) Deep learning model for the prediction of microsatellite instability in colorectal cancer: a diagnostic study. Lancet Oncol 22(1):132–141

    Article  Google Scholar 

  30. Kather JN (2019) Histological images for MSI vs. MSS classification in gastrointestinal cancer, FFPE samples. ZENODO.

  31. Zhang W et al (2020) MRI-based deep learning analysis can predict microsatellite instability in rectal cancer. SSRN Electron J. https://doi.org/10.2139/ssrn.3569821

    Article  Google Scholar 

  32. Wang T et al. (2020) Microsatellite instability prediction of uterine corpus endometrial carcinoma based on H&E histology whole-slide imaging. In: 2020 IEEE 17th international symposium on biomedical imaging (ISBI), IEEE, pp 1289–1292

  33. Hong R, Liu W, DeLair D, Razavian N, Fenyö D (2021) Predicting endometrial cancer subtypes and molecular features from histopathology images using multi-resolution deep learning models. Cell Rep Med 2(9):100400. https://doi.org/10.1016/j.xcrm.2021.100400

    Article  Google Scholar 

  34. Schmauch B et al (2020) A deep learning model to predict RNA-Seq expression of tumours from whole slide images. Nat Commun 11(1):1–15

    Article  Google Scholar 

  35. Zhu J et al. (2020) Computational analysis of pathological image enables interpretable prediction for microsatellite instability. ArXiv Prepr. ArXiv201003130

  36. Muti HS et al (2021) Development and validation of deep learning classifiers to detect Epstein-Barr virus and microsatellite instability status in gastric cancer: a retrospective multicentre cohort study. Lancet Digit Health 3(10):e654–e664

    Article  Google Scholar 

  37. Kather JN et al (2019) Deep learning can predict microsatellite instability directly from histology in gastrointestinal cancer. Nat Med 25(7):1054–1056. https://doi.org/10.1038/s41591-019-0462-y

    Article  Google Scholar 

  38. Xu TB, Liu CL (2022) Deep neural network self-distillation exploiting data representation invariance. IEEE Trans Neural Netw Learn Syst 33(1):257–269. https://doi.org/10.1109/TNNLS.2020.3027634

    Article  Google Scholar 

  39. Ayinde BO, Inanc T, Zurada JM (2019) Redundant feature pruning for accelerated inference in deep neural networks. Neural Netw 118:148–158. https://doi.org/10.1016/j.neunet.2019.04.021

    Article  Google Scholar 

  40. Xu C, Gao W, Li T, Bai N, Li G, Zhang Y (2023) Teacher-student collaborative knowledge distillation for image classification. Appl Intell 53(2):1997–2009

    Article  Google Scholar 

  41. van Erven T, Harremoes P (2014) Rényi divergence and Kullback-Leibler divergence. IEEE Trans Inf Theory 60(7):3797–3820. https://doi.org/10.1109/TIT.2014.2320500

    Article  Google Scholar 

  42. Anghel A et al (2019) A high-performance system for robust stain normalization of whole-slide images in histopathology. Front Med 6:193

    Article  Google Scholar 

  43. Reinhard E, Adhikhmin M, Gooch B, Shirley P (2001) Color transfer between images. IEEE Comput Graph Appl 21(5):34–41

    Article  Google Scholar 

  44. Khan AM, Rajpoot N, Treanor D, Magee D (2014) A nonlinear mapping approach to stain normalization in digital histopathology images using image-specific color deconvolution. IEEE Trans Biomed Eng 61(6):1729–1738

    Article  Google Scholar 

  45. Vahadane A et al (2016) Structure-preserving color normalization and sparse stain separation for histological images. IEEE Trans Med Imaging 35(8):1962–1971

    Article  Google Scholar 

  46. Macenko M et al. (2009) A method for normalizing histology slides for quantitative analysis. In: 2009 IEEE international symposium on biomedical imaging: from nano to macro, IEEE, pp 1107–1110

  47. The Cancer Genome Atlas - Colorectal Carcinoma Study - National Cancer Institute (2022). https://www.cancer.gov/about-nci/organization/ccg/research/structural-genomics/tcga/studied-cancers/colorectal. Accessed 03 Feb 2022

  48. UNKNOWN (2022) sklearn: a set of python modules for machine learning and data mining. https://pypi.python.org/pypi/scikit-learn/. Accessed 03 Feb 2022

  49. Keras: the Python deep learning API (2022). https://keras.io/. Accessed 03 Feb 2022

  50. TensorFlow (2022). https://www.tensorflow.org/. Accessed 03 Feb 2022

  51. Wang N, Zeng NN, Zhu W (2010) Sensitivity, specificity, accuracy, associated confidence interval and ROC analysis with practical SAS implementations. p 9

  52. Dahiya N, Gupta S, Garg M (2021) Microsatellite instability in gastrointestinal cancer using deep learning: a review. In: IOP conference series: materials science and engineering, IOP Publishing, p 012025

  53. Kuntz S et al (2021) Gastrointestinal cancer classification and prognostication from histology using deep learning: systematic review. Eur J Cancer 155:200–215

    Article  Google Scholar 

  54. Rawla P, Barsouk A (2019) Epidemiology of gastric cancer: global trends, risk factors and prevention. Przeglad Gastroenterol 14(1):26–38. https://doi.org/10.5114/pg.2018.80001

    Article  Google Scholar 

  55. Behzadi-Khormouji H et al (2020) Deep learning, reusable and problem-based architectures for detection of consolidation on chest X-ray images. Comput Methods Progr Biomed 185:105162

    Article  Google Scholar 

  56. Hu B, El Hajj N, Sittler S, Lammert N, Barnes R (2012) Gastric cancer: classification, histology and application of molecular pathology. AJ Gastrointest Oncol 3(3):251–261

    Google Scholar 

  57. Musallam AS, Sherif AS, Hussein MK (2022) Efficient framework for detecting COVID-19 and pneumonia from chest X-ray using deep convolutional network. Egypt Inf J 23:247–257

    Google Scholar 

  58. Mehrotra R, Agrawal R, Ansari MA (2022) Diagnosis of hypercritical chronic pulmonary disorders using dense convolutional network through chest radiography. Multimed Tools Appl 81(6):7625–7649

    Article  Google Scholar 

  59. Hou J, Gao T (2021) Explainable DCNN based chest X-ray image analysis and classification for COVID-19 pneumonia detection. Sci Rep 11(1):1–15

    Article  Google Scholar 

  60. Jin W, Li X, Hamarneh G (2022) Evaluating explainable AI on a multi-modal medical imaging task: can existing algorithms fulfill clinical requirements?

  61. Grün F, Rupprecht C, Navab N, Tombari F (2016) A taxonomy and library for visualizing learned features in convolutional neural networks. ArXiv Prepr. ArXiv160607757

  62. Zhang Y, Tiňo P, Leonardis A, Tang K (2021) A survey on neural network interpretability. IEEE Trans Emerg Top Comput Intell 5:726–742

    Article  Google Scholar 

  63. Bodria F, Giannotti F, Guidotti R, Naretto F, Pedreschi D, Rinzivillo S (2021) Benchmarking and survey of explanation methods for black box models. ArXiv Prepr. ArXiv210213076

  64. Wickramanayake S, Hsu W, Lee ML (2021) Towards fully interpretable deep neural networks: are we there yet?. ArXiv Prepr. ArXiv210613164

  65. Schwalbe G, Finzel B (2021) XAI method properties: a (Meta-) study. ArXiv Prepr. ArXiv210507190

  66. Behzadi-Khormouji H, Rostami H (2021) Fast multi-resolution occlusion: a method for explaining and understanding deep neural networks. Appl Intell 51(4):2431–2455

    Article  Google Scholar 

  67. Zeiler MD, Fergus R (2014) Visualizing and understanding convolutional networks. European conference on computer vision. Springer, Cham, pp 818–833

    Google Scholar 

  68. Ribeiro MT, Singh S, Guestrin C (2016) Why should i trust you? Explaining the predictions of any classifier. In: Proceedings of the 22nd ACM SIGKDD international conference on knowledge discovery and data mining, pp 1135–1144

  69. Petsiuk V, Das A, Saenko K (2018) Rise: randomized input sampling for explanation of black-box models. ArXiv Prepr. ArXiv180607421

  70. Simonyan K, Vedaldi A, Zisserman A (2013) Deep inside convolutional networks: visualising image classification models and saliency maps. ArXiv Prepr. ArXiv13126034

  71. Springenberg JT, Dosovitskiy A, Brox T, Riedmiller M (2014) Striving for simplicity: the all convolutional net. ArXiv Prepr. ArXiv14126806

  72. Zhou B, Khosla A, Lapedriza A, Oliva A, Torralba A (2016) Learning deep features for discriminative localization. In: Proceedings of the IEEE conference on computer vision and pattern recognition, pp 2921–2929

  73. Selvaraju RR, Cogswell M, Das A, Vedantam R, Parikh D, Batra D (2017) Grad-cam: visual explanations from deep networks via gradient-based localization. In: Proceedings of the IEEE international conference on computer vision, pp 618–626

  74. Qi Z, Khorram S, Li F (2019) Visualizing deep networks by optimizing with integrated gradients. In: CVPR workshops

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Habib Rostami.

Ethics declarations

Conflict of interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher's Note

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

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rostami, H., Ashkpour, M., Behzadi-Khormouji, H. et al. Automatic extraction of lightweight and efficient neural network architecture of heavy convolutional architectures to predict microsatellite instability from hematoxylin and eosin histology in gastric cancer. Neural Comput & Applic (2024). https://doi.org/10.1007/s00521-024-09882-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00521-024-09882-w

Keywords

Navigation