簡易檢索 / 詳目顯示

研究生: Nabila Puspita Firdi
Nabila Puspita Firdi
論文名稱: 基於多層注意力的多實例深度學習方法應用於子宮內膜癌H&E 染色全切片影像樣本的腫瘤突變負荷評估和癌症亞型分析
Multilayer Attention based Multiple Instance Deep Learning Methods in application to Tumor Mutational Burden assessment and Cancer Subtyping from H&E stained whole slide images of Endometrial cancer samples
指導教授: 王靖維
Ching-Wei Wang
口試委員: 王靖維
Ching-Wei Wang
許昕
Hsin Hsiu
鄭智嘉
Chih-Chia Cheng
馮輝文
Huei-Wen Ferng
趙載光
Tai-Kuang Chao
學位類別: 碩士
Master
系所名稱: 應用科技學院 - 醫學工程研究所
Graduate Institute of Biomedical Engineering
論文出版年: 2023
畢業學年度: 112
語文別: 英文
論文頁數: 54
外文關鍵詞: Endometrial cancer, Immunotherapy, Tumor mutational burden, Aggressive Cancer, Cancer Subtyping, Deep Learning
相關次數: 點閱:47下載:0
分享至:
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報

  • Endometrial cancer (EC) diagnosis traditionally relies on tumor morphology and nuclear grade, but personalized therapy requires a deeper understanding of tumor mutational burden (TMB), i.e. an established predictive biomarker for immune checkpoint inhibition and has been shown to be a predictive biomarker for response to immunotherapy. However, traditional methods for predicting TMB require sequencing comprehensive panels, exomes, or whole genomes, which are high-cost and not commonly available in healthcare settings. Addressing this challenge, we present three multilayer attention multiple instance deep learning models designed to the classification of aggressive and non-aggressive EC and predict TMB status in the non-aggressive and aggressive ECs using commonly acquired H&E-stained whole slide images. Our models were evaluated on a large dataset of gigapixel histopathology images from The Cancer Genome Atlas (TCGA). The proposed method 1 achieved outstanding performance in classification of the aggressive and non-aggressive ECs, with 97%, 93%, 89% and 89% for area under the receiver operating characteristic curve (AUROC), sensitivity, mean of sensitivity and specificity (MSS) and accuracy, respectively. For TMB prediction in the aggressive ECs, the proposed method 3 achieves 85% and 80% for the sensitivity and AUROC, respectively, and the proposed method 2 obtains 73% and 78% for the MSS and AUROC, respectively.
    In predicting TMB in the non-aggressive ECs, the proposed method 3 also demonstrated the best performance in comparison to the seven SOTA approaches, achieving 76% of sensitivity and 70% of AUROC. Fisher's exact test further validated that the association between the proposed model prediction and the actual cancer subtype or TMB status is both strong (p < 0.01).
    Furthermore, according to the Fisher's exact test, the association between the proposed model prediction and the actual cancer subtype or TMB status in the aggressive group is both extremely strong (p < 0.001), and the association between the prediction of the proposed model 3 and the actual TMB status in the non-aggressive group is strong (p<0.01). Notably, our methods consistently outperformed seven state-of-the-art benchmarked approaches in computational pathology.
    Importantly, according to the Kaplan-Meier survival analysis, the results show that the proposed method 1 successfully differentiates patients with longer disease-specific survival (DSS) and overall survival (OS) with significant difference (p < 0.01 for DSS, p < 0.05 for OS) between the TMB predicted classes in the aggressive EC. These compelling findings highlight the potential of the proposed methods to guide personalized treatment decisions by accurately predicting the EC cancer subtype and the TMB status for effective immunotherapy planning for EC patients. Moreover, a run time analysis demonstrates that the proposed method achieves high efficiency in inference time, taking only 26.21 seconds per slide on average, which makes the proposed methods feasible for practical clinical usage. Additionally, we explored the impact of incorporating a stable factor as a model covariate, resulting in an average performance improvement of 7% and 9% in the MSS and AUROC, respectively. This finding provides valuable insight for researchers aiming to optimize the performance of deep learning-based frameworks in future research.

    Abstract iii Acknowledgement v Table of Content vi List of Tables viii List of Figure x 1 Introduction 1 1.1 Motivation 1 1.2 Aim and Objectives 3 1.3 Contributions 4 1.4 Thesis Organization 5 2 Related Work 6 2.1 The Cancer Genome Atlas Cohort 6 2.2 Deep Learning-based Algorithm 7 2.2.1 ResNet 7 2.2.2 Weakly Supervised Learning Method 9 3 Materials and Method 13 3.1 Materials 13 3.2 Method 15 3.2.1 Tissue Segmentation and Patching 18 3.2.2 Feature Extraction 19 3.2.3 Multilayered Attention Module 20 3.2.4 Stable Covariate Integration and Classification 23 3.2.5 Model Selection with Early Stop Mechanism 24 3.2.6 Implementation Details 25 4 Experiments and Results 27 4.1 Introduction of Experimental Equipment and Evaluation Setup 27 4.1.1 Experimental Equipment 27 4.1.2 Quantitative Analysis Method 27 4.1.3 Statistical Analysis Method 30 4.2 Experimental Results 31 4.2.1 Quantitative evaluation in classification of the aggressive and non-aggressive cancer 31 4.2.2 Quantitative Evaluation in TMB prediction 32 4.2.3 Statistical Analysis 34 4.2.4 Ablation Studies 35 5 Conclusion and Future Work 45 5.1 Conclusion 45 5.2 Future Work 48 Reference 50

    [1] K. He, X. Zhang, S. Ren, and J. Sun, “Deep residual learning for image recognition,” in Proceedings of the IEEE conference on computer vision and pattern recognition, pp. 770–778, 2016.
    [2] O. Raglan, I. Kalliala, G. Markozannes, S. Cividini, M. J. Gunter, J. Nautiyal, H. Gabra, E. Paraskevaidis, P. Martin-Hirsch, K. K. Tsilidis, et al., “Risk factors for endometrial cancer: An umbrella review of the literature,” International journal of cancer, vol. 145, no. 7, pp. 1719–1730, 2019.
    [3] A. Santoro, G. Angelico, A. Travaglino, F. Inzani, D. Arciuolo, M. Valente, N. D’Alessandris, G. Scaglione, V. Fiorentino, A. Raffone, et al., “New pathological and clinical insights in endometrial cancer in view of the updated esgo/estro/esp guidelines,” Cancers, vol. 13, no. 11, p. 2623, 2021.
    [4] J. S. Berek, X. Matias-Guiu, C. Creutzberg, C. Fotopoulou, D. Gaffney, S. Kehoe, K. Lindemann, D. Mutch, N. Concin, F. W. C. C. Endometrial Cancer Staging Subcommittee, et al., “Figo staging of endometrial cancer: 2023,” International Journal of Gynecology & Obstetrics, 2023.
    [5] S. F. Lax, E. S. Pizer, B. M. Ronnett, and R. J. Kurman, “Comparison of estrogen and progesterone receptor, ki-67, and p53 immunoreactivity in uterine endometrioid carcinoma and endometrioid carcinoma with squamous, mucinous, secretory, and ciliated cell differentiation,” Human pathology, vol. 29, no. 9, pp. 924–931, 1998.
    [6] J. V. Bokhman, “Two pathogenetic types of endometrial carcinoma,” Gynecologic oncology, vol. 15, no. 1, pp. 10–17, 1983.
    [7] M. A. Voss, R. Ganesan, L. Ludeman, K. McCarthy, R. Gornall, G. Schaller, W. Wei, and S. Sundar, “Should grade 3 endometrioid endometrial carcinoma be considered a type 2 cancer—a clinical and pathological evaluation,” Gynecologic oncology, vol. 124, no. 1, pp. 15–20, 2012.
    [8] T. de Bortoli, M. Benary, P. Horak, M. Lamping, S. Stintzing, I. Tinhofer, S. Leyvraz, R. Sch ̈afer, F. Klauschen, U. Keller, et al., “Tumour mutational burden and survival with molecularly matched therapy,” European Journal of Cancer, vol. 190, p. 112925, 2023.
    [9] D. T. Rieke, T. de Bortoli, M. Benary, P. Horak, M. Lamping, S. Stintzing, I. Tinhofer, S. Leyvraz, R. Sch ̈afer, F. Klauschen, et al., “Tumor mutational burden as a predictive biomarker for molecularly matched therapy in two independent pan-cancer cohorts.,” 2023.
    [10] W. Cao, X. Ma, J. V. Fischer, C. Sun, B. Kong, and Q. Zhang, “Immunotherapy in endometrial cancer: rationale, practice and perspectives,” Biomarker Research, vol. 9, no. 1, pp. 1–30, 2021.
    [11] S. Lee, O. Lara, H. Karpel, and B. Pothuri, “The association of tumor mutational burden, microsatellite stability, and mismatch repair deficiency in an endometrial cancer patient cohort (194),” Gynecologic Oncology, vol. 166, p. S111, 2022.
    [12] X. Guo, X. Liang, Y. Wang, A. Cheng, H. Zhang, C. Qin, and Z. Wang, “Significance of tumor mutation burden combined with immune infiltrates in the progression and prognosis of advanced gastric cancer,” Frontiers in Genetics, vol. 12, p. 642608, 2021.
    [13] R. T. Lawlor, P. Mattiolo, A. Mafficini, S.-M. Hong, M. L. Piredda, S. V. Taormina, G. Malleo, G. Marchegiani, A. Pea, R. Salvia, et al., “Tumor mutational burden as a potential biomarker for immunotherapy in pancreatic cancer: systematic review and still-open questions,” Cancers, vol. 13, no. 13, p. 3119, 2021.
    [14] L. Fancello, S. Gandini, P. G. Pelicci, and L. Mazzarella, “Tumor mutational burden quantification from targeted gene panels: major advancements and challenges,” Journal for immunotherapy of cancer, vol. 7, pp. 1–13, 2019.
    [15] B. Mel ́endez, C. Van Campenhout, S. Rorive, M. Remmelink, I. Salmon, and N. D’Haene, “Methods of measurement for tumor mutational burden in tumor tissue,” Translational lung cancer research, vol. 7, no. 6, p. 661, 2018.
    [16] R. B ̈uttner, J. W. Longshore, F. L ́opez-R ́ıos, S. Merkelbach-Bruse, N. Normanno, E. Rouleau, and F. Penault-Llorca, “Implementing tmb measurement in clinical practice: considerations on assay requirements,” ESMO open, vol. 4, no. 1, p. e000442, 2019.
    [17] Z. R. Chalmers, C. F. Connelly, D. Fabrizio, L. Gay, S. M. Ali, R. Ennis, A. Schrock, B. Campbell, A. Shlien, J. Chmielecki, et al., “Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden,” Genome medicine, vol. 9, pp. 1–14, 2017.
    [18] A. Sadhwani, H.-W. Chang, A. Behrooz, T. Brown, I. Auvigne-Flament, H. Patel, R. Findlater, V. Velez, F. Tan, K. Tekiela, et al., “Comparative analysis of machine learning approaches to classify tumor mutation burden in lung adenocarcinoma using histopathology images,” Scientific reports, vol. 11, no. 1, p. 16605, 2021.
    [19] Y. Niu, L. Wang, X. Zhang, Y. Han, C. Yang, H. Bai, K. Huang, C. Ren, G. Tian, S. Yin, et al., “Predicting tumor mutational burden from lung adenocarcinoma histopathological images using deep learning,” Frontiers in Oncology, vol. 12, p. 927426, 2022.
    [20] K. Huang, B. Lin, J. Liu, Y. Liu, J. Li, G. Tian, and J. Yang, “Predicting colorectal cancer tumor mutational burden from histopathological images and clinical information using multi-modal deep learning,” Bioinformatics, vol. 38, no. 22, pp. 5108–5115, 2022.
    [21] H. Xu, J. R. Clemenceau, S. Park, J. Choi, S. H. Lee, and T. H. Hwang, “Spatial heterogeneity and organization of tumor mutation burden with immune infiltrates within tumors based on whole slide images correlated with patient survival in bladder cancer,” Journal of Pathology Informatics, vol. 13, p. 100105, 2022.
    [22] M. Y. Lu, T. Y. Chen, D. F. Williamson, M. Zhao, M. Shady, J. Lipkova, and F. Mahmood, “Ai-based pathology predicts origins for cancers of unknown primary,” Nature, vol. 594, no. 7861, pp. 106–110, 2021.
    [23] A. V. Konstantinov and L. V. Utkin, “Multi-attention multiple instance learning,” Neural Computing and Applications, vol. 34, no. 16, pp. 14029–14051, 2022.
    [24] K. Tomczak, P. Czerwi ́nska, and M. Wiznerowicz, “Review the cancer genome atlas (tcga): an immeasurable source of knowledge,” Contemporary Oncology/Wsp ́olczesna Onkologia, vol. 2015, no. 1, pp. 68–77, 2015.
    [25] N. Coudray, P. S. Ocampo, T. Sakellaropoulos, N. Narula, M. Snuderl, D. Feny ̈o, A. L. Moreira, N. Razavian, and A. Tsirigos, “Classification and mutation prediction from non–small cell lung cancer histopathology images using deep learning,” Nature medicine, vol. 24, no. 10, pp. 1559–1567, 2018.
    [26] G. Campanella, M. G. Hanna, L. Geneslaw, A. Miraflor, V. Werneck Krauss Silva, K. J. Busam, E. Brogi, V. E. Reuter, D. S. Klimstra, and T. J. Fuchs, “Clinical-grade computational pathology using weakly supervised deep learning on whole slide images,” Nature medicine, vol. 25, no. 8, pp. 1301–1309, 2019.
    [27] M. Y. Lu, D. F. Williamson, T. Y. Chen, R. J. Chen, M. Barbieri, and F. Mahmood, “Data-efficient and weakly supervised computational pathology on whole-slide images,” Nature biomedical engineering, vol. 5, no. 6, pp. 555–570, 2021.
    [28] C.-W. Wang, Y.-C. Lee, Y.-J. Lin, N. P. Firdi, H. Muzakky, T.-C. Liu, P.-J.Lai, C.-H. Wang, Y.-C. Wang, M.-H. Yu, et al., “Deep learning can predict bevacizumab therapeutic effect and microsatellite instability directly from histology in epithelial ovarian cancer,” Laboratory Investigation, vol. 103, no. 11, p. 100247, 2023.
    [29] Y. Zheng, J. Li, J. Shi, F. Xie, J. Huai, M. Cao, and Z. Jiang, “Kernel attention transformer for histopathology whole slide image analysis and assistant cancer diagnosis,” IEEE Transactions on Medical Imaging, 2023.
    [30] Z. Shao, H. Bian, Y. Chen, Y. Wang, J. Zhang, X. Ji, et al., “Transmil: Transformer based correlated multiple instance learning for whole slide image classification,” Advances in neural information processing systems, vol. 34, pp. 2136–2147, 2021.
    [31] H. Xiang, J. Shen, Q. Yan, M. Xu, X. Shi, and X. Zhu, “Multi-scale representation attention based deep multiple instance learning for gigapixel whole slide image analysis,” Medical Image Analysis, vol. 89, p. 102890, 2023.
    [32] D. N. Louis, M. Feldman, A. B. Carter, A. S. Dighe, J. D. Pfeifer, L. Bry, J. S. Almeida, J. Saltz, J. Braun, J. E. Tomaszewski, et al., “Computational pathology: a path ahead,” Archives of pathology & laboratory medicine, vol. 140, no. 1, pp. 41–50, 2016.
    [33] C.-L. Chen, C.-C. Chen, W.-H. Yu, S.-H. Chen, Y.-C. Chang, T.-I. Hsu, M. Hsiao, C.-Y. Yeh, and C.-Y. Chen, “An annotation-free whole-slide training approach to pathological classification of lung cancer types using deep learning,” Nature communications, vol. 12, no. 1, p. 1193, 2021.
    [34] J. Wang and F. Biljecki, “Unsupervised machine learning in urban studies: A systematic review of applications,” Cities, vol. 129, p. 103925, 2022.
    [35] V. Rani, S. T. Nabi, M. Kumar, A. Mittal, and K. Kumar, “Self-supervised learning: A succinct review,” Archives of Computational Methods in Engineering, vol. 30, no. 4, pp. 2761–2775, 2023.
    [36] C.-W. Wang, Y.-C. Lee, Y.-J. Lin, C.-C. Chang, C.-H. Wang, T.-K. Chao, et al., “Interpretable attention-based deep learning ensemble for personalized ovarian cancer treatment without manual annotations,” Computerized Medical Imaging and Graphics, vol. 107, p. 102233, 2023.
    [37] M. Tu, J. Huang, X. He, and B. Zhou, “Multiple instance learning with graph neural networks,” arXiv preprint arXiv:1906.04881, 2019.
    [38] Y. Xiong, Z. Zeng, R. Chakraborty, M. Tan, G. Fung, Y. Li, and V. Singh, “Nystr ̈omformer: A nystr ̈om-based algorithm for approximating self-attention,” in Proceedings of the AAAI Conference on Artificial Intelligence, vol. 35, pp. 14138–14148, 2021.
    [39] J. Shreffler and M. R. Huecker, “Diagnostic testing accuracy: Sensitivity, specificity, predictive values and likelihood ratios,” 2020.
    [40] D. M. Naeger, M. P. Kohi, E. M. Webb, A. Phelps, K. G. Ordovas, and T. B. Newman, “Correctly using sensitivity, specificity, and predictive values in clinical practice: how to avoid three common pitfalls,” American journal of roentgenology, vol. 200, no. 6, pp. W566–W570, 2013.
    [41] A. G. Glaros and R. B. Kline, “Understanding the accuracy of tests with cutting scores: The sensitivity, specificity, and predictive value model,” Journal of clinical psychology, vol. 44, no. 6, pp. 1013–1023, 1988.
    [42] E. Bolin and W. Lam, “A review of sensitivity, specificity, and likelihood ratios: evaluating the utility of the electrocardiogram as a screening tool in hypertrophic cardiomyopathy,” Congenital Heart Disease, vol. 8, no. 5, pp. 406–410, 2013.
    [43] J. Davis and M. Goadrich, “The relationship between precision-recall and roc curves,” in Proceedings of the 23rd international conference on Machine learning, pp. 233–240, 2006.
    [44] S. Inc, “Spss for windows, rel. 15.0. 1,” 2006.
    [45] H.-Y. Kim, “Statistical notes for clinical researchers: Chi-squared test and fisher’s exact test,” Restorative dentistry & endodontics, vol. 42, no. 2, pp. 152–155, 2017.
    [46] A. Nowacki, “Chi-square and fisher’s exact tests (from the” biostatistics and epidemiology lecture series, part 1”).,” Cleveland Clinic journal of medicine, vol. 84, no. 9 Suppl 2, pp. e20–e25, 2017.
    [47] B. Damato and A. Taktak, “Survival after treatment of intraocular melanoma,”in Outcome Prediction in Cancer, pp. 27–41, Elsevier, 2007.
    [48] M. Kang, H. Song, S. Park, D. Yoo, and S. Pereira, “Benchmarking self-supervised learning on diverse pathology datasets,” CVPR Open Access, 2023.
    [49] H.-Y. Zhou, Y. Yu, C. Wang, S. Zhang, Y. Gao, J. Pan, J. Shao, G. Lu, K. Zhang, and W. Li, “A transformer-based representation-learning model with unified processing of multimodal input for clinical diagnostics,” Nature Biomedical Engineering, pp. 1–13, 2023.
    [50] F. Shamshad, S. Khan, S. W. Zamir, M. H. Khan, M. Hayat, F. S. Khan, and H. Fu, “Transformers in medical imaging: A survey,” Medical Image Analysis, p. 102802, 2023.
    [51] Y. Zhang, J. Wang, J. M. Gorriz, and S. Wang, “Deep learning and vision transformer for medical image analysis,” 2023.
    [52] X. Wang, Y. Du, S. Yang, J. Zhang, M. Wang, J. Zhang, W. Yang, J. Huang, and X. Han, “Retccl: clustering-guided contrastive learning for whole-slide image retrieval,” Medical image analysis, vol. 83, p. 102645, 2023.
    [53] M. Caron, K. Misra, H. Touati, J. Steiner, H. Gurari, M. Uberti, and J. Raison, “Emerging properties in self-supervised vision transformers,” arXiv preprint arXiv:2104.14294, 2021.
    [54] E. Cengil and A. C ̧ ınar, “The effect of deep feature concatenation in the classification problem: an approach on covid-19 disease detection,” International journal of imaging systems and technology, vol. 32, no. 1, pp. 26–40, 2022.
    [55] Y.-J. Kang, S. O’Haire, F. Franchini, M. IJzerman, J. Zalcberg, F. Macrae, K. Canfell, and J. Steinberg, “A scoping review and meta-analysis on the prevalence of pan-tumour biomarkers (dmmr, msi, high tmb) in different solid tumours,” Scientific reports, vol. 12, no. 1, p. 20495, 2022.
    [56] K. Choucair, S. Morand, L. Stanbery, G. Edelman, L. Dworkin, and J. Nemunaitis, “Tmb: a promising immune-response biomarker, and potential spearhead in advancing targeted therapy trials,” Cancer gene therapy, vol. 27, no. 12, pp. 841–853, 2020.
    [57] C. S. Perone and J. Cohen-Adad, “Promises and limitations of deep learning for medical image segmentation,” Journal of Medical Artificial Intelligence, vol. 2, 2019.
    [58] C. R. Boland, S. N. Thibodeau, S. R. Hamilton, D. Sidransky, J. R. Eshleman, R. W. Burt, S. J. Meltzer, M. A. Rodriguez-Bigas, R. Fodde, G. N. Ranzani, et al., “A national cancer institute workshop on microsatellite instability for cancer detection and familial predisposition: development of international criteria for the determination of microsatellite instability in colorectal cancer,” Cancer research, vol. 58, no. 22, pp. 5248–5257, 1998.

    無法下載圖示 全文公開日期 2029/01/30 (校內網路)
    全文公開日期 2029/01/30 (校外網路)
    全文公開日期 2034/01/30 (國家圖書館:臺灣博碩士論文系統)
    QR CODE