請用此 Handle URI 來引用此文件:
http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/98327完整後設資料紀錄
| DC 欄位 | 值 | 語言 |
|---|---|---|
| dc.contributor.advisor | 李素華 | zh_TW |
| dc.contributor.advisor | Su-Hua Lee | en |
| dc.contributor.author | 蔡欣芮 | zh_TW |
| dc.contributor.author | Hsing-Jui Tsai | en |
| dc.date.accessioned | 2025-08-01T16:14:05Z | - |
| dc.date.available | 2025-08-02 | - |
| dc.date.copyright | 2025-08-01 | - |
| dc.date.issued | 2025 | - |
| dc.date.submitted | 2025-07-28 | - |
| dc.identifier.citation | 1. Abuelgasim KA, Jazieh AR. Quality Measures for Multidisciplinary Tumor Boards and Their Role in Improving Cancer Care. Glob J Qual Saf Healthc. 2024 Feb 12;7(1): 28-33.
2. ACC Linee Guida MTB, 2020. 3. Amsden, A. H. (1989). Asia's next giant: South Korea and late industrialization. Oxford University Press. 4. Analysis of the legal and human rights requirements for genomics in and outside the EU, Ref. Ares (2019)2271539 - 29/03/2019 5. Auzanneau C, Bacq D, Bellera C, et al. Feasibility of high-throughput sequencing in clinical routine cancer care: lessons from the cancer pilot project of the France Genomic Medicine 2025 plan. ESMO Open. 2020 Jul;5(4): e000744. 6. Bart Koopman, Harry J.M. et al. Multicenter Comparison of Molecular Tumor Boards in The Netherlands: Definition, Composition, Methods, and Targeted Therapy Recommendations, The Oncologist, Volume 26, Issue 8, August 2021, e1347–e1358. 7. Bartels M, Chibaudel B, et al. Evolving and Improving the Sustainability of Molecular Tumor Boards: The Value and Challenges. Cancers (Basel). 2024 Aug 20;16(16): 2888. 8. Berkman BE, Hull SC. The "right not to know" in the genomic era: time to break from tradition? Am J Bioeth. 2014;14(3): 28-31. 9. Bharadwaj M., Vallurupalli M., Huang F. W. Global precision oncology: a call to action on expanding access to targeted cancer therapies. Oncologist 26, 353‑355 (2021). 10. Bloomberg New Economy International Cancer Coalition; McKinsey Cancer Center; Cure4Cancer. Advancing Global Health Equity in Oncology Clinical Trial Access. Cancer Discov. 2024 Dec 2;14(12): 2317-2323. 11. Boos, L., Wicki, A. The molecular tumor board—a key element of precision oncology. memo 17, 190–193 (2024). 12. Burkard ME, Deming DA, Parsons BM, Kenny PA, Schuh MR, Leal T, et al. Implementation and Clinical Utility of an Integrated Academic-Community Regional Molecular Tumor Board. JCO Precis Oncol. 2017 Jul 5;1: PO.16.00022. 13. Burkard, Mark E., Dustin A. Deming, Benjamin M. Parsons, Paraic A. Kenny, Marissa R. Schuh, Ticiana Leal, Nataliya Uboha, et al. 2017. “Implementation and Clinical Utility of an Integrated Academic-community Regional Molecular Tumor Board.” JCO Precision Oncology 1: 1–10. 14. Cambrosio, A., Campbell, J., & Bourret, P. (2020). Beyond nosology? Molecular tumor boards, singularization, and the conflation of diagnosis and therapy. New Genetics and Society, 40(1), 95–111. 15. Castel, P., Tassy L., Lurkin A., et al. 2012. “Multidisciplinarity and Medical Decision, Impact for Patients with Cancer: Sociological Assessment of Two Tumour Committees' Organization.” Bulletin du Cancer 99, no. 4: E34–E42. 16. Centers for Medicare & Medicaid Services. (2018, March 16). National Coverage Determination (NCD) 90.2: Next generation sequencing (NGS). U.S. Department of Health & Human Services. 17. Centers for Medicare & Medicaid Services. (2018, March 16). National Coverage Determination (NCD) 90.2: Next generation sequencing (NGS). U.S. Department of Health & Human Services.; Daniel M. Sheinson et al. Association Between Medicare's National Coverage Determination and Utilization of Next-Generation Sequencing. JCO Oncol Pract 17, e1774-e1784(2021). 18. Chandra, A., Kao, J., Miller, K. L., Stern, A. D. (2023). Regulatory incentives for innovation: The FDA’s Breakthrough Therapy Designation (NBER Working Paper No. 30712). National Bureau of Economic Research. 19. Chen MH, Li WS, Liao BC, Wu CE, Li CF, Hsieh CH, et al. Expert consensus on molecular tumor boards in Taiwan: Joint position paper by the Taiwan Oncology Society and the Taiwan Society of Pathology. J Cancer Res Pract 2024;11: 18-27. 20. Ciliberto G, De Maria R, et al. A decalogue of Molecular Tumor Board (MTB) recommendations from the CAN.HEAL Consortium. Eur J Cancer. 2025 Jun 3;222: 115433. 21. Ciliberto, G., Canfora, M., Terrenato, I. et al. Bridging therapeutic opportunities: a survey by the Italian molecular tumor board workgroup of Alliance Against Cancer. J Exp Clin Cancer Res 41, 305 (2022). 22. Ciliberto, G., Giacomini, P., Trapani, V., D8.3 MTB Standardised Guidelines., 2025. 23. Collins, F. S., & Varmus, H. (2015). A new initiative on precision medicine. New England Journal of Medicine, 372(9), 793-795. 24. Daniel Hübschmann, Simon Kreutzfeldt, et al. Knowledge Connector: Decision support system for multiomics-based precision oncology. medRxiv 2025.02.23.25322403. 25. Daniel M. Sheinson et al. Association Between Medicare's National Coverage Determination and Utilization of Next-Generation Sequencing. JCO Oncol Pract 17, e1774-e1784(2021). 26. Debyani Chakravarty et al. OncoKB: A Precision Oncology Knowledge Base. JCO Precis Oncol. 2017 Jul;2017: PO.17.00011. 27. Dienstmann R. et al. A framework to rank genomic alterations as targets for cancer precision medicine: the ESCAT. Ann. Oncol. 29, 1895‑1902 (2018). 28. DKTK. (n.d.). The consortium. German Cancer Consortium. https: //dktk.dkfz.de/en/about-us/about-dktk. 29. Dutch Cancer Society, n.d. 30. Eid, M. et al., 156P Molecularly driven therapy recommended by a molecular tumor board: Accessible option or privilege for a minority of patients? A single-center experience from the Czech Republic, ESMO Open, Volume 8, Issue 1, 102005. 31. El-Deiry WS, Bresson C, Wunder F, et al. Worldwide Innovative Network (WIN) Consortium in Personalized Cancer Medicine: Bringing next-generation precision oncology to patients. Oncotarget. 2025 Mar 12;16: 140-162. 32. Erdmann A, Rehmann-Sutter C, Bozzaro C. Patients' and professionals' views related to ethical issues in precision medicine: a mixed research synthesis. BMC Med Ethics. 2021 Aug 31;22(1): 116. 33. European Cancer Organisation. (2023). The European Health Data Space and Cancer: Applying lessons learnt for successful implementation. 34. European Commission, Data privacy framework EU–US, 2023. 35. European Commission. (2025). European Health Data Space Regulation (EHDS): Unlocking the future of health data in Europe. 36. European Parliament & Council of the European Union. (2017). Regulation (EU) 2017/746 of the European Parliament and of the Council of 5 April 2017 on in vitro diagnostic medical devices and repealing Directive 98/79/EC and Commission Decision 2010/227/EU. Official Journal of the European Union, L 117/1. https: //eur-lex.europa.eu/eli/reg/2017/746/oj. 37. European Parliament and Council, Regulation (EU) 2016/679, 2016. 38. Fasola G, Barducci MC et al. Implementation of Precision Oncology in Clinical Practice: Results of a National Survey for Health Care Professionals. Oncologist. 2023 Jun 2;28(6): e324-e330. 39. Fernau S. et al., Zur Rolle und Verantwortung von Ärzten und Forschern in systemmedizinischen Kontexten, Ethik in der Medizin 30, 307-324 (2018). 40. FGM 2025 Workflow Study Group (Alliance nationale des Sciences de la Vie et de la Santé) 41. Filipp FV. Precision medicine driven by cancer systems biology. Cancer Metastasis Rev. 2017 Mar;36(1): 91-108. 42. Forbes SA, Beare D, Boutselakis H et al. COSMIC: Somatic cancer genetics at high-resolution. Nucleic Acids Res, 2017;45: D777–D783. 43. Fountzilas E, Said R, Tsimberidou AM. Expanded access to investigational drugs: balancing patient safety with potential therapeutic benefits. Expert Opin Investig Drugs. 2018 Feb;27(2): 155-162. 44. Gao J, Aksoy BA, Dogrusoz U et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6: pl1. 45. German Human Genome‑Phenome Archive. (2025, July 9). About us. https: //www.ghga.de. 46. Ghoreyshi N, Heidari R, Farhadi A, Chamanara M, Farahani N, Vahidi M, Behroozi J. Next-generation sequencing in cancer diagnosis and treatment: clinical applications and future directions. Discov Oncol. 2025 Apr 20;16(1): 578. 47. Ginsburg GS, Phillips KA. Precision Medicine: From Science to Value. Health Aff (Millwood). 2018 May;37(5): 694-701. 48. Ginsburg, G. S., & Phillips, K. A. (2018). Precision medicine: From science to value. Health Affairs, 37(5), 694–701. 49. Gladstone BP, Beha J, Hakariya A, Missios P, Malek NP, Bitzer M. Systematic review and meta-analysis of molecular tumor board data on clinical effectiveness and evaluation gaps. NPJ Precis Oncol. 2025 Apr 2;9(1): 96. 50. Grossman RL, Heath AP, Ferretti V, Varmus HE, Lowy DR, Kibbe WA, Staudt LM. Toward a Shared Vision for Cancer Genomic Data. N Engl J Med. 2016 Sep 22;375(12):1109-12. 51. Haj Mohammad SF, Timmer HJL, Zeverijn LJ, Geurts BS, Spiekman IAC, Verkerk K, Verbeek FAJ, Verheul HMW, Voest EE, Gelderblom H. The evolution of precision oncology: The ongoing impact of the Drug Rediscovery Protocol (DRUP). Acta Oncol. 2024 May 23;63: 368-372. 52. Hajdu, S. I., and Vadmal M. 2013. “A Note from History: Landmarks in History of Cancer, Part 6.” Cancer 119, no. 23: 4058–4082. 53. Hamamoto R, Koyama T, Kouno N, Yasuda T, Yui S, Sudo K, et al. Introducing AI to the molecular tumor board: one direction toward the establishment of precision medicine using large-scale cancer clinical and biological information. Exp Hematol Oncol. 2022 Oct 31;11(1): 82. 54. Hart, D. Haftungsrecht und Standardbildung in der modernen Medizin: e: med und Probleme der Definition des Standards. MedR 34, 669–675 (2016). 55. Hartwig Medical Foundation, https: //catalog.hartwigmedicalfoundation.nl/. 56. Hartwig Medical Foundation, https: //www.hartwigmedicalfoundation.nl/over-ons/anbi/. 57. Health Insurance Review & Assessment Service (HIRA). (2020). 2019 National health insurance statistical yearbook. HIRA. 58. Hofmann D, Esposito E. The Role of Algorithms in Molecular Tumour Boards-Managing the Gap Between Research and Clinic in Precision Medicine. Sociol Health Illn. 2025 May;47(4). 59. https: //nbct.nhri.org.tw/fileRename/fileRename.aspx?uid=24&fid=997&kid=2&site_id=0. 60. Hunter N, Dempsey N, Tbaishat F, Jahanzeb M, Al-Sukhun S, Gralow JR. Resource-Stratified Guideline-Based Cancer Care Should Be a Priority: Historical Context and Examples of Success. Am Soc Clin Oncol Educ Book. 2020 Mar;40: 1-10. 61. Institute of Medicine (US) Roundtable on Evidence-Based Medicine. The Learning Healthcare System: Workshop Summary. Olsen L, Aisner D, McGinnis JM, editors. Washington (DC): National Academies Press (US); 2007. 62. Jain NM, Schmalz L, Cann C, et al. Framework for Implementing and Tracking a Molecular Tumor Board at a National Cancer Institute-Designated Comprehensive Cancer Center. Oncologist. 2021 Nov;26(11): e1962-e1970. 63. Jang M, Pak HY, Heo JY, Lim H, Choi YL, Shim HS, Kim EK. Trends and Clinical Characteristics of Next-Generation Sequencing-Based Genetic Panel Tests: An Analysis of Korean Nationwide Claims Data. Cancer Res Treat. 2024 Jan;56(1): 27-36. doi: 10.4143/crt.2023.844. 64. Jennings, L. J. et al. Guidelines for validation of next-generation sequencing-based oncology panels: a Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists. J. Mol. Diagn. 19, 341–365 (2017). 65. Thiery J, Fahrner M. Integration of proteomics in the molecular tumor board. Proteomics. 2024 Jun;24(12-13): e2300002. 66. Jørgensen JT. Companion diagnostics: the key to personalized medicine. Foreword. Expert Rev Mol Diagn. 2015 Feb;15(2): 153-6. 67. Kaissis, G. A., Makowski, M. R., Rückert, D., & Braren, R. F. (2020). Secure, privacy-preserving and federated machine learning in medical imaging. Nature Machine Intelligence, 2(6), 305–311. 68. Kim SY, Kim JH, Kim TY, et al. Pragmatic nationwide master observational trial based on genomic alterations in advanced solid tumors: Korean Precision Medicine Networking Group Study of Molecular profiling guided therapy based on genomic alterations in advanced Solid tumors (KOSMOS)-II study protocol KCSG AL-22-09. BMC Cancer. 2024 May 9;24(1):574. 69. Kim TY, Kim SY, et al. Nationwide precision oncology pilot study: KOrean Precision Medicine Networking Group Study of Molecular profiling-guided therapy based on genomic alterations in advanced solid tumors (KOSMOS) KCSG AL-20-05. ESMO Open. 2024 Oct;9(10): 103709. 70. Kim Yun-mi, Experts call for equal insurance coverage of NGS testing for breast and lung cancer., Korea Biomedical Review, 2025. 71. Kingham TP, Alatise OI, Vanderpuye V, Casper C, Abantanga FA, Kamara TB, Olopade OI, Habeebu M, Abdulkareem FB, Denny L. Treatment of cancer in sub-Saharan Africa. Lancet Oncol. 2013 Apr;14(4): e158-67. 72. Knoppers BM, Thorogood A, Chadwick R. The Human Genome Organisation: towards next-generation ethics. Genome Med. 2013 Apr 29;5(4):38. 73. Knoppers BM, Zawati MH, Kirby ES. Sampling populations of humans across the world: ELSI issues. Annu Rev Genomics Hum Genet. 2012;13: 395-413. 74. Konukiewitz, B., Kellers, F., Nettersheim, A. et al. Das Molekulare Tumorboard. Onkologie 30, 916–921 (2024). 75. Kumar A, Owen JR, Sloat NT, Maynard E, Hill VM, Hubbard CB, McKinney MS, Sutton LM, McCall SJ, Datto MB, et al. Expansion of an Academic Molecular Tumor Board to Enhance Access to Biomarker-Driven Trials and Therapies in the Rural Southeastern United States. Current Oncology. 2024; 31(11): 7244-7257. 76. Kurtzer GM, Sochat V, Bauer MW. Singularity: Scientific containers for mobility of compute. PLoS One. 2017 May 11;12(5): e0177459. 77. Landrum MJ, Lee JM, Riley GR et al. ClinVar: Public archive of relationships among sequence variation and human phenotype. Nucleic Acids Res 2014;42: D980–D985. 78. Lee D, Kim K. Public R&D Projects-Based Investment and Collaboration Framework for an Overarching South Korean National Strategy of Personalized Medicine. Int J Environ Res Public Health. 2022 Jan 24;19(3): 1291. 79. Lee Y, Lee S, Sung JS, Chung HJ, Lim AR, Kim JW, Choi YJ, Park KH, Kim YH. Clinical Application of Targeted Deep Sequencing in Metastatic Colorectal Cancer Patients: Actionable Genomic Alteration in K-MASTER Project. Cancer Res Treat. 2021 Jan;53(1): 123-130. 80. Li, M. M. et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a Joint Consensus Recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J. Mol. Diagn. 19, 4–23 (2017). 81. Linee Guida per l’istituzione e la gestione dei Molecular Tumor Board negli Istituti di Alleanza Contro il Cancro. Gennaro Ciliberto, ALLEANZA CONTRO IL CANCRO | 2020. 82. Luchini C, Lawlor RT, Milella M, Scarpa A. Molecular Tumor Boards in Clinical Practice. Trends Cancer. 2020 Sep;6(9): 738-744. 83. Luthra R, Patel KP, Routbort MJ, Broaddus RR, Yau J, Simien C, Chen W, Hatfield DZ, Medeiros LJ, Singh RR. A Targeted High-Throughput Next-Generation Sequencing Panel for Clinical Screening of Mutations, Gene Amplifications, and Fusions in Solid Tumors. J Mol Diagn. 2017 Mar;19(2): 255-264. 84. Lutz S, D'Angelo A, Hammerl S, Schmutz M, Claus R, Fischer NM, Kramer F, Hammoud Z. Unveiling the Digital Evolution of Molecular Tumor Boards. Target Oncol. 2025 Jan;20(1): 27-43. 85. Mateo J, Chakravarty D, Dienstmann R, et al. A framework to rank genomic alterations as targets for cancer precision medicine: the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT). Ann Oncol. 2018 Sep 1;29(9): 1895-1902. 86. Matteo Pallocca, Martina Betti, Sara Baldinelli, Ramona Palombo, Gabriele Bucci, Luca Mazzarella, Giovanni Tonon, Gennaro Ciliberto, Clinical bioinformatics desiderata for molecular tumor boards, Briefings in Bioinformatics, Volume 25, Issue 5, September 2024, bbae447. 87. Mattingly TJ 2nd, Simoni-Wastila L. Patient-Centered Drug Approval: The Role of Patient Advocacy in the Drug Approval Process. J Manag Care Spec Pharm. 2017 Oct;23(10): 1078-1082. 88. Merkel D, Docker: lightweight Linux containers for consistent development and deployment. Linux J. 2014, 239, Article 2 (March 2014). 89. Merry D, Schickhardt C, Mehlis K, Winkler EC. Trust and responsibility in molecular tumour boards. Bioethics. 2018 Sep;32(7): 464-472. 90. Mikko Nieminen, Oliver Stolpe, Mathias Kuhring, January Weiner, Patrick Pett, Dieter Beule, Manuel Holtgrewe, SODAR: managing multiomics study data and metadata, GigaScience, Volume 12, 2023, giad052. 91. Miller RW, Hutchcraft ML, Weiss HL, Wu J, Wang C, Liu J, et al. Molecular Tumor Board-Assisted Care in an Advanced Cancer Population: Results of a Phase II Clinical Trial. JCO Precis Oncol. 2022 Aug;6: e2100524. 92. Molecular Tumor Boards: Realizing Precision Oncology Therapy, Clinical Pharmacology & Therapeutics Volume 103, Issue 2 pp. 206-209. 93. Moore, David Allan, Marina Kushnir, Gabriel Mak, Helen Winter, Teresa Curiel, Mark Voskoboynik, Michele Moschetta, et al. 2019. Prospective Analysis of 895 Patients on a UK Genomics Review Board.” ESMO Open 4 (2): e000469. 94. Morash M, Mitchell H, Beltran H, Elemento O, Pathak J. The role of next-generation sequencing in precision medicine: a review of outcomes in oncology. J Pers Med. 2018;8(3): 30. 95. Mosele F, Remon J, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020 Nov;31(11): 1491-1505. 96. National Academies of Sciences, Engineering, and Medicine; Health and Medicine Division; Board on Health Care Services; Board on the Health of Select Populations; Committee on the Evidence Base for Genetic Testing. An Evidence Framework for Genetic Testing. Washington (DC): National Academies Press (US); 2017 Mar 27. 97. National Coverage Determination 90.2, NCD 90.2. 98. National Institutes of Health, Cancer Research Data Commons (CRDC). https: //datascience.cancer.gov/collaborations/cancer-research-data-commons-crdc. 99. Nelson A. M., Milner D. A., Rebbeck T. R., Iliyasu Y. Oncologic care and pathology resources in Africa: survey and recommendations. J. Clin. Oncol. 34, 20‑26 (2016). 100. Nichetti F, Brambilla M et al. Real-World Outcomes of Molecular Tumor Board Treatment Recommendations. JCO Precis Oncol 9, e2400387(2025). 101. O'Dwyer PJ, Gray RJ, et al. The NCI-MATCH trial: lessons for precision oncology. Nat Med. 2023 Jun;29(6): 1349-1357. 102. Organisation for Economic Co‑operation and Development, “OECD Recommendation on AI”, 2021. 103. Organisation for Economic Co‑operation and Development, Recommendation on Health Data Governance, 2017. 104. Pagès A, Foulon S, Zou Z, Lacroix L, Lemare F, de Baère T, Massard C, Soria JC, Bonastre J. The cost of molecular-guided therapy in oncology: a prospective cost study alongside the MOSCATO trial. Genet Med. 2017 Jun;19(6):683-690. 105. Pallocca M, Betti M, Baldinelli S, Palombo R, Bucci G, Mazzarella L, Tonon G, Ciliberto G. Clinical bioinformatics desiderata for molecular tumor boards. Brief Bioinform. 2024 Jul 25;25(5): bbae447. 106. Pam K. Mangat et al. Rationale and Design of the Targeted Agent and Profiling Utilization Registry Study. JCO Precis Oncol 2, 1-14(2018). 107. Patterson SE, Liu R, Statz CM et al. The clinical trial landscape in oncology and connectivity of somatic mutational profiles to targeted therapies. Hum Genomics 2016;10: 4. 108. Pixberg C, Schulze M et al. Reimbursement in the Context of Precision Oncology Approaches in Metastatic Breast Cancer: Challenges and Experiences. Breast Care (Basel). 2024 Feb;19(1): 10-17. 109. Pope DG. Reacting to ranki NGS: evidence from "America's Best Hospitals". J Health Econ. 2009 Dec;28(6): 1154-65. 110. Porter ME, Teisberg EO. Redefining competition in health care. Harv Bus Rev. 2004 Jun;82(6): 64-76, 136. 111. PubMed. U.S. National Library of Medicine Web site. Available at https: //pubmed.ncbi.nlm.nih.gov/. 112. Ray, Turna. 2019.“Survey of Precision Oncology Programs Finds Agreement on Testing, Divergence in Care Delivery. ”Precision Oncology News, July 19. https: //www.precisiononcologynews.com/cancer/survey-precision-oncology-programs-finds-agreement-testing-divergence-care-delivery#.XdBbS797lR0. 113. Rehm HL, Page AJH, Smith L, et al. GA4GH: International policies and standards for data sharing across genomic research and healthcare. Cell Genom. 2021 Nov 10;1(2):100029. 114. Reisinger E, Genthner L, et al. OTP: An automatized system for managing and processing NGS data. J Biotechnol. 2017 Nov 10;261: 53-62. 115. Relling, M., Evans, W. Pharmacogenomics in the clinic. Nature 526, 343–350 (2015). https: //doi.org/10.1038/nature15817. 116. Richards S, Aziz N, Bale S, et al.; ACMG Laboratory Quality Assurance Committee. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015 May;17(5): 405-24. 117. Riely GJ, Wood DE, et al. Non-Small Cell Lung Cancer, Version 4.2024, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2024 May;22(4): 249-274. 118. Rouven Hoefflin et al. Personalized Clinical Decision Making Through Implementation of a Molecular Tumor Board: A German Single-Center Experience. JCO Precis Oncol 2, 1-16(2018). 119. Roychowdhury, S., Iyer M. K., Robinson D. R., et al. 2011. “Personalized Oncology Through Integrative High‐Throughput Sequencing: A Pilot Study.” Science Translational Medicine 3, no. 111: 111ra121. 120. Rulten SL, Grose RP, Gatz SA, Jones JL, Cameron AJM. The Future of Precision Oncology. Int J Mol Sci. 2023 Aug 9;24(16): 12613. 121. Schickhardt C. et al., Do patients and research subjects have a right to receive their genomic raw data? An ethical and legal analysis. BMC Medical Ethics. (2020) 21: 7 122. Shirota H, Komine K, Takahashi M, Takahashi S, Miyauchi E, Niizuma H, et al. Clinical decisions by the molecular tumor board on comprehensive genomic profiling tests in Japan: A retrospective observational study. Cancer Med. 2023 Mar;12(5): 6170-6181. 123. Soohyeon Lee, Clinical Implication of Molecular Tumor Board, Korean J Med 2022;97(5): 319-322. 124. Stiglitz, J. 1999. “Knowledge as a Global Public Good,” in Kaul, editor, Grunberg, editor and Stern, editor. (1999), Eds., 308-325. 125. Stoeklé HC, Mamzer-Bruneel MF, Frouart CH, Le Tourneau C, Laurent-Puig P, Vogt G, Hervé C. Molecular Tumor Boards: Ethical Issues in the New Era of Data Medicine. Sci Eng Ethics. 2018 Feb;24(1): 307-322. 126. Tamborero, D. et al. The Molecular Tumor Board Portal supports clinical decisions and automated reporting for precision oncology. Nat. Cancer 3, 251–261 (2022). 127. Ten years of Hartwig Medical Foundation: a retrospect and a glimpse of the future, Hartwig Medical Foundation, 11-04-2025. 128. The White House. (2015, January 30). Fact sheet: President Obama’s Precision Medicine Initiative. https: //obamawhitehouse.archives.gov/the-press-office/2015/01/30/fact-sheet-president-obama-s-precision-medicine-initiative. 129. The White House. (2016, October 17). Fact sheet: Vice President Biden delivers Cancer Moonshot Task Force report. https: //obamawhitehouse.archives.gov/the-press-office/2016/10/17/fact-sheet-vice-president-biden-delivers-cancer-moonshot-report. 130. Tsimberidou, A.M., Kahle, M., Vo, H.H. et al. Molecular tumour boards - current and future considerations for precision oncology. Nat Rev Clin Oncol 20, 843–863 (2023). 131. U.S. Food & Drug Administration. (2017, November 30). FDA grants marketing approval to FoundationOne CDx in vitro diagnostic test. 132. U.S. Food and Drug Administration, 2018. 133. van de Haar, Joris, Louisa Hoes, and Emile Voest. 2019. “Advancing Molecular Tumour Boards: Highly Needed to Maximise the Impact of Precision Medicine.” ESMO Open 4 (2): e000516. 134. van der Velden DL, Hoes LR, van der Wijngaart H, et al. The Drug Rediscovery protocol facilitates the expanded use of existing anticancer drugs. Nature. 2019 Oct;574(7776): 127-131. 135. van Waalwijk van Doorn-Khosrovani, S. et al.,1723P Improving access to molecular tumour boards for complex genomic profiles: A healthcare policy from the Netherlands, Annals of Oncology, Volume 34, S938. 136. Vassal, G. 2010. “L’IGR Lance Sa Première RCP Moléculaire.” Oncomagazine 4, no. 3: 6. 137. Wahida A, Kurzrock R. The Molecular Tumor Board Turns 10: The Age of Complexity. Oncologist. 2025 Mar 10;30(3): oyae271. 138. Westphalen, C. B., Boscolo Bielo, L., Aftimos, P. et al. ESMO Precision Oncology Working Group recommendations on the structure and quality indicators for molecular tumour boards in clinical practice. Annals of Oncology, 36(6), 614–625(2025). 139. Whole Genome Sequencing: Global Markets, BCC Research,June 25,2025,81. https: //www.giiresearch.com/report/bc1761723-whole-genome-sequencing-global-markets.html. 140. World Health Organization, “Ethics and Governance of AI for Health”, 2021. 141. Wright, F. C., De Vito C., Langer B., and Hunter A. 2007. “Multidisciplinary Cancer Conferences: A Systematic Review and Development of Practice Standards.” European Journal of Cancer 43, no. 36: 1002–1010. 142. Zeng, J. et al. Operationalization of Next-Generation Sequencing and Decision Support for Precision Oncology. JCO Clin Cancer Inform. 2019 Sep;3: 1-12. 143. 合作示範計畫打造我國本土癌症基因庫促次世代定序納健保, Rti 中央廣播電臺。https: //web01.rti.org.tw/news/view/id/22058173 144. 次世代基因定序檢測(NGS)專區,中央健康保險署, 2024。https: //www.nhi.gov.tw/ch/np-3636-1.html. 145. 國家人體生物資料庫整合平台。nbct.nhri.org.tw 146. 國家級人體生物資料庫整合平台(NBCT)向所有參與者致敬,國家衛生研究院電子報,1042期, 院務紀事。 147. 產官學合作迎來次世代定序給付,分子腫瘤委員會助力癌症精準治療, GeneOnline News,由羅氏大藥廠贊助,專文代碼 M-TW-00004301。https: //geneonline.news/roche-2023-mgto-MTB-precision-medicine-1/ 148. 癌症問康健,2023/12/07。https: //cancer.commonhealth.com.tw/article/1041 149. 癌症精準醫療與分子腫瘤委員會實踐,第六屆臺灣藥學聯合學術研討會, November 16, 2024,奇美醫院,蘇慧真。 150. 總統府新聞稿, 2023。president.gov.tw. 151. 羅氏併購 Foundation Medicine 為求基因檢測對「癌」下藥,今周刊, 2018/12/26。https: //www.businesstoday.com.tw/article/category/80392/post/201812260028/ | - |
| dc.identifier.uri | http://tdr.lib.ntu.edu.tw/jspui/handle/123456789/98327 | - |
| dc.description.abstract | 本研究聚焦於分子腫瘤委員會(molecular tumor board, MTB)在精準醫療時代下的治理樣貌與永續發展,旨在釐清其制度設計關鍵與最佳實踐模式。研究採用比較個案研究法與文件分析法,選取美國、歐盟(以荷蘭為代表)與韓國三大國家作為典範案例,從政策架構、產業參與與臨床實務等層面進行制度比較,並進一步映射至臺灣情境,探討其借鏡價值與制度移植的可行性。
分析結果顯示,三國分別發展出「市場導向生態系」(美國)、「公私協力網絡」(荷蘭)與「國家戰略競速」(韓國)三種差異化制度模式,反映出各自政治經濟背景與治理邏輯。然而,無論模式為何,皆面臨變異詮釋標準不一、藥物可近性不均與品質監管缺口等共通挑戰。對照國際經驗,臺灣具備全民健保、高品質醫療體系與國家級生醫資料平台等制度優勢,卻同時面臨「診斷-治療斷鏈」、跨域人才短缺與法規真空等結構性障礙,導致分子腫瘤委員會制度難以發揮其應有效能。 為回應上述挑戰,本研究提出「混合型策略」作為臺灣未來發展分子腫瘤委員會制度之優化途徑:一方面應建立國家級數據治理中心與條件式給付機制,以縮減實證與給付間之落差;另一方面,透過病患倡議與產業創新導入更多治療選項,提升制度彈性與臨床可行性。同時,建議推動分子腫瘤委員會品質指標與責任階梯化模型之制定,形塑問責明確、透明治理的永續制度設計。 本研究認為,分子腫瘤委員會的成功關鍵不僅在於技術導入,更取決於制度設計的完整性與治理機制的韌性。本研究對臨床實務端提供跨領域人才培育與流程優化之參考建議,對政策制定端則勾勒出監管強化與財務永續並行的改革藍圖,期盼能為臺灣乃至其他全民健保國家推動精準腫瘤醫學發展提供具體借鏡與策略參考。 | zh_TW |
| dc.description.abstract | This study focuses on the governance and sustainability of molecular tumor boards (MTBs) in the era of precision medicine, aiming to identify the institutional design elements and best practices critical to their success. Employing a comparative case study methodology and documentary analysis, the research examines three representative national models-the market-driven ecosystem of the United States, the public-private collaborative network of the Netherlands, and South Korea’s state-led strategic initiative-analyzing their policy frameworks, industrial dynamics, and clinical implementation. These international insights are then mapped onto the Taiwanese context to assess the feasibility of institutional transfer and adaptation.
Findings reveal that each country has developed a distinct governance model reflecting its unique sociopolitical and healthcare systems. However, common challenges persist across all cases, including inconsistent standards for variant interpretation, unequal access to targeted therapies, and a lack of quality oversight mechanisms. In comparison, Taiwan benefits from strong institutional foundations, such as a universal health insurance system, high-quality healthcare services, and a national biomedical data infrastructure. Nonetheless, it faces structural barriers-most notably the diagnostic–therapeutic gap, shortages in cross-disciplinary expertise, and regulatory ambiguity-which hinder the full functionality of its MTB framework. In response, this study proposes a hybrid strategy as the optimal pathway for advancing MTB governance in Taiwan. Key recommendations include establishing a centralized data governance hub and implementing conditional reimbursement policies to bridge the evidence–reimbursement divide; encouraging patient advocacy and fostering industry-driven innovation to diversify treatment access; and developing standardized quality metrics and a tiered accountability model to promote transparent and sustainable institutional design. The study concludes that the effectiveness of MTBs depends not only on technological capabilities but, more importantly, on coherent system architecture, adequate resource allocation, and the establishment of a learning-oriented healthcare framework. It provides practical recommendations for healthcare institutions regarding talent development and process optimization, while outlining a policy roadmap that balances regulatory robustness with financial sustainability. The findings aim to serve as a strategic reference for Taiwan and other countries with single-payer systems seeking to advance precision oncology. | en |
| dc.description.provenance | Submitted by admin ntu (admin@lib.ntu.edu.tw) on 2025-08-01T16:14:05Z No. of bitstreams: 0 | en |
| dc.description.provenance | Made available in DSpace on 2025-08-01T16:14:05Z (GMT). No. of bitstreams: 0 | en |
| dc.description.tableofcontents | 口試委員會審定書 i
誌謝 ii 中文摘要 iv ABSTRACT v 目次 vii 圖次 x 表次 xi 第一章 緒論 1 第一節 研究背景 1 第二節 研究動機與目的 4 第一項 研究動機 4 第二項 研究目的 7 第三節 研究架構與方法 9 第二章 分子腫瘤委員會制度與精準醫療之理論基礎 12 第一節 次世代定序(next-generation sequencing, NGS)與精準醫療發展 12 第二節 分子腫瘤委員會的組成、功能與運作模式 16 第一項 組成:跨領域的專業整合 16 第二項 功能:從數據詮釋到臨床決策支援 18 第三項 運作模式:標準化的決策流程 23 第四項 分子腫瘤委員會之全球發展趨勢 27 第三節 法規與倫理議題:責任歸屬與數據治理 31 第一項 國際指導原則與規範框架 31 第二項 決策責任的階梯化歸屬 36 第三項 數據隱私與治理挑戰 39 第四項 病患參與及知情同意之倫理挑戰 42 第三章 國際分子腫瘤委員會制度比較分析:政策、產業與實踐 46 第一節 政策與保險覆蓋作為分子腫瘤委員會發展的關鍵驅動力 46 第二節 實務案例比較分析 47 第一項 美國模式:市場、產業與法規共同驅動的生態系 48 第二項 歐盟模式:倫理框架與公衛網絡驅動-以荷蘭為例 56 第三項 韓國模式:國家意志與健保給付驅動 63 第三節 跨國多維度比較與制度根源分析:三大典範的比較性解釋 68 第四章 臺灣分子腫瘤委員會制度與現況分析 74 第一節 推動歷程與政策背景 74 第一項 制度起源與規劃背景 74 第二項 發展歷程與重要里程碑 76 第二節 現況分析 80 第一項 分子腫瘤委員會之組織與運作現況 80 第二項 醫院設置情形與成果 84 第三節 臺灣分子腫瘤委員會面臨之挑戰 86 第一項 政府與醫療機構的角色定位 87 第二項 健保與自費制度 88 第三項 對診斷流程與醫院營運之影響 89 第四項 跨領域整合與臨床應用成效 90 第五項 法規與倫理挑戰 91 第五章 比較分析與政策建議(臺灣分子腫瘤委員會機遇與挑戰) 95 第一節 國際典範的總結與啟示 95 第二節 臺灣模式的定位與策略性課題 97 第一項 綜合評估:臺灣的利基與發展困境 97 第二項 國際經驗的策略性應用 98 第三節 制度優化與發展建議:邁向永續的臺灣模式 100 第一項 組織架構與標準作業流程明確化 100 第二項 具體政策工具創新 101 第三項 資源整合與永續發展策略 103 第四項 倫理與法律規範建議 104 第五項 政策制度創新與未來展望 106 第六章 結論與研究展望 108 第一節 研究發現總結 108 第二節 實務與政策貢獻 109 第三節 後續研究建議 111 參考文獻 114 | - |
| dc.language.iso | zh_TW | - |
| dc.subject | 分子腫瘤委員會 | zh_TW |
| dc.subject | 精準醫療 | zh_TW |
| dc.subject | 醫療政策 | zh_TW |
| dc.subject | 制度治理 | zh_TW |
| dc.subject | 次世代定序 | zh_TW |
| dc.subject | MTB | en |
| dc.subject | NGS | en |
| dc.subject | Next Generation Sequencing | en |
| dc.subject | Institutional Governance | en |
| dc.subject | Health Policy | en |
| dc.subject | Precision Medicine | en |
| dc.subject | Molecular Tumor Board | en |
| dc.title | 分子腫瘤委員會制度之研究:精準醫療實踐下的挑戰、機會與未來發展 | zh_TW |
| dc.title | Exploring the Molecular Tumor Board(MTB) System:Challenges, Opportunities, and Prospects in Precision Medicine Practice | en |
| dc.type | Thesis | - |
| dc.date.schoolyear | 113-2 | - |
| dc.description.degree | 碩士 | - |
| dc.contributor.oralexamcommittee | 張濱璿;吳全峰 | zh_TW |
| dc.contributor.oralexamcommittee | Pin-Hsuan Chang;Chuan-Feng Wu | en |
| dc.subject.keyword | 分子腫瘤委員會,精準醫療,醫療政策,制度治理,次世代定序, | zh_TW |
| dc.subject.keyword | Molecular Tumor Board,MTB,Precision Medicine,Health Policy,Institutional Governance,Next Generation Sequencing,NGS, | en |
| dc.relation.page | 126 | - |
| dc.identifier.doi | 10.6342/NTU202502482 | - |
| dc.rights.note | 未授權 | - |
| dc.date.accepted | 2025-07-30 | - |
| dc.contributor.author-college | 進修推廣學院 | - |
| dc.contributor.author-dept | 生物科技管理碩士在職學位學程 | - |
| dc.date.embargo-lift | N/A | - |
| 顯示於系所單位: | 生物科技管理碩士在職學位學程 | |
文件中的檔案:
| 檔案 | 大小 | 格式 | |
|---|---|---|---|
| ntu-113-2.pdf 未授權公開取用 | 2.31 MB | Adobe PDF |
系統中的文件,除了特別指名其著作權條款之外,均受到著作權保護,並且保留所有的權利。
