帳號:guest(18.116.80.34)          離開系統
字體大小: 字級放大   字級縮小   預設字形  

詳目顯示

以作者查詢圖書館館藏以作者查詢臺灣博碩士論文系統以作者查詢全國書目
作者(中文):廖敏伊
作者(外文):Liao, Min-I
論文名稱(中文):以微流道晶片影像辨識系統藉液態活檢之循環腫瘤細胞與其團塊評估肺癌病人於免疫治療後之預後
論文名稱(外文):Using the microfluidic chip image recognition system to evaluate the prognosis of lung cancer patients after treatment by liquid biopsy of circulating tumor cells and their masses
指導教授(中文):饒達仁
指導教授(外文):Yao, Da-Jeng
口試委員(中文):曾繁根
李岡遠
江啓勲
口試委員(外文):Tseng, Fan-Gang
Lee, Kang-Yun
Chiang, Chi-Shiun
學位類別:碩士
校院名稱:國立清華大學
系所名稱:動力機械工程學系
學號:109033520
出版年(民國):111
畢業學年度:110
語文別:中文
論文頁數:73
中文關鍵詞:肺癌免疫治療非小細胞肺癌循環腫瘤細胞循環腫瘤細胞簇
外文關鍵詞:Lung cancerImmunotherapycirculating tumor cellsCirculating Tunor Microemboli
相關次數:
  • 推薦推薦:0
  • 點閱點閱:294
  • 評分評分:*****
  • 下載下載:0
  • 收藏收藏:0
肺癌在近年已經成為癌症死亡率中的首位,其可分為非小細胞肺癌及小細胞肺癌,包括此兩者在內,轉移性肺癌在五年內的存活率極低,在早期的治療手段中,主要以放射治療、化學治療以及標靶治療為主,但是這幾種治療效果仍然有限,故在近年免疫治療成為治療肺癌的一種新手段。
免疫治療主要是透過自體免疫的方式來治療癌症,其中又分為許多種激活自體免疫的方式,常見的兩大類為CTLA-4抑制劑以及PD-1抑制劑,這兩種方式所帶來的治療效果為肺癌的治療注入一股新流,在臨床上更是取得了前所未有的成功。
在另一方面,循環腫瘤細胞也一直是癌症領域研究的重點,他與臨床上的關係引發人們的興趣。但是在腫瘤細胞的富集以及分析上,卻面臨許多困難。為了更好的進行研究,實驗室研發出細胞自組裝排列晶片以及自動化拍攝系統被大量地使用在了這一項研究之中。在先前的許多研究中,已經提出了腫瘤細胞的數量與不良的預後具有正相關,但應用在免疫治療上的研究卻少之又少,故本研究期望建立起腫瘤細胞的數量以及免疫治療的效果的關聯性。
在方法的建立上主要涵蓋兩種預測因子,其一為循環腫瘤細胞(Circulating Tunor Cells, CTC),其二為循環腫瘤細胞簇(Circulating Tunor Microemboli, CTM),透過ROC Curve的分析方法,我們找到CTC以及CTM的最適切點分別為35以及7.5,預測準確率分別為0.887以及0.885,為了使預測的結果可以涵蓋CTC以及CTM的預測效力,我們建立一指標,即C Score指標,不同於過去僅用CTC作為預測因子的分析方法,合併CTM進行分析後,使得預測的準確率更為提升,透過ROC Curve計算後,準確率達到0.975高於使用單一指標進行預測,最適切點為-0.566,為了使分析結果更為直觀,故我們將此進行偏移,使判斷基準點為0,也就是說,當C Score大於0則病情惡化機會高,反之則為病情穩定,C Score可以直接的反應出患者體內腫瘤的變化,相較於許多目前臨床上的指標,容易受到其他因素所影響,此指標具有更好的判斷性以及穩定性。
透過此項指標,我們將患者之用藥方式分為三組,第一組為免疫治療合併化療,這組的患者目前病況都為穩定,第二組為於第一組的基礎上加上血管抑制劑,此組患者病情均惡化,造成此項差異的原因可能是因為血管抑制劑切斷了腫瘤的新生血管使得藥物進入腫瘤更為困難,進而導致效果不如直接用藥來的好。第三組為新藥TIGIT(T Cell Ig and ITIM Domain)的藥物測試,此為一新型免疫抑制劑,它可能影響CD8+ T細胞的功能,近兩年作為一新型藥物被廣泛研究。結果發現第一組的效果最好,其次為第三組新要測試組合,最後為第二組合併血管抑制劑的用藥方式。
Lung cancer has become the first cancer mortality rate in recent years. It can be divided into non-small cell lung cancer and small cell lung cancer. Including these two, the survival rate of metastatic lung cancer within five years is extremely low. Among them, radiotherapy, chemotherapy, and targeted therapy are the mainstays, but the effects of these treatments are still limited. Therefore, in recent years, immunotherapy has become a new method for the treatment of lung cancer.
Immunotherapy is mainly to treat cancer through autoimmunity, which is divided into many ways to activate autoimmunity. The two common types are CTLA-4 inhibitors and PD-1 inhibitors, which are carried by these two methods. The new therapeutic effect has injected a new wave into the treatment of lung cancer, and it has achieved unprecedented success in clinical practice.
On the other hand, circulating tumor cells have also been the focus of research in the field of cancer, and their clinical relevance has sparked interest. However, there are many difficulties in the enrichment and analysis of tumor cells. In order to better conduct research, the laboratory developed a cell self-assembly array wafer and an automated imaging system that was extensively used in this research. In many previous studies, it has been proposed that the number of tumor cells has a positive correlation with poor prognosis, but there are very few studies on the application of immunotherapy, so this study is expected to establish the number of tumor cells and the effect of immunotherapy. correlation of effects.
The establishment of the method mainly covers two predictors, one is Circulating Tunor Cells, and the other one is Circulating Tunor Microemboli. Through the ROC Curve analysis method, we found the optimal cut-points of CTC and CTM are 35 and 7.5, respectively, and the prediction accuracy is 0.887 and 0.885, respectively. In order to make the prediction results cover the prediction effectiveness of CTC and CTM, we establish an indicator, the C Score indicator, which is different from the past analysis method only using CTC as a predictor and combining CTM for analysis can improve the accuracy of the prediction. After calculating through the ROC Curve, the accuracy rate reaches 0.975, which is higher than that of using a single indicator for prediction. The optimal cut point is -0.566. In order to make the analysis results more intuitive, we offset this to make the judgment reference point 0. That is to say, when the C Score is greater than 0, the probability of disease deterioration is high; otherwise, the disease is stable, and the C Score can directly reflect the disease. Compared with many current clinical indicators, tumor changes in patients are easily affected by other factors, and this indicator has better judgment and stability.
Through this indicator, we divided the patients into three groups. The first group was immunotherapy combined with chemotherapy. The patients in this group were all stable at present. The second group was based on the first group plus vascular inhibition. The condition of the patients in this group deteriorated. The reason for this difference may be that the vascular inhibitor cuts off the new blood vessels of the tumor, making it more difficult for the drug to enter the tumor, and thus the effect is not as good as the direct drug. The third group is the drug test of the new drug TIGIT (T Cell Ig and ITIM Domain), which is a new type of immunosuppressant, which may affect the function of CD8+ T cells. It has been widely studied as a new type of drug in the past two years. The results showed that the first group had the best effect, followed by the third group of new combinations to be tested, and finally the second group combined with vascular inhibitors.
致 謝..........................I
摘 要.........................II
ABSTRACT......................IV
章節目錄.......................VI
表目錄........................IX
圖目錄.......................X
第一章 緒論....................1
1.1 研究背景.....................1
1.2 研究動機.....................9
1.3 研究目的....................11
1.4 研究架構....................14
第二章 文獻探討................16
2.1 循環腫瘤細胞(CIRCULATING TUMOR CELLS, CTC)基本介紹.........16
2.2 CTC富集方法生物學基礎......................................17
2.2.1 腫瘤細胞大小...............................................17
2.2.2 腫瘤細胞變形能力...........................................19
2.2.3 腫瘤細胞電學特性...........................................20
2.3 CTC富集方法的分類和原理.....................................20
2.3.1 基於物理特性的CTC富集.......................................22
2.3.2 基於生物學特性的CTC富集.....................................29
2.4 CTC檢測...................................................31
第三章 實驗設置與研究架構..........................................33
3.1 實驗設備...................................................33
3.2 臨床血液樣本來源............................................35
3.3 血液採集與準備..............................................35
3.3.1 血液的收集方式與CTC分離方法..................................36
3.4 血液染色處理................................................37
3.4.1 免疫螢光染劑選擇............................................38
3.5 細胞影像辨識................................................38
3.6 統計分析方法................................................41
3.6.1 ROC Curve..................................................41
3.6.2 計算ROC Curve之面積.........................................42
3.6.3 Kaplan-Meierc Curve........................................43
第四章 實驗結果與討論..............................................44
4.1 患者之收案情況..............................................44
4.2 CTC/CTM數量之變化與臨床之相關性..............................45
4.3 臨床預測模型之建立...........................................47
4.4 臨床預測模型之預測效力.......................................53
4.5 利用C SCORE進行不同藥物治療之臨床分析.........................54
4.6 綜合討論....................................................63
第五章 結論與未來展望...............................................66
5.1 結論........................................................66
5.2 未來展望....................................................67
參考文獻............................................................68
[1] M. Akhtar et al., “Paget’s “seed and soil” theory of cancer metastasis: an idea whose time has come,” Advances in anatomic pathology, vol. 26, no. 1, pp. 69-74, 2019.
[2] I. Fidler, “Selection of successive tumour lines for metastasis,” Nature New Biology, vol. 242, no. 118, pp. 148-149, 1973.
[3] I. J. Fidler, “Metastasis: quantitative analysis of distribution and fate of tumor emboli labeled with 125I-5-iodo-2′-deoxyuridine,” Journal of the National Cancer Institute, vol. 45, no. 4, pp. 773-782, 1970.
[4] I. J. Fidler, “Modulation of the organ microenvironment for treatment of cancer metastasis,” JNCI: Journal of the National Cancer Institute, vol. 87, no. 21, pp. 1588-1592, 1995.
[5] I. J. Fidler, and I. R. Hart, “Biological diversity in metastatic neoplasms: origins and implications,” Science, vol. 217, no. 4564, pp. 998-1003, 1982.
[6] V. Akpe et al., “Circulating tumour cells: a broad perspective,” Journal of the Royal Society Interface, vol. 17, no. 168, pp. 20200065, 2020.
[7] K. C. Andree, G. van Dalum, and L. W. Terstappen, “Challenges in circulating tumor cell detection by the CellSearch system,” Molecular oncology, vol. 10, no. 3, pp. 395-407, 2016.
[8] D. S. Micalizzi, S. Maheswaran, and D. A. Haber, “A conduit to metastasis: circulating tumor cell biology,” Genes & development, vol. 31, no. 18, pp. 1827-1840, 2017.
[9] P. Paterlini-Brechot, and N. L. Benali, “Circulating tumor cells (CTC) detection: clinical impact and future directions,” Cancer letters, vol. 253, no. 2, pp. 180-204, 2007.
[10] K. Pantel et al., “Circulating epithelial cells in patients with benign colon diseases,” Clinical chemistry, vol. 58, no. 5, pp. 936-940, 2012.
[11] J. Inhestern et al., “Prognostic role of circulating tumor cells during induction chemotherapy followed by curative surgery combined with postoperative radiotherapy in patients with locally advanced oral and oropharyngeal squamous cell cancer,” PloS one, vol. 10, no. 7, pp. e0132901, 2015.
[12] O. A. Martin et al., “Does the mobilization of circulating tumour cells during cancer therapy cause metastasis?,” Nature reviews Clinical oncology, vol. 14, no. 1, pp. 32-44, 2017.
[13] M. Mego et al., “Prognostic value of EMT-circulating tumor cells in metastatic breast cancer patients undergoing high-dose chemotherapy with autologous hematopoietic stem cell transplantation,” Journal of Cancer, vol. 3, pp. 369, 2012.
[14] H. S. Kaplan, “THE EFFECT OF LOCAL ROENTGEN IRRADIATION ON THE BIOLOGICAL BEHAVIOR,” Journal, vol. 9, pp. 407, 1948.
[15] P. Sheldon, and J. Fowler, “The effect of low-dose pre-operative X-irradiation of implanted mouse mammary carcinomas on local recurrence and metastasis,” British journal of cancer, vol. 34, no. 4, pp. 401-407, 1976.
[16] K. Camphausen et al., “Radiation therapy to a primary tumor accelerates metastatic growth in mice,” Cancer research, vol. 61, no. 5, pp. 2207-2211, 2001.
[17] R. Bütof, A. Dubrovska, and M. Baumann, “Clinical perspectives of cancer stem cell research in radiation oncology,” Radiotherapy and Oncology, vol. 108, no. 3, pp. 388-396, 2013.
[18] E. K. Rofstad, K. Galappathi, and B. S. Mathiesen, “Tumor interstitial fluid pressure—a link between tumor hypoxia, microvascular density, and lymph node metastasis,” Neoplasia, vol. 16, no. 7, pp. 586-594, 2014.
[19] O. A. Martin et al., “Mobilization of viable tumor cells into the circulation during radiation therapy,” International Journal of Radiation Oncology* Biology* Physics, vol. 88, no. 2, pp. 395-403, 2014.
[20] J. Kusukawa et al., “Dissemination of cancer cells into circulation occurs by incisional biopsy of oral squamous cell carcinoma,” Journal of oral pathology & medicine, vol. 29, no. 7, pp. 303-307, 2000.
[21] T. J. Polascik et al., “Influence of sextant prostate needle biopsy or surgery on the detection and harvest of intact circulating prostate cancer cells,” The Journal of urology, vol. 162, no. 3 Part 1, pp. 749-752, 1999.
[22] P. Sharma et al., “Novel cancer immunotherapy agents with survival benefit: recent successes and next steps,” Nature Reviews Cancer, vol. 11, no. 11, pp. 805-812, 2011.
[23] H. Sadeghi Rad et al., “Understanding the tumor microenvironment for effective immunotherapy,” Medicinal Research Reviews, vol. 41, no. 3, pp. 1474-1498, 2021.
[24] G. Q. Phan et al., “Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma,” Proceedings of the National Academy of Sciences, vol. 100, no. 14, pp. 8372-8377, 2003.
[25] A. Ribas et al., “Antitumor activity in melanoma and anti-self responses in a phase I trial with the anti-cytotoxic T lymphocyte-associated antigen 4 monoclonal antibody CP-675,206,” J Clin Oncol, vol. 23, no. 896877.25, 2005.
[26] J. R. Brahmer, and D. M. Pardoll, “Immune checkpoint inhibitors: making immunotherapy a reality for the treatment of lung cancer,” Cancer immunology research, vol. 1, no. 2, pp. 85-91, 2013.
[27] C.-Y. Mu et al., “High expression of PD-L1 in lung cancer may contribute to poor prognosis and tumor cells immune escape through suppressing tumor infiltrating dendritic cells maturation,” Medical oncology, vol. 28, no. 3, pp. 682-688, 2011.
[28] W. J. Lesterhuis, H. Steer, and R. A. Lake, “PD-L2 is predominantly expressed by Th2 cells,” Molecular immunology, vol. 49, no. 1-2, pp. 1-3, 2011.
[29] E. Soularue et al., “Enterocolitis due to immune checkpoint inhibitors: a systematic review,” Gut, vol. 67, no. 11, pp. 2056-2067, 2018.
[30] K.-A. Hyun et al., “Isolation and enrichment of circulating biomarkers for cancer screening, detection, and diagnostics,” Analyst, vol. 141, no. 2, pp. 382-392, 2016.
[31] L. A. Torre et al., “Global cancer statistics, 2012,” CA: a cancer journal for clinicians, vol. 65, no. 2, pp. 87-108, 2015.
[32] P. Goldstraw et al., “The IASLC Lung Cancer Staging Project: proposals for the revision of the TNM stage groupings in the forthcoming (seventh) edition of the TNM Classification of malignant tumours,” Journal of thoracic oncology, vol. 2, no. 8, pp. 706-714, 2007.
[33] C. K. Lee et al., “Impact of EGFR inhibitor in non–small cell lung cancer on progression-free and overall survival: a meta-analysis,” Journal of the National Cancer Institute, vol. 105, no. 9, pp. 595-605, 2013.
[34] A. T. Shaw et al., “Ceritinib in ALK-rearranged non–small-cell lung cancer,” N Engl j med, vol. 370, pp. 1189-1197, 2014.
[35] B. J. Solomon et al., “First-line crizotinib versus chemotherapy in ALK-positive lung cancer,” N Engl j Med, vol. 371, pp. 2167-2177, 2014.
[36] P. C. Bailey, and S. S. Martin, “Insights on CTC biology and clinical impact emerging from advances in capture technology,” Cells, vol. 8, no. 6, pp. 553, 2019.
[37] T. Ashworth, “A case of cancer in which cells similar to those in the tumours were seen in the blood after death,” Aust Med J., vol. 14, pp. 146, 1869.
[38] S. Braun et al., “A pooled analysis of bone marrow micrometastasis in breast cancer,” New England journal of medicine, vol. 353, no. 8, pp. 793-802, 2005.
[39] B. Rack et al., “CTCs in primary breast cancer (I),” Minimal Residual Disease and Circulating Tumor Cells in Breast Cancer, pp. 179-185, 2012.
[40] V. Mikulová et al., “Detection of circulating tumor cells during follow-up of patients with early breast cancer: Clinical utility for monitoring of therapy efficacy,” Scandinavian journal of clinical and laboratory investigation, vol. 74, no. 2, pp. 132-142, 2014.
[41] A. Lucci et al., “Circulating tumour cells in non-metastatic breast cancer: a prospective study,” The lancet oncology, vol. 13, no. 7, pp. 688-695, 2012.
[42] V. Gupta et al., “ApoStream™, a new dielectrophoretic device for antibody independent isolation and recovery of viable cancer cells from blood,” Biomicrofluidics, vol. 6, no. 2, pp. 024133, 2012.
[43] M. M. Wintrobe, Wintrobe's clinical hematology: Lippincott Williams & Wilkins, 2008.
[44] G. Schmid-Schonbein, Y. Y. Shih, and S. Chien, “Morphometry of human leukocytes,” 1980.
[45] R. A. Harouaka, M. Nisic, and S.-Y. Zheng, “Circulating tumor cell enrichment based on physical properties,” Journal of laboratory automation, vol. 18, no. 6, pp. 455-468, 2013.
[46] R. A. Weinberg, The biology of cancer: Garland science, 2013.
[47] F. L. Meyskens, S. P. Thomson, and T. E. Moon, “Quantitation of the number of cells within tumor colonies in semisolid medium and their growth as oblate spheroids,” Cancer research, vol. 44, no. 1, pp. 271-277, 1984.
[48] J. J. Nieva, and P. Kuhn, “Fluid biopsy for solid tumors: a patient’s companion for lifelong characterization of their disease,” Future oncology, vol. 8, no. 8, pp. 989-998, 2012.
[49] J. Nieva et al., “High-definition imaging of circulating tumor cells and associated cellular events in non-small cell lung cancer patients: a longitudinal analysis,” Physical biology, vol. 9, no. 1, pp. 016004, 2012.
[50] H. Mohamed et al., “Isolation of tumor cells using size and deformation,” Journal of Chromatography A, vol. 1216, no. 47, pp. 8289-8295, 2009.
[51] Q. Li et al., “AFM indentation study of breast cancer cells,” Biochemical and biophysical research communications, vol. 374, no. 4, pp. 609-613, 2008.
[52] D. R. Gossett et al., “Hydrodynamic stretching of single cells for large population mechanical phenotyping,” Proceedings of the National Academy of Sciences, vol. 109, no. 20, pp. 7630-7635, 2012.
[53] A. Han, L. Yang, and A. B. Frazier, “Quantification of the heterogeneity in breast cancer cell lines using whole-cell impedance spectroscopy,” Clinical cancer research, vol. 13, no. 1, pp. 139-143, 2007.
[54] G. Galletti et al., “Using circulating tumor cells to advance precision medicine in prostate cancer,” Journal of cancer metastasis and treatment, vol. 3, pp. 190, 2017.
[55] S. Seal, “Silicone flotation: A simple quantitative method for the isolation of free‐floating cancer cells from the blood,” Cancer, vol. 12, no. 3, pp. 590-595, 1959.
[56] R. Rosenberg et al., “Comparison of two density gradient centrifugation systems for the enrichment of disseminated tumor cells in blood,” Cytometry: The Journal of the International Society for Analytical Cytology, vol. 49, no. 4, pp. 150-158, 2002.
[57] S. Seal, “A sieve for the isolation of cancer cells and other large cells from the blood,” Cancer, vol. 17, no. 5, pp. 637-642, 1964.
[58] G. Vona et al., “Isolation by size of epithelial tumor cells: a new method for the immunomorphological and molecular characterization of circulating tumor cells,” The American journal of pathology, vol. 156, no. 1, pp. 57-63, 2000.
[59] S. Zheng et al., “3D microfilter device for viable circulating tumor cell (CTC) enrichment from blood,” Biomedical microdevices, vol. 13, no. 1, pp. 203-213, 2011.
[60] S. J. Tan et al., “Microdevice for the isolation and enumeration of cancer cells from blood,” Biomedical microdevices, vol. 11, no. 4, pp. 883-892, 2009.
[61] A. A. S. Bhagat et al., “Pinched flow coupled shear-modulated inertial microfluidics for high-throughput rare blood cell separation,” Lab on a Chip, vol. 11, no. 11, pp. 1870-1878, 2011.
[62] S. Hur, A. Mach, and D. Di Carlo, “High-Throughput Size-Based Rare Cell Enrichment Using Microscale Vortices. Biomicrofluidics, 5,” Article ID, vol. 22206, 2011.
[63] H. Hou et al., "Isolation and retrieval of circulating tumor cells using centrifugal forces. Sci. Rep. 3, 1259," 2013.
[64] F. Becker et al., “The removal of human leukaemia cells from blood using interdigitated microelectrodes,” Journal of Physics D: Applied Physics, vol. 27, no. 12, pp. 2659, 1994.
[65] G. Moldenhauer et al., “Epithelium-specific surface glycoprotein of M r 34,000 is a widely distributed human carcinoma marker,” British journal of cancer, vol. 56, no. 6, pp. 714-721, 1987.
[66] G. Deng et al., “Enrichment with anti-cytokeratin alone or combined with anti-EpCAM antibodies significantly increases the sensitivity for circulating tumor cell detection in metastatic breast cancer patients,” Breast Cancer Research, vol. 10, no. 4, pp. 1-11, 2008.
[67] N. Pamme, “On-chip bioanalysis with magnetic particles,” Current opinion in chemical biology, vol. 16, no. 3-4, pp. 436-443, 2012.
[68] A. H. Talasaz et al., “Isolating highly enriched populations of circulating epithelial cells and other rare cells from blood using a magnetic sweeper device,” Proceedings of the National Academy of Sciences, vol. 106, no. 10, pp. 3970-3975, 2009.
[69] A. A. Ghazani et al., “Comparison of select cancer biomarkers in human circulating and bulk tumor cells using magnetic nanoparticles and a miniaturized micro-NMR system,” Nanomedicine: Nanotechnology, Biology and Medicine, vol. 9, no. 7, pp. 1009-1017, 2013.
[70] P. D. Potdar, and N. K. Lotey, “Role of circulating tumor cells in future diagnosis and therapy of cancer,” Journal of Cancer Metastasis and Treatment, vol. 1, pp. 44-56, 2015.
[71] L. H. Broersen et al., “Clinical application of circulating tumor cells in breast cancer,” Cellular oncology, vol. 37, no. 1, pp. 9-15, 2014.
[72] S. Riethdorf et al., “Detection of circulating tumor cells in peripheral blood of patients with metastatic breast cancer: a validation study of the CellSearch system,” Clinical cancer research, vol. 13, no. 3, pp. 920-928, 2007.
[73] A. Toss et al., “CTC enumeration and characterization: moving toward personalized medicine,” Annals of translational medicine, vol. 2, no. 11, 2014.

 
 
 
 
第一頁 上一頁 下一頁 最後一頁 top
* *