參考文獻 |
[1]Ramalho-Santos, M. and H. Willenbring (2007). "On the origin of the term “stem cell”." Cell stem cell 1(1): 35-38.
[2]Chagastelles, P. C. and N. B. Nardi (2011). "Biology of stem cells: an overview." Kidney international supplements 1(3): 63-67.
[3]Cai, J., et al. (2022). "Research Progress of Totipotent Stem Cells." Stem Cells and Development 31(13-14): 335-345.
[4]Malik, V. and J. Wang (2022). "Pursuing totipotency: authentic totipotent stem cells in culture." Trends in Genetics.
[5]Shen, H., et al. (2021). "Mouse totipotent stem cells captured and maintained through spliceosomal repression." Cell 184(11): 2843-2859. e2820.
[6]Hu, Y., et al. (2023). "Induction of mouse totipotent stem cells by a defined chemical cocktail." Nature 617(7962): 792-797.
[7]Xu, Y., et al. (2022). "Derivation of totipotent-like stem cells with blastocyst-like structure forming potential." Cell Research 32(6): 513-529.
[8]Smith, A. (2010). "Pluripotent stem cells: private obsession and public expectation." EMBO Molecular Medicine 2(4): 113-116.
[9]de Figueiredo Pessôa, L. V., et al. (2019). "Induced pluripotent stem cells throughout the animal kingdom: availability and applications." World Journal of Stem Cells 11(8): 491.
[10]Evans, M. J. and M. H. Kaufman (1981). "Establishment in culture of pluripotential cells from mouse embryos." Nature 292(5819): 154-156.
[11]Thomson, J. A., et al. (1998). "Embryonic stem cell lines derived from human blastocysts." science 282(5391): 1145-1147.
[12]Nichols, J., et al. (1998). "Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4." Cell 95(3): 379-391.
[13]Takahashi, K., et al. (2007). "Induction of pluripotent stem cells from adult human fibroblasts by defined factors." Cell 131(5): 861-872.
[14]Takahashi, K. and S. Yamanaka (2006). "Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors." Cell 126(4): 663-676.
[15]Yu, J., et al. (2007). "Induced pluripotent stem cell lines derived from human somatic cells." science 318(5858): 1917-1920.
[16]Nakagawa, M., et al. (2008). "Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts." Nature biotechnology 26(1): 101-106.
[17]Konstantinov, I. E. (2000). "In search of Alexander A. Maximow: the man behind the unitarian theory of hematopoiesis." Perspectives in biology and medicine 43(2): 269-276.
[18]Till, J. E., et al. (1964). "A stochastic model of stem cell proliferation, based on the growth of spleen colony-forming cells." Proceedings of the National Academy of Sciences 51(1): 29-36.
[19]Lilja, A. M., et al. (2018). "Clonal analysis of Notch1-expressing cells reveals the existence of unipotent stem cells that retain long-term plasticity in the embryonic mammary gland." Nature cell biology 20(6): 677-687.
[20]Estrada-Meza, C., et al. (2022). "Recent insights into the microRNA and long non-coding RNA-mediated regulation of stem cell populations." 3 Biotech 12(10): 270.
[21]Zhou, J. N., et al. (2022). "A Functional Screening Identifies a New Organic Selenium Compound Targeting Cancer Stem Cells: Role of c‐Myc Transcription Activity Inhibition in Liver Cancer." Advanced Science 9(22): 2201166.
[22]Koury, J., et al. (2017). "Targeting signaling pathways in cancer stem cells for cancer treatment." Stem cells international 2017.
[23]Cho, D.-Y., et al. (2013). "Targeting cancer stem cells for treatment of glioblastoma multiforme." Cell transplantation 22(4): 731-739.
[24]Zhou, H.-M., et al. (2021). "Targeting cancer stem cells for reversing therapy resistance: Mechanism, signaling, and prospective agents." Signal transduction and targeted therapy 6(1): 62.
[25]Abubaker, K., et al. (2013). "Short-term single treatment of chemotherapy results in the enrichment of ovarian cancer stem cell-like cells leading to an increased tumor burden." Molecular cancer 12: 1-15.
[26]López de Andrés, J., et al. (2020). "Cancer stem cell secretome in the tumor microenvironment: a key point for an effective personalized cancer treatment." Journal of hematology & oncology 13(1): 136.
[27]Zhou, Y., et al. (2023). "The role of mesenchymal stem cells derived exosomes as a novel nanobiotechnology target in the diagnosis and treatment of cancer." Frontiers in Bioengineering and Biotechnology 11.
[28]Sarkar, M., et al. (2023). "Cancer-associated fibroblasts: The chief architect in the tumor microenvironment." Frontiers in Cell and Developmental Biology 11: 1089068.
[29]Wright, K., et al. (2023). "Cancer-Associated Fibroblasts: Master Tumor Microenvironment Modifiers." Cancers 15(6): 1899.
[30]Luo, H., et al. (2022). "Pan-cancer single-cell analysis reveals the heterogeneity and plasticity of cancer-associated fibroblasts in the tumor microenvironment." Nature Communications 13(1): 6619.
[31]Peng, Z., et al. (2022). "Spatial transcriptomics atlas reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment components in colorectal cancer." Journal of Translational Medicine 20(1): 302.
[32]Mao, X., et al. (2021). "Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives." Molecular cancer 20(1): 1-30.
[33]Brassart-Pasco, S., et al. (2020). "Tumor microenvironment: extracellular matrix alterations influence tumor progression." Frontiers in oncology 10: 397.
[34]Henke, E., et al. (2020). "Extracellular matrix in the tumor microenvironment and its impact on cancer therapy." Frontiers in molecular biosciences 6: 160.
[35]Popova, N. V. and M. Jücker (2022). "The functional role of extracellular matrix proteins in cancer." Cancers 14(1): 238.
[36]Winkler, J., et al. (2020). "Concepts of extracellular matrix remodelling in tumour progression and metastasis." Nature Communications 11(1): 5120.
[37]Yuan, Z., et al. (2023). "Extracellular matrix remodeling in tumor progression and immune escape: from mechanisms to treatments." Molecular cancer 22(1): 48.
[38]Yang, Q., et al. (2020). "The role of tumor-associated macrophages (TAMs) in tumor progression and relevant advance in targeted therapy." Acta Pharmaceutica Sinica B 10(11): 2156-2170.
[39]Chen, Y., et al. (2019). "Tumor-associated macrophages: an accomplice in solid tumor progression." Journal of biomedical science 26(1): 1-13.
[40]Yang, L., et al. (2020). "Targeting cancer stem cell pathways for cancer therapy." Signal transduction and targeted therapy 5(1): 8.
[41]Li, Y.-R., et al. (2022). "An ex vivo 3D tumor microenvironment-mimicry culture to study TAM modulation of cancer immunotherapy." Cells 11(9): 1583.
[42]Wang, J., et al. (2019). "Crosstalk between cancer and immune cells: Role of tumor‐associated macrophages in the tumor microenvironment." Cancer medicine 8(10): 4709-4721.
[43]Rahma, O. E. and F. S. Hodi (2019). "The intersection between tumor angiogenesis and immune suppression." Clinical Cancer Research 25(18): 5449-5457.
[44]Fong, C. Y., et al. (2009). "Separation of SSEA-4 and TRA-1–60 labelled undifferentiated human embryonic stem cells from a heterogeneous cell population using magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting (FACS)." Stem Cell Reviews and Reports 5: 72-80.
[45]Manoli, M. and W. Driever (2012). "Fluorescence-activated cell sorting (FACS) of fluorescently tagged cells from zebrafish larvae for RNA isolation." Cold Spring Harbor Protocols 2012(8): pdb. prot069633.
[46]Sharon, Y., et al. (2013). "Isolation of normal and cancer-associated fibroblasts from fresh tissues by Fluorescence Activated Cell Sorting (FACS)." JoVE (Journal of Visualized Experiments)(71): e4425.
[47]Baker, M. K., et al. (2003). "Molecular detection of breast cancer cells in the peripheral blood of advanced-stage breast cancer patients using multimarker real-time reverse transcription-polymerase chain reaction and a novel porous barrier density gradient centrifugation technology." Clinical Cancer Research 9(13): 4865-4871.
[48]Liu, W.-h., et al. (2012). "Efficient enrichment of hepatic cancer stem-like cells from a primary rat HCC model via a density gradient centrifugation-centered method." PloS one 7(4): e35720.
[49]Grunt, T. W., et al. (1991). "Separation of clonogenic and differentiated cell phenotypes of ovarian cancer cells (HOC-7) by discontinuous density gradient centrifugation." Cancer letters 58(1-2): 7-16.
[50]Baday, M., et al. (2019). "Density based characterization of mechanical cues on cancer cells using magnetic levitation." Advanced healthcare materials 8(10): 1801517.
[51]Lin, Z., et al. (2020). "Rapid assessment of surface markers on cancer cells using immuno-magnetic separation and multi-frequency impedance cytometry for targeted therapy." Scientific reports 10(1): 3015.
[52]Bankó, P., et al. (2019). "Technologies for circulating tumor cell separation from whole blood." Journal of hematology & oncology 12: 1-20.
[53]Seal, S. (1964). "A sieve for the isolation of cancer cells and other large cells from the blood." Cancer 17(5): 637-642.
[54]Higuchi, A., et al. (2006). "Separation of CD34+ cells from human peripheral blood through polyurethane foaming membranes." Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials 78(3): 491-499.
[55]Higuchi, A., et al. (2008). "Separation of hematopoietic stem cells from human peripheral blood through modified polyurethane foaming membranes." Journal of Biomedical Materials Research Part A: An Official Journal of The Society for Biomaterials, The Japanese Society for Biomaterials, and The Australian Society for Biomaterials and the Korean Society for Biomaterials 85(4): 853-861.
[56]Chen, D.-C., et al. (2014). "Purification of human adipose-derived stem cells from fat tissues using PLGA/silk screen hybrid membranes." Biomaterials 35(14): 4278-4287.
[57]Wu, C.-H., et al. (2012). "The isolation and differentiation of human adipose-derived stem cells using membrane filtration." Biomaterials 33(33): 8228-8239.
[58]Sung, T.-C., et al. (2020). "Enrichment of cancer-initiating cells from colon cancer cells through porous polymeric membranes by a membrane filtration method." Journal of Materials Chemistry B 8(46): 10577-10585.
[59]Kim, W.-T. and C. J. Ryu (2017). "Cancer stem cell surface markers on normal stem cells." BMB reports 50(6): 285.
[60]Xia, P. (2014). "Surface markers of cancer stem cells in solid tumors." Current stem cell research & therapy 9(2): 102-111.
[61]Walcher, L., et al. (2020). "Cancer stem cells—origins and biomarkers: perspectives for targeted personalized therapies." Frontiers in immunology 11: 539291.
[62]Woodward, W. A. and E. P. Sulman (2008). "Cancer stem cells: markers or biomarkers?" Cancer and metastasis reviews 27: 459-470.
[63]Leon, G., et al. (2016). "Cancer stem cells in drug resistant lung cancer: Targeting cell surface markers and signaling pathways." Pharmacology & therapeutics 158: 71-90.
[64]Tang, K. H., et al. (2012). "CD133+ liver tumor‐initiating cells promote tumor angiogenesis, growth, and self‐renewal through neurotensin/interleukin‐8/CXCL1 signaling." Hepatology 55(3): 807-820.
[65]Yang, Z. F., et al. (2008). "Significance of CD90+ cancer stem cells in human liver cancer." Cancer cell 13(2): 153-166.
[66]Yamashita, T., et al. (2009). "EpCAM-positive hepatocellular carcinoma cells are tumor-initiating cells with stem/progenitor cell features." Gastroenterology 136(3): 1012-1024. e1014.
[67]Hermann, P. C., et al. (2007). "Distinct populations of cancer stem cells determine tumor growth and metastatic activity in human pancreatic cancer." Cell stem cell 1(3): 313-323.
[68]Skoda, J., et al. (2016). "Co-expression of cancer stem cell markers corresponds to a pro-tumorigenic expression profile in pancreatic adenocarcinoma." PloS one 11(7): e0159255.
[69]Durko, L., et al. (2017). "Expression and clinical significance of cancer stem cell markers CD24, CD44, and CD133 in pancreatic ductal adenocarcinoma and chronic pancreatitis." Disease markers 2017.
[70]Wang, L., et al. (2023). "Potential markers of cancer stem-like cells in ESCC: a review of the current knowledge." Frontiers in oncology 13.
[71]Wu, Q., et al. (2019). "Cancer stem cells in esophageal squamous cell cancer." Oncology letters 18(5): 5022-5032.
[72]Zhang, X., et al. (2016). "Identification of stem-like cells and clinical significance of candidate stem cell markers in gastric cancer." Oncotarget 7(9): 9815.
[73]Nishikawa, S., et al. (2015). "Surgically resected human tumors reveal the biological significance of the gastric cancer stem cell markers CD44 and CD26." Oncology letters 9(5): 2361-2367.
[74]Huang, J. L., et al. (2021). "Clinical relevance of stem cell surface markers CD133, CD24, and CD44 in colorectal cancer." American Journal of Cancer Research 11(10): 5141.
[75]Andrews, T. E., et al. (2013). "Cell surface markers of cancer stem cells: diagnostic macromolecules and targets for drug delivery." Drug delivery and translational research 3: 121-142.
[76]Calaf, G. M., et al. (2018). "Effect of curcumin on the cell surface markers CD44 and CD24 in breast cancer." Oncology Reports 39(6): 2741-2748.
[77]Barsky, S. H., et al. (2009). "CD133, a stem cell surface marker in inflammatory breast cancer, functions in tumor cell survival." The FASEB Journal 23: 363.367-363.367.
[78]Shigdar, S., et al. (2011). "RNA aptamer against a cancer stem cell marker epithelial cell adhesion molecule." Cancer science 102(5): 991-998.
[79]Klemba, A., et al. (2018). "Surface markers of cancer stem-like cells of ovarian cancer and their clinical relevance." Contemporary Oncology/Współczesna Onkologia 2018(1): 48-55.
[80]Muñoz-Galván, S. and A. Carnero (2020). "Targeting cancer stem cells to overcome therapy resistance in ovarian cancer." Cells 9(6): 1402.
[81]Singh, S. K., et al. (2003). "Identification of a cancer stem cell in human brain tumors." Cancer research 63(18): 5821-5828.
[82]Tang, D. G. (2022). Understanding and targeting prostate cancer cell heterogeneity and plasticity. Seminars in cancer biology, Elsevier.
[83]Culen, M., et al. (2019). "Multicolor Immunophenotyping of Candidate Leukemic Stem Cell Markers in CD34+ CD38-Chronic Myeloid Leukemia Stem Cells." Blood 134: 2922.
[84]Goeminne, J.-C., et al. (1999). "Unreliability of carcinoembryonic antigen (CEA) reverse transcriptase-polymerase chain reaction (RT-PCR) in detecting contaminating breast cancer cells in peripheral blood stem cells due to induction of CEA by growth factors." Bone marrow transplantation 24(7): 769-775.
[85]Shah, K. and P. Maghsoudlou (2016). "Enzyme-linked immunosorbent assay (ELISA): the basics." British journal of hospital medicine 77(7): C98-C101.
[86]Lázaro-Gorines, R., et al. (2019). "A novel carcinoembryonic antigen (CEA)-targeted trimeric immunotoxin shows significantly enhanced antitumor activity in human colorectal cancer xenografts." Scientific reports 9(1): 11680.
[87]Sandberg, M. L., et al. (2022). "A carcinoembryonic antigen-specific cell therapy selectively targets tumor cells with HLA loss of heterozygosity in vitro and in vivo." Science Translational Medicine 14(634): eabm0306.
[88]Das, A., et al. (2014). "A monoclonal antibody against neem leaf glycoprotein recognizes carcinoembryonic antigen (CEA) and restricts CEA expressing tumor growth." Journal of Immunotherapy 37(8): 394-406.
[89]Lin, A. V. (2015). "Direct Elisa." ELISA: methods and protocols: 61-67.
[90]Lin, A. V. (2015). "Indirect elisa." ELISA: methods and protocols: 51-59.
[91]Abd Temur, A. and F. A. Rashid (2021). "Irisin and carcinoembryonic antigen (CEA) as potential diagnostic biomarkers in gastric and colorectal cancers." Reports of Biochemistry & Molecular Biology 10(3): 488.
[92]Franken, N. A., et al. (2006). "Clonogenic assay of cells in vitro." Nature protocols 1(5): 2315-2319.
[93]Nakamura, D. (2023). "The evaluation of tumorigenicity and characterization of colonies in a soft agar colony formation assay using polymerase chain reaction." Scientific reports 13(1): 5405.
[94]Gordon, J. L., et al. (2018). "Cell-based methods for determination of efficacy for candidate therapeutics in the clinical management of cancer." Diseases 6(4): 85.
[95]Vargas, J., et al. (2018). "Sequential multiplex immunofluorescence technology (SMIFT): A new staining strategy for immunotherapy." Cancer research 78(13_Supplement): 3965-3965.
[96]Gillespie, J. L., et al. (2016). "A versatile method for immunofluorescent staining of cells cultured on permeable membrane inserts." Medical Science Monitor Basic Research 22: 91.
[97]Zaqout, S., et al. (2020). "Immunofluorescence staining of paraffin sections step by step." Frontiers in neuroanatomy 14: 582218.
[98]Im, K., et al. (2019). "An introduction to performing immunofluorescence staining." Biobanking: methods and protocols: 299-311.
[99]Taube, J. M., et al. (2020). "The Society for Immunotherapy in Cancer statement on best practices for multiplex immunohistochemistry (IHC) and immunofluorescence (IF) staining and validation." Journal for immunotherapy of cancer 8(1). |