Intricate relationship between cancer stemness, metastasis, and drug resistance

Tikam Chand Dakal , Ravi Bhushan , Caiming Xu , Bhana Ram Gadi , Swaranjit Singh Cameotra , Vikas Yadav , Jarek Maciaczyk , Ingo G. H. Schmidt-Wolf , Abhishek Kumar , Amit Sharma

MedComm ›› 2024, Vol. 5 ›› Issue (10) : e710

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (10) : e710 DOI: 10.1002/mco2.710
REVIEW

Intricate relationship between cancer stemness, metastasis, and drug resistance

Author information +
History +
PDF

Abstract

Cancer stem cells (CSCs) are widely acknowledged as the drivers of tumor initiation, epithelial-mesenchymal transition (EMT) progression, and metastasis. Originating from both hematologic and solid malignancies, CSCs exhibit quiescence, pluripotency, and self-renewal akin to normal stem cells, thus orchestrating tumor heterogeneity and growth. Through a dynamic interplay with the tumor microenvironment (TME) and intricate signaling cascades, CSCs undergo transitions from differentiated cancer cells, culminating in therapy resistance and disease recurrence. This review undertakes an in-depth analysis of the multifaceted mechanisms underlying cancer stemness and CSC-mediated resistance to therapy. Intrinsic factors encompassing the TME, hypoxic conditions, and oxidative stress, alongside extrinsic processes such as drug efflux mechanisms, collectively contribute to therapeutic resistance. An exploration into key signaling pathways, including JAK/STAT, WNT, NOTCH, and HEDGEHOG, sheds light on their pivotal roles in sustaining CSCs phenotypes. Insights gleaned from preclinical and clinical studies hold promise in refining drug discovery efforts and optimizing therapeutic interventions, especially chimeric antigen receptor (CAR)-T cell therapy, cytokine-induced killer (CIK) cell therapy, natural killer (NK) cell-mediated CSC-targeting and others. Ultimately use of cell sorting and single cell sequencing approaches for elucidating the fundamental characteristics and resistance mechanisms inherent in CSCs will enhance our comprehension of CSC and intratumor heterogeneity, which ultimately would inform about tailored and personalized interventions.

Keywords

cancer stem cells / cancer stemness / drug resistance / EMT / metastasis / signaling pathways / tumor microenvironment

Cite this article

Download citation ▾
Tikam Chand Dakal, Ravi Bhushan, Caiming Xu, Bhana Ram Gadi, Swaranjit Singh Cameotra, Vikas Yadav, Jarek Maciaczyk, Ingo G. H. Schmidt-Wolf, Abhishek Kumar, Amit Sharma. Intricate relationship between cancer stemness, metastasis, and drug resistance. MedComm, 2024, 5(10): e710 DOI:10.1002/mco2.710

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Torre LA, Siegel RL, Ward EM, et al. Global cancer incidence and mortality rates and trends—an update. Cancer Epidemiol Biomarkers Prev. 2016; 25(1): 16-27.

[2]

Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA A Cancer J Clinicians. 2016; 66(1): 7-30.

[3]

Mathers CD, Loncar D. Projections of global mortality and burden of disease from 2002 to 2030. Samet J, ed. PLoS Med. 2006; 3(11): e442.

[4]

Visvader JE. Cells of origin in cancer. Nature. 2011; 469(7330): 314-322.

[5]

Timp W, Feinberg AP. Cancer as a dysregulated epigenome allowing cellular growth advantage at the expense of the host. Nat Rev Cancer. 2013; 13(7): 497-510.

[6]

Rycaj K, Tang DG. Cell-of-origin of cancer versus cancer stem cells: assays and interpretations. Cancer Res. 2015; 75(19): 4003-4011.

[7]

Coates AS, Winer EP, Goldhirsch A, et al. Tailoring therapies—improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol. 2015; 26(8): 1533-1546.

[8]

Kaseb HO, Fohrer-Ting H, Lewis DW, et al. Identification, expansion and characterization of cancer cells with stem cell properties from head and neck squamous cell carcinomas. Exp Cell Res. 2016; 348(1): 75-86.

[9]

Gedye C, Sirskyj D, Lobo NC, et al. Cancer stem cells are underestimated by standard experimental methods in clear cell renal cell carcinoma. Sci Rep. 2016; 6(1): 25220.

[10]

Cabrera MC. Cancer stem cell plasticity and tumor hierarchy. WJSC. 2015; 7(1): 27.

[11]

Borovski T, De Sousa E Melo F, Vermeulen L, et al. Cancer stem cell niche: the place to be. Cancer Res. 2011; 71(3): 634-639.

[12]

Moore N, Lyle S. Quiescent, slow-cycling stem cell populations in cancer: a review of the evidence and discussion of significance. J Oncol. 2011; 2011: 1-11.

[13]

Bezuidenhout N, Shoshan M. A shifty target: tumor-initiating cells and their metabolism. Int J Mol Sci. 2019; 20(21): 5370.

[14]

Aponte PM, Caicedo A. Stemness in cancer: stem cells, cancer stem cells, and their microenvironment. Stem Cells Int. 2017; 2017: 1-17.

[15]

Wuputra K, Ku CC, Wu DC, et al. Prevention of tumor risk associated with the reprogramming of human pluripotent stem cells. J Exp Clin Cancer Res. 2020; 39(1): 100.

[16]

Visvader JE, Clevers H. Tissue-specific designs of stem cell hierarchies. Nat Cell Biol. 2016; 18(4): 349-355.

[17]

Chaffer CL, Brueckmann I, Scheel C, et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc Natl Acad Sci USA. 2011; 108(19): 7950-7955.

[18]

Plaks V, Kong N, Werb Z. The cancer stem cell niche: how essential is the niche in regulating stemness of tumor cells? Cell Stem Cell. 2015; 16(3): 225-238.

[19]

Chen W, Dong J, Haiech J, et al. Cancer stem cell quiescence and plasticity as major challenges in cancer therapy. Stem Cells Int. 2016; 2016: 1-16.

[20]

Chu X, Tian W, Ning J, et al. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther. 2024; 9(1): 170. Published online 2024.

[21]

Schott AF, Landis MD, Dontu G, et al. Preclinical and clinical studies of gamma secretase inhibitors with docetaxel on human breast tumors. Clin Cancer Res. 2013; 19(6): 1512-1524.

[22]

Patel H, Nilendu P, Jahagirdar D, et al. Modulating secreted components of tumor microenvironment: a masterstroke in tumor therapeutics. Cancer Biol Ther. 2018; 19(1): 3-12.

[23]

Joshi RS, Kanugula SS, Sudhir S, et al. The role of cancer-associated fibroblasts in tumor progression. Cancers. 2021; 13(6): 1399.

[24]

Yamaguchi H, Sakai R. Direct interaction between carcinoma cells and cancer associated fibroblasts for the regulation of cancer invasion. Cancers (Basel). 2015; 7(4): 2054-2062.

[25]

Augsten M. Cancer-associated fibroblasts as another polarized cell type of the tumor microenvironment. Front Oncol. 2014; 4: 62.

[26]

Guo L, Li B, Yang J, et al. Fibroblast-derived exosomal microRNA-369 potentiates migration and invasion of lung squamous cell carcinoma cells via NF1-mediated MAPK signaling pathway. Int J Mol Med. 2020; 46(2): 595-608.

[27]

Ren J, Ding L, Zhang D, et al. Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19. Theranostics. 2018; 8(14): 3932-3948.

[28]

Lacina L, Plzak J, Kodet O, et al. Cancer microenvironment: what can we learn from the stem cell niche. Int J Mol Sci. 2015; 16(10): 24094-24110.

[29]

Chen WJ, Ho CC, Chang YL, et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat Commun. 2014; 5(1): 3472.

[30]

Paraiso KHT, Smalley KSM. Fibroblast-mediated drug resistance in cancer. Biochem Pharmacol. 2013; 85(8): 1033-1041.

[31]

Valcz G, Sipos F, Tulassay Z, et al. Importance of carcinoma-associated fibroblast-derived proteins in clinical oncology. J Clin Pathol. 2014; 67(12): 1026-1031.

[32]

Choe C, Shin YS, Kim SH, et al. Tumor–stromal interactions with direct cell contacts enhance motility of non-small cell lung cancer cells through the hedgehog signaling pathway. Anticanc Res. 2013; 33(9): 3715-3723.

[33]

Borah A, Raveendran S, Rochani A, et al. Targeting self-renewal pathways in cancer stem cells: clinical implications for cancer therapy. Oncogenesis. 2015; 4(11): e177-e177.

[34]

Quail DF, Joyce JA. The microenvironmental landscape of brain tumors. Cancer Cell. 2017; 31(3): 326-341.

[35]

Osman A, Afify SM, Hassan G, et al. Revisiting cancer stem cells as the origin of cancer-associated cells in the tumor microenvironment: a hypothetical view from the potential of iPSCs. Cancers. 2020; 12(4): 879.

[36]

Evans J, Essex A, Xin H, et al. Registered report: wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Gilbertson RJ, ed. eLife. 2015; 4: e07301.

[37]

Vermeulen L, De Sousa E Melo F, van der Heijden M, et al. Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 2010; 12(5): 468-476.

[38]

Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell. 2010; 40(2): 294-309.

[39]

Yang MH, Wu MZ, Chiou SH, et al. Direct regulation of TWIST by HIF-1α promotes metastasis. Nat Cell Biol. 2008; 10(3): 295-305.

[40]

Lester RD, Jo M, Montel V, et al. uPAR induces epithelial–mesenchymal transition in hypoxic breast cancer cells. J Cell Biol. 2007; 178(3): 425-436.

[41]

Imai T, Horiuchi A, Wang C, et al. Hypoxia attenuates the expression of E-cadherin via up-regulation of SNAIL in ovarian carcinoma cells. Am J Pathol. 2003; 163(4): 1437-1447.

[42]

Kim Y, Lin Q, Zelterman D, et al. Hypoxia-regulated delta-like 1 homologue enhances cancer cell stemness and tumorigenicity. Cancer Res. 2009; 69(24): 9271-9280.

[43]

Keith B, Simon MC. Hypoxia-inducible factors, stem cells, and cancer. Cell. 2007; 129(3): 465-472.

[44]

Krishnamachary B, Zagzag D, Nagasawa H, et al. Hypoxia-inducible factor-1-dependent repression of E-cadherin in von Hippel-Lindau tumor suppressor–null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1B. Cancer Res. 2006; 66(5): 2725-2731.

[45]

Esteban MA, Tran MGB, Harten SK, et al. Regulation of E-cadherin expression by VHL and hypoxia-inducible factor. Cancer Res. 2006; 66(7): 3567-3575.

[46]

Wang K, Zhang T, Dong Q, et al. Redox homeostasis: the linchpin in stem cell self-renewal and differentiation. Cell Death Dis. 2013; 4(3): e537-e537.

[47]

Skvortsova I, Debbage P, Kumar V, et al. Radiation resistance: cancer stem cells (CSCs) and their enigmatic pro-survival signaling. Semin Cancer Biol. 2015; 35: 39-44.

[48]

Zhao H, Duan Q, Zhang Z, et al. Up-regulation of glycolysis promotes the stemness and EMT phenotypes in gemcitabine-resistant pancreatic cancer cells. J Cell Mol Med. 2017; 21(9): 2055-2067.

[49]

Versini A, Colombeau L, Hienzsch A, et al. Salinomycin derivatives kill breast cancer stem cells by lysosomal iron targeting. Chem A Eur J. 2020; 26(33): 7416-7424.

[50]

geu RyooI, n, hyun ChoiB, Ku SK, et al. High CD44 expression mediates p62-associated NFE2L2/NRF2 activation in breast cancer stem cell-like cells: implications for cancer stem cell resistance. Redox Biol. 2018; 17: 246-258.

[51]

Liao J, Liu PP, Hou G, et al. Regulation of stem-like cancer cells by glutamine through β-catenin pathway mediated by redox signaling. Mol Cancer. 2017; 16(1): 51.

[52]

Lee KM, Giltnane JM, Balko JM, et al. MYC and MCL1 cooperatively promote chemotherapy-resistant breast cancer stem cells via regulation of mitochondrial oxidative phosphorylation. Cell Metab. 2017; 26(4): 633-647. e7.

[53]

Hwang RF, Moore T, Arumugam T, et al. Cancer-Associated stromal fibroblasts promote pancreatic tumor progression. Cancer Res. 2008; 68(3): 918-926.

[54]

Mao Y, Keller ET, Garfield DH, et al. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev. 2013; 32(1-2): 303-315.

[55]

Calon A, Espinet E, Palomo-Ponce S, et al. Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell. 2012; 22(5): 571-584.

[56]

Takebe N, Miele L, Harris PJ, et al. Targeting Notch, Hedgehog, and Wnt pathways in cancer stem cells: clinical update. Nat Rev Clin Oncol. 2015; 12(8): 445-464.

[57]

Dandawate PR, Subramaniam D, Jensen RA, et al. Targeting cancer stem cells and signaling pathways by phytochemicals: novel approach for breast cancer therapy. Semin Cancer Biol. 2016; 40-41: 192-208.

[58]

Thomas SJ, Snowden JA, Zeidler MP, et al. The role of JAK/STAT signalling in the pathogenesis, prognosis and treatment of solid tumours. Br J Cancer. 2015; 113(3): 365-371.

[59]

Kim JH, Yoon MS, Chen J. Signal transducer and activator of transcription 3 (STAT3) mediates amino acid inhibition of insulin signaling through serine 727 phosphorylation. J Biol Chem. 2009; 284(51): 35425-35432.

[60]

Carpenter RL, Lo HW. STAT3 target genes relevant to human cancers. Cancers (Basel). 2014; 6(2): 897-925.

[61]

Galoczova M, Coates P, Vojtesek B. STAT3, stem cells, cancer stem cells and p63. Cell Mol Biol Lett. 2018; 23: 12.

[62]

Yoshimatsu T, Kawaguchi D, Oishi K, et al. Non-cell-autonomous action of STAT3 in maintenance of neural precursor cells in the mouse neocortex. Development. 2006; 133(13): 2553-2563.

[63]

Peng L, Jiang D. Resveratrol eliminates cancer stem cells of osteosarcoma by STAT3 pathway inhibition. PLoS One. 2018; 13(10): e0205918.

[64]

Liu Y, Gao X, Wang S, et al. Cancer stem cells are regulated by STAT3 signalling in wilms tumour. J Cancer. 2018; 9(8): 1486-1499.

[65]

Lin L, Jou D, Wang Y, et al. STAT3 as a potential therapeutic target in ALDH+ and CD44+/CD24+ stem cell-like pancreatic cancer cells. Int J Oncol. 2016; 49(6): 2265-2274.

[66]

Shastri A, Choudhary G, Teixeira M, et al. Antisense STAT3 inhibitor decreases viability of myelodysplastic and leukemic stem cells. J Clin Invest. 2018; 128(12): 5479-5488.

[67]

Sherry MM, Reeves A, Wu JK, et al. STAT3 is required for proliferation and maintenance of multipotency in glioblastoma stem cells. Stem Cells. 2009; 27(10): 2383-2392.

[68]

Gupta PB, Chaffer CL, Weinberg RA. Cancer stem cells: mirage or reality? Nat Med. 2009; 15(9): 1010-1012.

[69]

Zhou J, Wulfkuhle J, Zhang H, et al. Activation of the PTEN/mTOR/STAT3 pathway in breast cancer stem-like cells is required for viability and maintenance. Proc Natl Acad Sci USA. 2007; 104(41): 16158-16163.

[70]

Yang W, Liu Y, Gao R, et al. HDAC6 inhibition induces glioma stem cells differentiation and enhances cellular radiation sensitivity through the SHH/Gli1 signaling pathway. Cancer Lett. 2018; 415: 164-176.

[71]

Kahn M. Can we safely target the WNT pathway? Nat Rev Drug Discov. 2014; 13(7): 513-532.

[72]

Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. 2017; 36(11): 1461-1473.

[73]

Malhotra S, Kincade PW. Wnt-related molecules and signaling pathway equilibrium in hematopoiesis. Cell Stem Cell. 2009; 4(1): 27-36.

[74]

Andl T, Reddy ST, Gaddapara T, et al. WNT signals are required for the initiation of hair follicle development. Dev Cell. 2002; 2(5): 643-653.

[75]

Jaks V, Barker N, Kasper M, et al. Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat Genet. 2008; 40(11): 1291-1299.

[76]

Wang R, Sun Q, Wang P, et al. Notch and Wnt/β-catenin signaling pathway play important roles in activating liver cancer stem cells. Oncotarget. 2015; 7(5): 5754-5768.

[77]

Pramanik KC, Fofaria NM, Gupta P, et al. Inhibition of β-Catenin signaling suppresses pancreatic tumor growth by disrupting nuclear β-Catenin/TCF-1 complex: critical role of STAT-3. Oncotarget. 2015; 6(13): 11561-11574.

[78]

Li J, Ji L, Chen J, et al. Wnt/β-catenin signaling pathway in skin carcinogenesis and therapy. Biomed Res Int. 2015; 2015: 964842.

[79]

Stewart DJ. Wnt signaling pathway in non–small cell lung cancer. J Natl Cancer Inst. 2014; 106(1): djt356.

[80]

Chen Z, Zhou L, Wang L, et al. HBO1 promotes cell proliferation in bladder cancer via activation of Wnt/β-catenin signaling. Mol Carcinog. 2018; 57(1): 12-21.

[81]

Valkenburg KC, Graveel CR, Zylstra-Diegel CR, et al. Wnt/β-catenin signaling in normal and cancer stem cells. Cancers (Basel). 2011; 3(2): 2050-2079.

[82]

de Sousa e Melo F, Vermeulen L. Wnt signaling in cancer stem cell biology. Cancers (Basel). 2016; 8(7): 60.

[83]

Prasetyanti PR, Zimberlin CD, Bots M, et al. Regulation of stem cell self-renewal and differentiation by Wnt and Notch are conserved throughout the adenoma-carcinoma sequence in the colon. Mol Cancer. 2013; 12(1): 126.

[84]

Liu S, KP U, Zhang J, et al. R-spodin2 enhances canonical Wnt signaling to maintain the stemness of glioblastoma cells. Cancer Cell Int. 2018; 18: 156.

[85]

Wang Z, Ahmad A, Li Y, et al. Targeting notch to eradicate pancreatic cancer stem cells for cancer therapy. Anticancer Res. 2011; 31(4): 1105-1113.

[86]

Koulis A, Buckle A, Boussioutas A. Premalignant lesions and gastric cancer: current understanding. World J Gastrointest Oncol. 2019; 11(9): 665-678.

[87]

Fortini ME. Notch signaling: the core pathway and its posttranslational regulation. Dev Cell. 2009; 16(5): 633-647.

[88]

Xiao W, Gao Z, Duan Y, et al. Notch signaling plays a crucial role in cancer stem-like cells maintaining stemness and mediating chemotaxis in renal cell carcinoma. J Exp Clin Cancer Res. 2017; 36: 41.

[89]

Chiba S. Concise review: notch signaling in stem cell systems. Stem Cells. 2006; 24(11): 2437-2447.

[90]

Wang R, Li Y, Tsung A, et al. iNOS promotes CD24+CD133+ liver cancer stem cell phenotype through a TACE/ADAM17-dependent Notch signaling pathway. Proc Natl Acad Sci USA. 2018; 115(43): E10127-E10136.

[91]

Yan Y, Liu F, Han L, et al. HIF-2α promotes conversion to a stem cell phenotype and induces chemoresistance in breast cancer cells by activating Wnt and Notch pathways. J Exp Clin Cancer Res. 2018; 37(1): 256.

[92]

Yu JB, Jiang H, Zhan RY. Aberrant Notch signaling in glioblastoma stem cells contributes to tumor recurrence and invasion. Mol Med Rep. 2016: 1263-1268.

[93]

Barnawi R, Al-Khaldi S, Majed Sleiman G, et al. Fascin is critical for the maintenance of breast cancer stem cell pool predominantly via the activation of the notch self-renewal pathway. Stem Cells. 2016; 34(12): 2799-2813.

[94]

Zeng F, Chen H, Zhang Z, et al. Regulating glioma stem cells by hypoxia through the Notch1 and Oct3/4 signaling pathway. Onco Lett. 2018; 16(5): 6315-6322.

[95]

Bayin NS, Frenster JD, Sen R, et al. Notch signaling regulates metabolic heterogeneity in glioblastoma stem cells. Oncotarget. 2017; 8(39): 64932-64953.

[96]

Merchant A, Matsui W. Targeting hedgehog—a cancer stem cell pathway. Clin Cancer Res. 2010; 16(12): 3130-3140.

[97]

Clement V, Sanchez P, de Tribolet N, et al. HEDGEHOG-GLI1 signaling regulates human glioma growth, cancer stem cell self-renewal and tumorigenicity. Curr Biol. 2007; 17(2): 165-172.

[98]

Ranjan T, Sengupta S, Glantz MJ, et al. Cancer stem cell assay-guided chemotherapy improves survival of patients with recurrent glioblastoma in a randomized trial. Cell Rep Med. 2023; 4(5): 101025.

[99]

Peacock CD, Wang Q, Gesell GS, et al. Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma. Proc Natl Acad Sci USA. 2007; 104(10): 4048-4053.

[100]

Stuart SA, Minami Y, Wang JYJ. The CML stem cell: evolution of the progenitor. Cell Cycle. 2009; 8(9): 1338-1343.

[101]

Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest. 2003; 112(12): 1776-1784.

[102]

Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002; 2(6): 442-454.

[103]

Creighton CJ, Li X, Landis M, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA. 2009; 106(33): 13820-13825.

[104]

Schmidt S, Friedl P. Interstitial cell migration: integrin-dependent and alternative adhesion mechanisms. Cell Tissue Res. 2009; 339(1): 83-92.

[105]

Hsiao CT, Cheng HW, Huang CM, et al. Fibronectin in cell adhesion and migration via N-glycosylation. Oncotarget. 2017; 8(41): 70653-70668.

[106]

Kim WT, Ryu CJ. Cancer stem cell surface markers on normal stem cells. BMB Rep. 2017; 50(6): 285-298.

[107]

Zhao W, Li Y, Zhang X. Stemness-related markers in cancer. Cancer Transl Med. 2017; 3(3): 87-95.

[108]

Nakahata K, Uehara S, Nishikawa S, et al. Aldehyde dehydrogenase 1 (ALDH1) is a potential marker for cancer stem cells in embryonal rhabdomyosarcoma. PLoS One. 2015; 10(4): e0125454.

[109]

Clark DW, Palle K. Aldehyde dehydrogenases in cancer stem cells: potential as therapeutic targets. Ann Transl Med. 2016; 4(24): 518.

[110]

Liu T, Kong WX, Tang XY, et al. The transcription factor Zfp90 regulates the self-renewal and differentiation of hematopoietic stem cells. Cell Death Dis. 2018; 9(6): 1-12.

[111]

Chen G, Yin S, Zeng H, et al. Regulation of embryonic stem cell self-renewal. Life (Basel). 2022; 12(8): 1151.

[112]

Varzideh F, Gambardella J, Kansakar U, et al. Molecular mechanisms underlying pluripotency and self-renewal of embryonic stem cells. Int J Mol Sci. 2023; 24(9): 8386.

[113]

Liu A, Yu X, Liu S. Pluripotency transcription factors and cancer stem cells: small genes make a big difference. Chin J Cancer. 2013; 32(9): 483-487.

[114]

Tsai CC, Hung SC. Functional roles of pluripotency transcription factors in mesenchymal stem cells. Cell Cycle. 2012; 11(20): 3711-3712.

[115]

Zhang W, Sui Y, Ni J, et al. Insights into the Nanog gene: a propeller for stemness in primitive stem cells. Int J Biol Sci. 2016; 12(11): 1372-1381.

[116]

Yang L, Shi P, Zhao G, et al. Targeting cancer stem cell pathways for cancer therapy. Signal Transduct Target Ther. 2020; 5: 8.

[117]

Basu S, Cheriyamundath S, Ben-Ze’ev A. Cell–cell adhesion: linking Wnt/β-catenin signaling with partial EMT and stemness traits in tumorigenesis. F1000Res. 2018; 7. F1000 Faculty Rev-1488.

[118]

Janiszewska M, Primi MC, Izard T. Cell adhesion in cancer: beyond the migration of single cells. J Biol Chem. 2020; 295(8): 2495-2505.

[119]

Gires O, Pan M, Schinke H, et al. Expression and function of epithelial cell adhesion molecule EpCAM: where are we after 40 years? Cancer Metastasis Rev. 2020; 39(3): 969-987.

[120]

Mrozik KM, Blaschuk OW, Cheong CM, et al. N-cadherin in cancer metastasis, its emerging role in haematological malignancies and potential as a therapeutic target in cancer. BMC Cancer. 2018; 18(1): 939.

[121]

Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial–mesenchymal transition. Nat Rev Mol Cell Biol. 2014; 15(3): 178-196.

[122]

Sharma P, Alsharif S, Fallatah A, et al. Intermediate filaments as effectors of cancer development and metastasis: a focus on keratins, vimentin, and nestin. Cells. 2019; 8(5): 497.

[123]

Xu Q, Deng F, Qin Y, et al. Long non-coding RNA regulation of epithelial–mesenchymal transition in cancer metastasis. Cell Death Dis. 2016; 7(6): e2254-e2254.

[124]

Usman S, Waseem NH, Nguyen TKN, et al. Vimentin is at the heart of epithelial mesenchymal transition (EMT) mediated metastasis. Cancers (Basel). 2021; 13(19): 4985.

[125]

Xu J, Lamouille S, Derynck R. TGF-β-induced epithelial to mesenchymal transition. Cell Res. 2009; 19(2): 156-172.

[126]

Serrano-Gomez SJ, Maziveyi M, Alahari SK. Regulation of epithelial-mesenchymal transition through epigenetic and post-translational modifications. Mol Cancer. 2016; 15(1): 18.

[127]

Ingruber J, Dudás J, Savic D, et al. EMT-related transcription factors and protein stabilization mechanisms involvement in cadherin switch of head and neck squamous cell carcinoma. Exp Cell Res. 2022; 414(1): 113084.

[128]

Debnath P, Huirem RS, Dutta P, et al. Epithelial–mesenchymal transition and its transcription factors. Biosci Rep. 2021; 42(1): BSR20211754.

[129]

Ansieau S, Collin G, Hill L. EMT or EMT-promoting transcription factors, where to focus the light? Front Oncol. 2014; 4: 353.

[130]

Seo J, Ha J, Kang E, et al. The role of epithelial–mesenchymal transition-regulating transcription factors in anti-cancer drug resistance. Arch Pharmacal Res. 2021; 44(3): 281-292.

[131]

Qin Q, Xu Y, He T, et al. Normal and disease-related biological functions of Twist1 and underlying molecular mechanisms. Cell Res. 2012; 22(1): 90-106.

[132]

Jena MK, Janjanam J. Role of extracellular matrix in breast cancer development: a brief update. F1000Res. 2018; 7: 274.

[133]

Pote MS, Gacche RN. ATP-binding cassette efflux transporters and MDR in cancer. Drug Discov Today. 2023; 28(5): 103537.

[134]

Choi YH, Yu AM. ABC transporters in multidrug resistance and pharmacokinetics, and strategies for drug development. Curr Pharm Des. 2014; 20(5): 793-807.

[135]

Choi CH. ABC transporters as multidrug resistance mechanisms and the development of chemosensitizers for their reversal. Cancer Cell Int. 2005; 5: 30.

[136]

Sun YL, Patel A, Kumar P, et al. Role of ABC transporters in cancer chemotherapy. Chin J Cancer. 2012; 31(2): 51-57.

[137]

Phi LTH, Sari IN, Yang YG, et al. Cancer stem cells (CSCs) in drug resistance and their therapeutic implications in cancer treatment. Stem Cells Int. 2018; 2018: 5416923.

[138]

Ciccone V, Morbidelli L, Ziche M, et al. How to conjugate the stemness marker ALDH1A1 with tumor angiogenesis, progression, and drug resistance. Cancer Drug Resist. 2020; 3(1): 26-37.

[139]

Chai JY, Sugumar V, Alshanon AF, et al. Defining the Role of GLI/Hedgehog signaling in chemoresistance: implications in therapeutic approaches. Cancers (Basel). 2021; 13(19): 4746.

[140]

Lu Y, Zhu Y, Deng S, et al. Targeting the sonic hedgehog pathway to suppress the expression of the cancer stem cell (CSC)—related transcription factors and CSC-driven thyroid tumor growth. Cancers (Basel). 2021; 13(3): 418.

[141]

Shi Q, Xue C, Zeng Y, et al. Notch signaling pathway in cancer: from mechanistic insights to targeted therapies. Signal Transduct Target Ther. 2024; 9(1): 1-37.

[142]

You WK, Schuetz TJ, Lee SH. Targeting the DLL/Notch signaling pathway in cancer: challenges and advances in clinical development. Mol Cancer Ther. 2023; 22(1): 3-11.

[143]

Mohammad RM, Muqbil I, Lowe L, et al. Broad targeting of resistance to apoptosis in cancer. Semin Cancer Biol. 2015; 35: S78-S103.

[144]

Rathore R, McCallum JE, Varghese E, et al. Overcoming chemotherapy drug resistance by targeting inhibitors of apoptosis proteins (IAPs). Apoptosis. 2017; 22(7): 898-919.

[145]

Kaloni D, Diepstraten ST, Strasser A, et al. BCL-2 protein family: attractive targets for cancer therapy. Apoptosis. 2023; 28(1-2): 20-38.

[146]

Qian S, Wei Z, Yang W, et al. The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front Oncol. 2022; 12: 985363.

[147]

Fu M, Hu Y, Lan T, et al. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther. 2022; 7(1): 1-20.

[148]

Talukdar PD, Chatterji U. Transcriptional co-activators: emerging roles in signaling pathways and potential therapeutic targets for diseases. Signal Transduct Target Ther. 2023; 8: 427.

[149]

Zhang H, Steed A, Co M, et al. Cancer stem cells, epithelial-mesenchymal transition, ATP and their roles in drug resistance in cancer. Cancer Drug Resist. 2021; 4(3): 684-709.

[150]

Shibue T, Weinberg RA. EMT, CSCs, and drug resistance: the mechanistic link and clinical implications. Nat Rev Clin Oncol. 2017; 14(10): 611-629.

[151]

Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci. 2012; 125(23): 5591-5596.

[152]

Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008; 14(6): 818-829.

[153]

Lindsey S, Langhans SA. Crosstalk of oncogenic signaling pathways during epithelial-mesenchymal transition. Front Oncol. 2014; 4: 358.

[154]

Shi Y, Massagué J. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell. 2003; 113(6): 685-700.

[155]

Jechlinger M, Grunert S, Tamir IH, et al. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003; 22(46): 7155-7169.

[156]

Andor N, Graham TA, Jansen M, et al. Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat Med. 2016; 22(1): 105-113.

[157]

Tanabe S, Quader S, Cabral H, et al. Interplay of EMT and CSC in cancer and the potential therapeutic strategies. Front Pharmacol. 2020; 11: 904.

[158]

Davies KJ. The complex interaction of matrix metalloproteinases in the migration of cancer cells through breast tissue stroma. Int J Breast Cancer. 2014; 2014: 1-5.

[159]

Radisky ES, Radisky DC. Matrix metalloproteinase-induced epithelial-mesenchymal transition in breast cancer. J Mammary Gland Biol Neoplasia. 2010; 15(2): 201-212.

[160]

Lee M, Vasioukhin V. Cell polarity and cancer—cell and tissue polarity as a non-canonical tumor suppressor. J Cell Sci. 2008; 121(8): 1141-1150.

[161]

Chatterjee S, Seifried L, Feigin ME, et al. Dysregulation of cell polarity proteins synergize with oncogenes or the microenvironment to induce invasive behavior in epithelial cells. Schneider-Stock R, ed. PLoS One. 2012; 7(4): e34343.

[162]

Finak G, Bertos N, Pepin F, et al. Stromal gene expression predicts clinical outcome in breast cancer. Nat Med. 2008; 14(5): 518-527.

[163]

Noë V, Fingleton B, Jacobs K, et al. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J Cell Sci. 2001; 114(1): 111-118.

[164]

Hu X, Li D, Zhang W, et al. Matrix metalloproteinase-9 expression correlates with prognosis and involved in ovarian cancer cell invasion. Arch Gynecol Obstet. 2012; 286(6): 1537-1543.

[165]

Li HC, Cao DC, Liu Y, et al. Prognostic value of matrix metalloproteinases (MMP-2 and MMP-9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 2004; 88(1): 75-85.

[166]

Inoue T, Yashiro M, Nishimura S, et al. Matrix metalloproteinase-1 expression is a prognostic factor for patients with advanced gastric cancer. Int J Mol Med. 1999; 4(1): 73-77.

[167]

Brabletz T, Jung A, Reu S, et al. Variable β-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA. 2001; 98(18): 10356-10361.

[168]

Yu M, Bardia A, Wittner BS, et al. Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013; 339(6119): 580-584.

[169]

Lecharpentier A, Vielh P, Perez-Moreno P, et al. Detection of circulating tumour cells with a hybrid (epithelial/mesenchymal) phenotype in patients with metastatic non-small cell lung cancer. Br J Cancer. 2011; 105(9): 1338-1341.

[170]

Ruscetti M, Quach B, Dadashian EL, et al. Tracking and functional characterization of epithelial-mesenchymal transition and mesenchymal tumor cells during prostate cancer metastasis. Cancer Res. 2015; 75(13): 2749-2759.

[171]

Rhim AD, Mirek ET, Aiello NM, et al. EMT and dissemination precede pancreatic tumor formation. Cell. 2012; 148(1-2): 349-361.

[172]

Aceto N, Bardia A, Miyamoto DT, et al. Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell. 2014; 158(5): 1110-1122.

[173]

Podsypanina K, Du YCN, Jechlinger M, et al. Seeding and propagation of untransformed mouse mammary cells in the lung. Science. 2008; 321(5897): 1841-1844.

[174]

Jolly MK, Boareto M, Huang B, et al. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015; 5: 155.

[175]

Liu L, Salnikov AV, Bauer N, et al. Triptolide reverses hypoxia-induced epithelial–mesenchymal transition and stem-like features in pancreatic cancer by NF-κB downregulation. Int J Cancer. 2014; 134(10): 2489-2503.

[176]

Lai M, Liu L, Zhu L, et al. Triptolide reverses epithelial-mesenchymal transition in glioma cells via inducing autophagy. Ann Transl Med. 2021; 9(16): 1304.

[177]

Celià-Terrassa T, Meca-Cortés Ó, Mateo F, et al. Epithelial-mesenchymal transition can suppress major attributes of human epithelial tumor-initiating cells. J Clin Invest. 2012; 122(5): 1849-1868.

[178]

Weiswald LB, Bellet D, Dangles-Marie V. Spherical cancer models in tumor biology. Neoplasia. 2015; 17(1): 1-15.

[179]

Ocaña OH, Córcoles R, Fabra Á, et al. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012; 22(6): 709-724.

[180]

Bedard PL, Hansen AR, Ratain MJ, et al. Tumour heterogeneity in the clinic. Nature. 2013; 501(7467): 355-364.

[181]

McGranahan N, Swanton C. Biological and therapeutic impact of intratumor heterogeneity in cancer evolution. Cancer Cell. 2015; 27(1): 15-26.

[182]

Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem Cell. 2014; 14(3): 275-291.

[183]

Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013; 19(11): 1423-1437.

[184]

McGranahan N, Furness AJS, Rosenthal R, et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science. 2016; 351(6280): 1463-1469.

[185]

Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer. Science. 2015; 348(6230): 124-128.

[186]

Germano G, Lamba S, Rospo G, et al. Inactivation of DNA repair triggers neoantigen generation and impairs tumour growth. Nature. 2017; 552(7683): 116-120.

[187]

Greaves M. Cancer stem cells as ‘units of selection’. Evol Appl. 2013; 6(1): 102-108.

[188]

Thorsson V, Gibbs DL, Brown SD, et al. The immune landscape of cancer. Immunity. 2018; 48(4): 812-830. e14.

[189]

Xiao M, Zhang X, Zhang D, et al. Complex interaction and heterogeneity among cancer stem cells in head and neck squamous cell carcinoma revealed by single-cell sequencing. Front Immunol. 2022; 13: 1050951.

[190]

Dirkse A, Golebiewska A, Buder T, et al. Stem cell-associated heterogeneity in Glioblastoma results from intrinsic tumor plasticity shaped by the microenvironment. Nat Commun. 2019; 10(1): 1787.

[191]

Smit MA, Peeper DS. Epithelial-mesenchymal transition and senescence: two cancer-related processes are crossing paths. Aging (Albany NY). 2010; 2(10): 735-741.

[192]

Fischer KR, Durrans A, Lee S, et al. EMT is not required for lung metastasis but contributes to chemoresistance. Nature. 2015; 527(7579): 472-476. Published online November 26, 2015.

[193]

Ohashi S, Natsuizaka M, Wong GS, et al. EGFR and mutant p53 expand esophageal cellular subpopulation capable of epithelial-to-mesenchymal transition through ZEB transcription factors. Cancer Res. 2010; 70(10): 4174-4184.

[194]

Ansieau S, Bastid J, Doreau A, et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell. 2008; 14(1): 79-89.

[195]

Robson EJD, Khaled WT, Abell K, et al. Epithelial-to-mesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation. 2006; 74(5): 254-264.

[196]

Bellomo C, Caja L, Fabregat I, et al. Snail mediates crosstalk between TGFβ and LXRα in hepatocellular carcinoma. Cell Death Differ. 2018; 25(5): 885-903.

[197]

Soundararajan R, Fradette JJ, Konen JM, et al. Targeting the interplay between epithelial-to-mesenchymal-transition and the immune system for effective immunotherapy. Cancers (Basel). 2019; 11(5): 714.

[198]

Shintani Y, Okimura A, Sato K, et al. Epithelial to mesenchymal transition is a determinant of sensitivity to chemoradiotherapy in non-small cell lung cancer. Ann Thorac Surg. 2011; 92(5): 1794-1804.

[199]

Kawamoto A, Yokoe T, Tanaka K, et al. Radiation induces epithelial-mesenchymal transition in colorectal cancer cells. Oncol Rep. 2012; 27(1): 51-57.

[200]

Aiello N, Brabletz T, Kang Y, et al. Upholding a role for EMT in pancreatic cancer metastasis. Nature. 2017; 547(7661): E7-E8.

[201]

Wellner U, Brabletz T, Keck T. ZEB1 in Pancreatic Cancer. Cancers (Basel). 2010; 2(3): 1617-1628.

[202]

Pradella D, Naro C, Sette C, et al. EMT and stemness: flexible processes tuned by alternative splicing in development and cancer progression. Mol Cancer. 2017; 16(1): 8.

[203]

Kurrey NK, Jalgaonkar SP, Joglekar AV, et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells. 2009; 27(9): 2059-2068.

[204]

Visvader JE, Lindeman GJ. Cancer stem cells: current status and evolving complexities. Cell Stem Cell. 2012; 10(6): 717-728.

[205]

Fuchs E, Tumbar T, Guasch G. Socializing with the neighbors: stem cells and their niche. Cell. 2004; 116(6): 769-778.

[206]

Lau EYT, Ho NPY, Lee TKW. Cancer stem cells and their microenvironment: biology and therapeutic implications. Stem Cells Int. 2017; 2017: 1-11.

[207]

Shepard HM. Breaching the Castle Walls: hyaluronan depletion as a therapeutic approach to cancer therapy. Front Oncol. 2015; 5: 192.

[208]

Okuda H, Kobayashi A, Xia B, et al. Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of breast cancer stem–like cells with macrophages and stromal cells. Cancer Res. 2012; 72(2): 537-547.

[209]

Lathia JD, Li M, Hall PE, et al. Laminin alpha 2 enables glioblastoma stem cell growth. Ann Neurol. 2012; 72(5): 766-778.

[210]

Bourguignon LYW, Wong G, Earle C, et al. Hyaluronan-CD44v3 interaction with Oct4-Sox2-Nanog promotes miR-302 expression leading to self-renewal, clonal formation, and cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma. J Biol Chem. 2012; 287(39): 32800-32824.

[211]

Batlle E, Clevers H. Cancer stem cells revisited. Nat Med. 2017; 23(10): 1124-1134.

[212]

Awasthi R, Roseblade A, Hansbro PM, et al. Nanoparticles in cancer treatment: opportunities and obstacles. CDT. 2018; 19(14): 1696-1709.

[213]

Azevedo-Pereira RL, Rangel B, Tovar-Moll F, et al. Superparamagnetic iron oxide nanoparticles as a tool to track mouse neural stem cells in vivo. Mol Biol Rep. 2019; 46(1): 191-198.

[214]

Seebacher NA, Krchniakova M, Stacy AE, et al. Tumour microenvironment stress promotes the development of drug resistance. Antioxidants (Basel). 2021; 10(11): 1801.

[215]

Mitra A, Mishra L, EMT LiS. CTCs and CSCs in tumor relapse and drug-resistance. Oncotarget. 2015; 6(13): 10697-10711.

[216]

Reda A, Hosseiny S, El-Sherbiny IM. Next-generation nanotheranostics targeting cancer stem cells. Nanomedicine (Lond). 2019; 14(18): 2487-2514.

[217]

Hou Y, Zhu Q, Li Z, et al. The FOXM1–ABCC5 axis contributes to paclitaxel resistance in nasopharyngeal carcinoma cells. Cell Death Dis. 2017; 8(3): e2659.

[218]

Jiang Y, He Y, Li H, et al. Expressions of putative cancer stem cell markers ABCB1, ABCG2, and CD133 are correlated with the degree of differentiation of gastric cancer. Gastric Cancer. 2012; 15(4): 440-450.

[219]

Zhou S, Schuetz JD, Bunting KD, et al. The ABC transporter Bcrp1/ABCG2 is expressed in a wide variety of stem cells and is a molecular determinant of the side-population phenotype. Nat Med. 2001; 7(9): 1028-1034.

[220]

Wang XK, He JH, Xu JH, et al. Afatinib enhances the efficacy of conventional chemotherapeutic agents by eradicating cancer stem–like cells. Cancer Res. 2014; 74(16): 4431-4445.

[221]

Das B, Pal B, Bhuyan R, et al. MYC regulates the HIF-2α stemness pathway via Nanog and Sox2 to maintain self-renewal in cancer stem cells versus non-stem cancer cells. Cancer Res. 2019; 79(16): 4015-4025.

[222]

Sasaki N, Ishiwata T, Hasegawa F, et al. Stemness and anti-cancer drug resistance in ATP-binding cassette subfamily G member 2 highly expressed pancreatic cancer is induced in 3D culture conditions. Cancer Sci. 2018; 109(4): 1135-1146.

[223]

Patrawala L, Calhoun T, Schneider-Broussard R, et al. Side population is enriched in tumorigenic, stem-like cancer cells, whereas abcg2+ and abcg2– cancer cells are similarly tumorigenic. Cancer Res. 2005; 65(14): 6207-6219.

[224]

Miranda-Lorenzo I, Dorado J, Lonardo E, et al. Intracellular autofluorescence: a biomarker for epithelial cancer stem cells. Nat Methods. 2014; 11(11): 1161-1169.

[225]

Grimm M, Krimmel M, Polligkeit J, et al. ABCB5 expression and cancer stem cell hypothesis in oral squamous cell carcinoma. Eur J Cancer. 2012; 48(17): 3186-3197.

[226]

El-Khattouti A, Sheehan NT, Monico J, et al. CD133+ melanoma subpopulation acquired resistance to caffeic acid phenethyl ester-induced apoptosis is attributed to the elevated expression of ABCB5: significance for melanoma treatment. Cancer Lett. 2015; 357(1): 83-104.

[227]

Wilson BJ, Saab KR, Ma J, et al. ABCB5 maintains melanoma-initiating cells through a pro-inflammatory cytokine signaling circuit. Cancer Res. 2014; 74(15): 4196-4207.

[228]

Lee CAA, Banerjee P, Wilson BJ, et al. Targeting the ABC transporter ABCB5 sensitizes glioblastoma to temozolomide-induced apoptosis through a cell-cycle checkpoint regulation mechanism. J Biol Chem. 2020; 295(22): 7774-7788.

[229]

Sugano T, Seike M, Noro R, et al. Inhibition of ABCB1 overcomes cancer stem cell–like properties and acquired resistance to MET inhibitors in non–small cell lung cancer. Mol Cancer Ther. 2015; 14(11): 2433-2440.

[230]

Huang B, Fu SJ, Fan WZ, et al. PKCϵ inhibits isolation and stemness of side population cells via the suppression of ABCB1 transporter and PI3K/Akt, MAPK/ERK signaling in renal cell carcinoma cell line 769P. Cancer Lett. 2016; 376(1): 148-154.

[231]

Wu ZX, Teng QX, Cai CY, et al. Tepotinib reverses ABCB1-mediated multidrug resistance in cancer cells. Biochem Pharmacol. 2019; 166: 120-127.

[232]

Tsou SH, Chen TM, Hsiao HT, et al. A critical dose of doxorubicin is required to alter the gene expression profiles in MCF-7 cells acquiring multidrug resistance. PLoS One. 2015; 10(1): e0116747.

[233]

Zhou S, Zong Y, Lu T, et al. Hematopoietic cells from mice that are deficient in both Bcrp1/Abcg2 and Mdr1a/1b develop normally but are sensitized to mitoxantrone. BioTechniques. 2003; 35(6): 1248-1252.

[234]

Voutsadakis IA. Expression and function of immune ligand-receptor pairs in NK cells and cancer stem cells: therapeutic implications. Cell Oncol. 2018; 41(2): 107-121.

[235]

Mandal A, Viswanathan C. Natural killer cells: in health and disease. Hematol Oncol Stem Cell Ther. 2015; 8(2): 47-55.

[236]

Zheng PP, Kros JM, Li J. Approved CAR T cell therapies: ice bucket challenges on glaring safety risks and long-term impacts. Drug Discov Today. 2018; 23(6): 1175-1182.

[237]

June CH, O’Connor RS, Kawalekar OU, et al. CAR T cell immunotherapy for human cancer. Science. 2018; 359(6382): 1361-1365.

[238]

Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: mechanisms, manifestations and management. Blood Rev. 2019; 34: 45-55.

[239]

Zhu X, Prasad S, Gaedicke S, et al. Patient-derived glioblastoma stem cells are killed by CD133-specific CAR T cells but induce the T cell aging marker CD57. Oncotarget. 2014; 6(1): 171-184.

[240]

Morgan RA, Johnson LA, Davis JL, et al. Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther. 2012; 23(10): 1043-1053.

[241]

O’Rourke DM, Nasrallah MP, Desai A, et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017; 9(399): eaaa0984.

[242]

Gammaitoni L, Giraudo L, Leuci V, et al. Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features. Clin Cancer Res. 2013; 19(16): 4347-4358.

[243]

Sangiolo D, Mesiano G, Gammaitoni L, et al. Cytokine-induced killer cells eradicate bone and soft-tissue sarcomas. Cancer Res. 2014; 74(1): 119-129.

[244]

Yang T, Zhang W, Wang L, et al. Co-culture of dendritic cells and cytokine-induced killer cells effectively suppresses liver cancer stem cell growth by inhibiting pathways in the immune system. BMC Cancer. 2018; 18: 984.

[245]

Mesiano G, Grignani G, Fiorino E, et al. Cytokine induced killer cells are effective against sarcoma cancer stem cells spared by chemotherapy and target therapy. Oncoimmunology. 2018; 7(11): e1465161.

[246]

Thankamony AP, Saxena K, Murali R, et al. Cancer stem cell plasticity—a deadly deal. Front Mol Biosci. 2020; 7: 79.

[247]

Patel AP, Tirosh I, Trombetta JJ, et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science. 2014; 344(6190): 1396-1401.

[248]

Dirkse A, Golebiewska A, Buder T, et al. Stem cell-associated heterogeneity in glioblastoma results from intrinsic tumor plasticity shaped by the microenvironment. Nat Commun. 2019; 10: 1787.

[249]

Caras IW. Two cancer stem cell-targeted therapies in clinical trials as viewed from the standpoint of the cancer stem cell model. Stem Cells Transl Med. 2020; 9(8): 821-826.

[250]

Locatelli MA, Aftimos P, Dees EC, et al. Phase I study of the gamma secretase inhibitor PF-03084014 in combination with docetaxel in patients with advanced triple-negative breast cancer. Oncotarget. 2016; 8(2): 2320-2328.

[251]

Debeb BG, Cohen EN, Boley K, et al. Pre-Clinical studies of notch signaling inhibitor RO4929097 in inflammatory breast cancer cells. Breast Cancer Res Treat. 2012; 134(2): 495-510.

[252]

Kartini D, Taher A, Panigoro SS, et al. Effect of melatonin supplementation in combination with neoadjuvant chemotherapy to miR-210 and CD44 expression and clinical response improvement in locally advanced oral squamous cell carcinoma: a randomized controlled trial. J Egypt Natl Canc Inst. 2020; 32(1): 12.

[253]

Rupp U, Schoendorf-Holland E, Eichbaum M, et al. Safety and pharmacokinetics of bivatuzumab mertansine in patients with CD44v6-positive metastatic breast cancer: final results of a phase I study. Anticancer Drugs. 2007; 18(4): 477-485.

[254]

Lu L, Tao H, Chang AE, et al. Cancer stem cell vaccine inhibits metastases of primary tumors and induces humoral immune responses against cancer stem cells. Oncoimmunology. 2015; 4(3): e990767.

[255]

Hu Y, Lu L, Xia Y, et al. Therapeutic efficacy of cancer stem cell vaccines in the adjuvant setting. Cancer Res. 2016; 76(16): 4661-4672.

[256]

Vik-Mo EO, Nyakas M, Mikkelsen BV, et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol Immunother. 2013; 62(9): 1499-1509.

[257]

Ngai LL, Ma CY, Maguire O, et al. Bimodal expression of potential drug target CLL-1 (CLEC12A) on CD34+ blasts of AML patients. Eur J Haematol. 2021; 107(3): 343-353.

[258]

Yanagisawa B, Perkins B, Karantanos T, et al. Expression of putative leukemia stem cell targets in genetically-defined acute myeloid leukemia subtypes. Leuk Res. 2020; 99: 106477.

[259]

Hanekamp D, Cloos J, Schuurhuis GJ. Leukemic stem cells: identification and clinical application. Int J Hematol. 2017; 105(5): 549-557.

[260]

Das N, Gupta R, Gupta SK, et al. A real-world perspective of CD123 expression in acute leukemia as promising biomarker to predict treatment outcome in B-ALL and AML. Clin Lymphoma Myeloma Leuk. 2020; 20(10): e673-e684.

[261]

Mardiros A, Dos Santos C, McDonald T, et al. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood. 2013; 122(18): 3138-3148.

[262]

Haferlach T, Bennett JM, Löffler H, et al. Acute myeloid leukemia with translocation (8;21). Cytomorphology, dysplasia and prognostic factors in 41 cases. Leuk Lymphoma. 1996; 23(3-4): 227-234

[263]

Tosic N, Petrovic I, Grujicic NK, et al. Prognostic significance of SOX2, SOX3, SOX11, SOX14 and SOX18 gene expression in adult de novo acute myeloid leukemia. Leuk Res. 2018; 67: 32-38.

[264]

Zhou Y, Xia L, Wang H, et al. Cancer stem cells in progression of colorectal cancer. Oncotarget. 2017; 9(70): 33403-33415.

[265]

Lugli A, Iezzi G, Hostettler I, et al. Prognostic impact of the expression of putative cancer stem cell markers CD133, CD166, CD44s, EpCAM, and ALDH1 in colorectal cancer. Br J Cancer. 2010; 103(3): 382-390.

[266]

Piao L, Feng Y, Yang Z, et al. LETM1 is a potential cancer stem-like cell marker and predicts poor prognosis in colorectal adenocarcinoma. Pathol Res Pract. 2019; 215(7): 152437.

[267]

Leng Z, Xia Q, Chen J, et al. Lgr5+CD44+EpCAM+ strictly defines cancer stem cells in human colorectal cancer. Cell Physiol Biochem. 2018; 46(2): 860-872.

[268]

Suman S, Hota SK, Misra P, et al. Immunohistochemical expression of the stem cell marker CD133 in colorectal carcinoma. Cureus; 15(7):e41242.

[269]

Glumac PM, LeBeau AM. The role of CD133 in cancer: a concise review. Clin Transl Med. 2018; 7: 18.

[270]

Fujino S, Miyoshi N. Oct4 gene expression in primary colorectal cancer promotes liver metastasis. Stem Cells Int. 2019; 2019: 7896524.

[271]

Sun B, Xu L, Bi W, et al. SALL4 oncogenic function in cancers: mechanisms and therapeutic relevance. Int J Mol Sci. 2022; 23(4): 2053.

[272]

Forghanifard MM, Moghbeli M, Raeisossadati R, et al. Role of SALL4 in the progression and metastasis of colorectal cancer. J Biomed Sci. 2013; 20(1): 6.

[273]

Zhu Y, Huang S, Chen S, et al. SOX2 promotes chemoresistance, cancer stem cells properties, and epithelial–mesenchymal transition by β-catenin and Beclin1/autophagy signaling in colorectal cancer. Cell Death Dis. 2021; 12(5): 1-16.

[274]

Ji J, Wang XW. Clinical implications of cancer stem cell biology in hepatocellular carcinoma. Semin Oncol. 2012; 39(4): 461-472. Published online August 2012.

[275]

Leung RWH, Lee TKW. Wnt/β-catenin signaling as a driver of stemness and metabolic reprogramming in hepatocellular carcinoma. Cancers (Basel). 2022; 14(21): 5468.

[276]

Han S, Guo J, Liu Y, et al. Knock out CD44 in reprogrammed liver cancer cell C3A increases CSCs stemness and promotes differentiation. Oncotarget. 2015; 6(42): 44452-44465.

[277]

Jeter CR, Yang T, Wang J, et al. NANOG in cancer stem cells and tumor development: an update and outstanding questions. Stem Cells. 2015; 33(8): 2381-2390.

[278]

Mancarella S, Serino G, Gigante I, et al. CD90 is regulated by notch1 and hallmarks a more aggressive intrahepatic cholangiocarcinoma phenotype. J Exp Clin Cancer Res. 2022; 41: 65.

[279]

Singh A, Srivastava AN, Akhtar S, et al. Correlation of CD133 and Oct-4 expression with clinicopathological and demographic parameters in oral squamous cell carcinoma patients. Natl J Maxillofac Surg. 2018; 9(1): 8-13.

[280]

Hsieh HL, Yu MC, Cheng LC, et al. Molecular mechanism of therapeutic approaches for human gastric cancer stem cells. World J Stem Cells. 2022; 14(1): 76-91.

[281]

Wang J, Qu J, Hou Q, et al. Strategies for the isolation and identification of gastric cancer stem cells. Stem Cells Int. 2024; 2024: 5553852.

[282]

Watanabe T, Okumura T, Hirano K, et al. Circulating tumor cells expressing cancer stem cell marker CD44 as a diagnostic biomarker in patients with gastric cancer. Oncol Lett. 2017; 13(1): 281-288.

[283]

Li L, Ni R, Zheng D, et al. Eradicating the tumor “seeds”: nanomedicines-based therapies against cancer stem cells. Daru. 2023; 31(1): 83-94.

[284]

Walcher L, Kistenmacher AK, Suo H, et al. Frontiers | cancer stem cells—origins and biomarkers: perspectives for targeted personalized therapies. Front Immunol. 2020; 11: 1280.

[285]

Sipos F, Műzes G. Cancer stem cell relationship with pro-tumoral inflammatory microenvironment. Biomedicines. 2023; 11(1): 189.

[286]

Jo JH, Park SB, Park S, et al. Novel gastric cancer stem cell-related marker LINGO2 is associated with cancer cell phenotype and patient outcome. Int J Mol Sci. 2019; 20(3): 555.

[287]

Pospieszna J, Dams-Kozlowska H, Udomsak W, et al. Unmasking the deceptive nature of cancer stem cells: the role of CD133 in revealing their secrets. Int J Mol Sci. 2023; 24(13): 10910.

[288]

El-Guindy DM, Wasfy RE, Abdel Ghafar MT, et al. Oct4 expression in gastric carcinoma: association with tumor proliferation, angiogenesis and survival. J Egypt Natl Canc Inst. 2019; 31(1): 3.

[289]

Carrasco-Garcia E, Santos JC, Garcia I, et al. Paradoxical role of SOX2 in gastric cancer. Am J Cancer Res. 2016; 6(4): 701-713.

[290]

Sin WC, Lim CL. Breast cancer stem cells—from origins to targeted therapy. Stem Cell Investig. 2017; 4: 96.

[291]

Zheng Q, Zhang M, Zhou F, et al. The breast cancer stem cells traits and drug resistance. Front Pharmacol. 2021; 11: 599965.

[292]

Yang X, Wang H, Jiao B. Mammary gland stem cells and their application in breast cancer. Oncotarget. 2016; 8(6): 10675-10691.

[293]

Oakes SR, Gallego-Ortega D, Ormandy CJ. The mammary cellular hierarchy and breast cancer. Cell Mol Life Sci. 2014; 71(22): 4301-4324.

[294]

Wang ML, Chiou SH, Wu CW. Targeting cancer stem cells: emerging role of Nanog transcription factor. Onco Targets Ther. 2013; 6: 1207-1220.

[295]

Saito M. Novel roles of nanog in cancer cells and their extracellular vesicles. Cells. 2022; 11(23): 3881.

[296]

Braune EB, Seshire A, Lendahl U. Notch and Wnt dysregulation and its relevance for breast cancer and tumor initiation. Biomedicines. 2018; 6(4): 101.

[297]

Wong NKY, Fuller M, Sung S, et al. Heterogeneity of breast cancer stem cells as evidenced with Notch-dependent and Notch-independent populations. Cancer Med. 2012; 1(2): 105-113.

[298]

Lo PK, Kanojia D, Liu X, et al. CD49f and CD61 identify Her2/neu-induced mammary tumor initiating cells that are potentially derived from luminal progenitors and maintained by the integrin-TGFβ signaling. Oncogene. 2012; 31(21): 2614-2626.

[299]

Sampayo RG, Bissell MJ. Cancer stem cells in breast and prostate: fact or fiction? Adv Cancer Res. 2019; 144: 315-341.

[300]

Zhang X, Powell K, Li L. Breast cancer stem cells: biomarkers, identification and isolation methods, regulating mechanisms, cellular origin, and beyond. Cancers (Basel). 2020; 12(12): 3765.

[301]

Lobba ARM, Carreira ACO, Cerqueira OLD, et al. High CD90 (THY-1) expression positively correlates with cell transformation and worse prognosis in basal-like breast cancer tumors. PLoS One. 2018; 13(6): e0199254.

[302]

Brenet M, Martínez S, Pérez-Nuñez R, et al. Thy-1 (CD90)-induced metastatic cancer cell migration and invasion are β3 integrin-dependent and involve a Ca2+/P2×7 receptor signaling axis. Front Cell Dev Biol. 2021; 8: 592442.

[303]

Utnal PA, A H, Pn S, Gn M. Expression of CD 133 in invasive ductal carcinoma of breast. Asian Pac J Cancer Prev. 2020; 21(10): 3055-3059.

[304]

Brugnoli F, Grassilli S, Al-Qassab Y, et al. CD133 in breast cancer cells: more than a stem cell marker. J Oncol. 2019; 2019: 7512632.

[305]

Carrà G, Cartellà A, Maffeo B, et al. Strategies for targeting chronic myeloid leukaemia stem cells. Blood Lymphat Cancer. 2019; 9: 45-52.

[306]

Soverini S, De Santis S, Monaldi C, et al. Targeting leukemic stem cells in chronic myeloid leukemia: is it worth the effort? Int J Mol Sci. 2021; 22(13): 7093.

[307]

Houshmand M, Simonetti G, Circosta P, et al. Chronic myeloid leukemia stem cells. Leukemia. 2019; 33(7): 1543-1556.

[308]

Patterson SD, Copland M. The bone marrow immune microenvironment in CML: treatment responses, treatment-free remission, and therapeutic vulnerabilities. Curr Hematol Malig Rep. 2023; 18(2): 19-32.

[309]

Cochrane CR, Szczepny A, Watkins DN, et al. Hedgehog signaling in the maintenance of cancer stem cells. Cancers (Basel). 2015; 7(3): 1554-1585.

[310]

Landberg N, von Palffy S, Askmyr M, et al. CD36 defines primitive chronic myeloid leukemia cells less responsive to imatinib but vulnerable to antibody-based therapeutic targeting. Haematologica. 2018; 103(3): 447-455.

[311]

Dybko J, Haus O, Jaźwiec B, et al. CD117 (c-kit) expression on CD34+ cells participates in the cytogenetic response to imatinib in patients with chronic myeloid leukemia in the first chronic phase. Acta Haematol. 2014; 132(2): 166-171.

[312]

El Achi H, Dupont E, Paul S, et al. CD123 as a biomarker in hematolymphoid malignancies: principles of detection and targeted therapies. Cancers (Basel). 2020; 12(11): 3087.

[313]

Masciale V, Grisendi G, Banchelli F, et al. CD44+/EPCAM+ cells detect a subpopulation of ALDHhigh cells in human non-small cell lung cancer: a chance for targeting cancer stem cells? Oncotarget. 2020; 11(17): 1545-1555.

[314]

Hochmair M, Rath B, Klameth L, et al. Effects of salinomycin and niclosamide on small cell lung cancer and small cell lung cancer circulating tumor cell lines. Invest New Drugs. 2020; 38(4): 946-955.

[315]

Zheng Y, Wang L, Yin L, et al. Lung cancer stem cell markers as therapeutic targets: an update on signaling pathways and therapies. Front Oncol. 2022; 12: 873994.

[316]

Mudra SE, Sadhukhan P, Ugurlu MT, et al. Therapeutic targeting of cancer stem cells in lung, head and neck, and bladder cancers. Cancers (Basel). 2021; 13(20): 5098.

[317]

TAN Y, CHEN B, XU W, et al. Clinicopathological significance of CD133 in lung cancer: a meta-analysis. Mol Clin Oncol. 2014; 2(1): 111-115.

[318]

El-Ashmawy NE, Salem ML, Abd El-Fattah EE, et al. Targeting CD166+ lung cancer stem cells: molecular study using murine dendritic cell vaccine. Toxicol Appl Pharmacol. 2021; 429: 115699.

[319]

Hussar P. Apoptosis regulators Bcl-2 and caspase-3. Encyclopedia. 2022; 2(4): 1624-1636.

[320]

Vigneswara V, Ahmed Z. The role of caspase-2 in regulating cell fate. Cells. 2020; 9(5): 1259.

[321]

Chen S, Zhu J, Wang F, et al. LncRNAs and their role in cancer stem cells. Oncotarget. 2017; 8(66): 110685-110692.

[322]

Bi J, Liu H, Dong W, et al. Circular RNA circ-ZKSCAN1 inhibits bladder cancer progression through miR-1178-3p/p21 axis and acts as a prognostic factor of recurrence. Mol Cancer. 2019; 18: 133.

[323]

Li P, Chen J, Zou J, et al. Circular RNA coiled-coil domain containing 66 regulates malignant development of papillary thyroid carcinoma by upregulating La ribonucleoprotein 1 via the sponge effect on miR-129-5p. Bioengineered. (2022); 13(3): 7181-7196.

[324]

Castro I, Carvajal P, Jara D, et al. Small RNA expression profiling reveals hsa-mir-181d-5p downregulation associated with TNF-α overexpression in Sjögren’s syndrome patients. Front Immunol. 2022; 13: 870094.

[325]

Tesolato SE, González-Gamo D, Barabash A, et al. Expression analysis of hsa-miR-181a-5p, hsa-miR-143-3p, hsa-miR-132-3p and hsa-miR-23a-3p as biomarkers in colorectal cancer—relationship to the body mass index. Cancers. 2023; 15(13): 3324.

[326]

Aguirre M, Escobar M, Forero Amézquita S, et al. Application of the Yamanaka transcription factors Oct4, Sox2, Klf4, and c-Myc from the laboratory to the clinic. Genes. 2023; 14(9): 1697.

[327]

Vishnoi K, Viswakarma N, Rana A, et al. Transcription Factors in cancer development and therapy. Cancers (Basel). 2020; 12(8): 2296.

[328]

He J, Fortunati E, Liu DX, et al. Pleiotropic roles of ABC transporters in breast cancer. Int J Mol Sci. 2021; 22(6): 3199.

[329]

Dei S, Braconi L, Romanelli MN, et al. Recent advances in the search of BCRP-and dual P-gp/BCRP-based multidrug resistance modulators. Cancer Drug Resist. 2019; 2(3): 710-743.

[330]

da Costa KM, Freire-de-Lima L, da Fonseca LM, et al. ABCB1 and ABCC1 function during TGF-β-induced epithelial-mesenchymal transition: relationship between multidrug resistance and tumor progression. Int J Mol Sci. 2023; 24(7): 6046.

[331]

Prasad S, Ramachandran S, Gupta N, et al. Cancer cells stemness: a doorstep to targeted therapy. Biochim Biophys Acta Mol Basis Dis. 2020; 1866(4): 165424.

[332]

Manni W, Min W. Signaling pathways in the regulation of cancer stem cells and associated targeted therapy. MedComm. 2022; 3(4): e176.

[333]

Espinosa-Sánchez A, Suárez-Martínez E, Sánchez-Díaz L, et al. Therapeutic targeting of signaling pathways related to cancer stemness. Front Oncol. 2020; 10: 1533.

[334]

Koguchi M, Nakahara Y, Ito H, et al. BMP4 induces asymmetric cell division in human glioma stem-like cells. Oncol Lett. 2020; 19(2): 1247-1254.

[335]

Hodgkinson T, Wignall F, Hoyland JA, et al. High BMPR2 expression leads to enhanced SMAD1/5/8 signalling and GDF6 responsiveness in human adipose-derived stem cells: implications for stem cell therapies for intervertebral disc degeneration. J Tissue Eng. 2020; 11: 2041731420919334.

[336]

Eskiizmir G. Tumor microenvironment in head and neck squamous cell carcinomas. Turk Arch Otorhinolaryngol. 2015; 53(3): 120-127.

[337]

Spinelli FM, Vitale DL, Demarchi G, et al. The immunological effect of hyaluronan in tumor angiogenesis. Clin Transl Immunol. 2015; 4(12): e52.

[338]

WANG C, XIE J, GUO J, et al. Evaluation of CD44 and CD133 as cancer stem cell markers for colorectal cancer. Oncol Rep. 2012; 28(4): 1301-1308.

[339]

Jaggupilli A, Elkord E. Significance of CD44 and CD24 as cancer stem cell markers: an enduring ambiguity. Clin Dev Immunol. 2012; 2012: 708036.

[340]

Jahangiri L. Cancer stem cell markers and properties across gastrointestinal cancers. Curr Tissue Microenviron Rep. 2023; 4(4): 77-89.

[341]

Werbowetski-Ogilvie TE. From sorting to sequencing in the molecular era: the evolution of the cancer stem cell model in medulloblastoma. FEBS J. 2022; 289(7): 1765-1778.

[342]

Anaparthy N, Ho YJ, Martelotto L, et al. Single-cell applications of next-generation sequencing. Cold Spring Harb Perspect Med. 2019; 9(10): a026898.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

164

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/