Abstract
As one of the common cancers that threaten human life, the recurrence and metastasis of colorectal cancer seriously affect the prognosis of patients. Although new drugs and comprehensive treatments have been adopted, the current treatment effect on this tumor, especially in advanced colorectal cancer, is still not satisfactory. More and more evidence shows that tumors are likely to be a stem cell disease. In recent years, the rise of cancer stem cell theory has provided a new way for cancer treatment. Studies have found that a small number of special cells in colorectal cancer tissues that induce tumorigenesis, proliferation, and promote tumor migration and metastasis, namely, colorectal cancer stem cells. Colorectal cancer stem cells are defined with a group of cell-surface markers, such as CD44, CD133, CD24, epithelial cell adhesion factor molecule, LGR5, and acetaldehyde dehydrogenase. They are highly tumorigenic, aggressive, and chemoresistant and thus are critical in the metastasis and recurrence of colorectal cancer. Therefore, targeting colorectal cancer stem cells may become an important research direction for the future cure of colorectal cancer.
Keywords: CRC, CSC, CCSCs, targeted therapy, drug resistance
Colorectal cancer (CRC) is one of the most common malignancies in the digestive system. The incidence of CRC in the United States between 2009 and 2013 was as high as 40.7 per 100 000, and the mortality rate from 2010 to 2014 was 14.8 per 100 000.1-3 It is the third largest malignant tumor in the United States and is also widely prevalent throughout the world. At present, the treatment plan for CRC is still mainly combined with surgery, radiotherapy, and chemotherapy.4 In recent years, with the development of endoscopy and various assistive technologies, although the mortality rate of patients has decreased, there is still no effective method to prevent the recurrence and metastasis of CRC.5 Hence, targeted therapy for the key pathogenic factors of CRC has become a major breakthrough, and the birth of cancer stem cell (CSC) theory has opened a new window for cancer treatment.6
Cells that have self-renewal capacity in tumors and that produce heterogeneous tumor cells are called cancer stem cells (CSCs).7 Cancer stem cells play an important role in the occurrence, development, recurrence, and metastasis of tumors.8 Traditional treatment methods can only reduce the volume of tumors and kill differentiated cells in tumors, whereas CSCs can resist radiotherapy and chemotherapy, eventually leading to tumor recurrence and metastasis.9 The research currently focuses on the observation of colorectal cancer stem cells (CCSCs) biological characteristics.10-12 With the development of stem cell theory, research on targeting CSC therapy has received increasing attention.
Discovery of CCSCs
The German pathologist Rudolf Virchow first proposed the theory that “every cell comes from another cell” in 1858.13 Makino et al 14 first proposed the CSC hypothesis in 1956 and believed that tumors were produced by CSCs. In the 1950s and 1960s, researchers found that a small number of cells in leukemia and myeloma cells can form clones in vitro and in vivo.15-17 This small number of cells is considered to be CSCs. In 2007, O’Brien et al 18 purified and expressed CD133-positive human CRC primary cells and transplanted them into the renal capsule of immunodeficient mice. These cells were found to maintain self-renewal and differentiation while forming tumors and therefore are called colorectal cancer stem cells (CCSCs).
Since these cells account for a small proportion of tumor tissues, identifying and understanding the characteristics of CCSCs in the past is difficult. However, with the development of molecular biology technology and related research, a variety of biomarkers have been available for the isolation of CCSCs. More cell-surface markers related to CCSCs have been discovered.4,19,20 It has been validated in different studies, which provides a basis for further study of the characteristics of CCSCs.
Cell-Surface Markers of CCSCs
Several cell-surface markers (solid tumors) have been used for the identification and isolation of CSCs and CCSCs (Table 1). In 2007, O’Brien et al 21 demonstrated that all CCSCs were CD133 expressing, while CD133-negative cells were unable to form tumors and functionally confirmed the rationality of CD133 as a surface marker for CCSCs. Haraguchi et al 22 found that CD133+CD44+ cells were tumorigenic, while CD133+CD44− and CD133−CD44− cells were not tumorigenic, and CD44 was further introduced into CD133 cells as a screening marker for CCSCs. Huang et al 27 found that acetaldehyde dehydrogenase (ALDH) was better as a CCSCs marker than CD133 and CD44. Dalerba et al 24 successfully isolated human CCSCs using CD44 and epithelial cell adhesion factor molecule (EpCAM) as a surface marker. Tumor cells of CD44+/EpCAMhigh were found to have the characteristics of CSCs and were verified in nonobese diabetic/severe combined immunodeficient mice, and the value of CD166 as a synergistic marker was also found.23 Pang et al 26 isolated CD26+ cells from CRC specimens and injected them into the cecal wall of mice and found that CD26+ cells were stronger in invasiveness and drug resistance than CD26− cells.
Table 1.
Name of Marker | Cancer | Function | References |
---|---|---|---|
EpCAM | Colon cancer, liver cancer | Chemoresistance, tumorigenesis, invasiveness, self-renewal | 18,21-23 |
CD133 | Colon cancer, lung cancer, gastric cancer, ovarian cancer, pancreatic cancer, liver cancer | Chemoresistance, self-renewal, tumorigenesis, invasiveness, proliferation, differentiation, angiogenesis, resistant to apoptosis | 18,21,22 |
CD166 | Colon cancer | proliferation, tumorigenesis, self-renewal, chemoresistance | 23 |
CD44 | Breast cancer, prostate cancer, colon cancer, lung cancer, gastric cancer, ovarian cancer, pancreatic cancer, liver cancer | Self-renewal, chemoresistance, metastasis, invasiveness, tumorigenesis | 18,21-24 |
CD90 | Gastric cancer, head and neck cancer, liver cancer | Metastasis, chemoresistance, tumorigenesis, self-renewal | 25 |
CD26 | Colon cancer | Self-renewal, tumorigenesis, invasiveness | 26 |
ALDH | Breast cancer, prostate cancer, colon cancer, lung cancer, ovarian cancer | Self-renewal, tumorigenesis, invasiveness, metastasis | 18,27 |
Aurora-A | Colon cancer | Proliferation, chemoresistance, EMT, invasiveness, metastasis | 28 |
CXCR4 | Lung cancer, colon cancer | Metastasis, chemoresistance, tumorigenesis, self-renewal | 29,30 |
Lamin A | Colon cancer | chemoresistance, tumorigenesis, self-renewal | 31 |
CD24 | Colon cancer, gastric cancer, prostate cancer | Metastasis, chemoresistance, tumorigenesis, self-renewal | 18 |
Abbreviations: ALDH, acetaldehyde dehydrogenase; EMT, epithelial–mesenchymal transition; EpCAM, epithelial cell adhesion factor molecule.
Studies have shown that the serine-threonine kinase Aurora-A is highly expressed in CCSCs, and downregulation of this gene leads to decreased tumorigenicity and metastasis of CCSCs, indicating that Aurora-A is used as a potential biomarker for CCSCs.28 Dessein et al 29 found that drug-resistant CRC cells have stronger invasiveness and distant metastasis and drug resistance. These cells express CXCR4 highly, and the macrophage migration inhibitor MIF, which is a key ligand of CXCR4, promotes cell malignancy. Behavior found that the CXCR4-MIF axis plays an important role in the regulation of CCSCs and may be a candidate for screening CCSCs.32 In addition, studies by Willis et al 31 showed that Lamin A has potential value for identifying CCSCs.
Currently reported surface markers of CCSCs have a certain scientific basis, but a highly specific surface marker or method has not been found to isolate and identify CCSCs. The markers of CCSCs newly screened in the future are likely to be important genes and products in the signaling pathway that plays an important role in regulating the cancer stemness.
Biological Characteristics of CCSCs
A study of ALDH+ CCSCs populations revealed that insulin-like growth factor 1 overexpresses the constitutively activated mutant of the silk/threonine protein kinase (AKT) in a β-catenin-dependent manner, resulting in increased stem cell numbers and tumor growth.33 In addition, ALDH+CD133+ CCSCs express high levels of phosphorylated signal transducer and activator of transcription 3 (STAT3).34 Curcumin and its analog GO-Y030 can inhibit this change, reduce the expression of target genes downstream of STAT3, and induce apoptosis of CCSCs to inhibit tumor formation.35 Other studies have shown that interleukin 6 (IL-6) can enhance the tumorigenic ability of cancer cell by inducing the expression of Notch homolog 1 and CD44 in CCSCs of HCT-116 and HT-29 CRC cell lines.36 Interferon γ and tumor necrosis factor α can also stimulate CCSCs in C26 mice and accelerate tumor growth.25 In addition, inhibition of hypoxia-inducible factor signaling pathway in CCSCs can effectively reduce the tumorigenic ability of CCSCs in an inflammatory environment in vivo.37 Leng et al 30 found that the overexpressed kruppel-like factor 4 in DLD-1 cell line can induce somatic cells to produce pluripotent stem cells and promote tumor formation in vitro and in vivo.
The Wnt signaling pathway is highly activated in CCSCs and inhibition of the reactive protein in this pathway will block signal transduction, thereby affecting the growth of CCSCs and achieving the purpose of treating CRC.38-40 High levels of Yap polypeptides are thought to be important factors in promoting the proliferation of CCSCs.41 High expression of Yap can cause the expansion of intestinal stem cell population and promote cancer cell proliferation, while the elimination of Yap can attenuate β-catenin and Notch signaling and inhibit cell proliferation and survival.42 In the HCT116 cell line, epidermal growth factor (EGF) was also found to be essential for maintaining stem cell proliferation.43-46 Inhibition of autophosphorylation of EGF receptor (EGFR1) and downstream signaling pathway protein AKT and extracellular signal-regulated kinase ERK1/2 decreases proliferation and induces apoptosis in CCSCs.47 Conditional inactivation of the telomerase reverse transcriptase (TERT) gene in colonic epithelial cells of newborn mice can induce overexpression of colonic crypts in mice, causing an increase in mucosal thickness and an increase in the number of goblet cells.48 This change in the differentiation status of selective crypts suggests that deficiency of TERT induces the proliferation and behavior of CCSCs to lead to tumorigenesis.
The study found that 5hmC is abundant in normal colon tissues of rats and humans, but significantly decreased in colon cancer tissues and CCSCs, indicating that 5hmC plays an important role in tissue differentiation.49-51 Another study found that activation of bone morphogenetic protein (BMP) signaling pathway in CD133+ stem cells can promote terminal differentiation and apoptosis of CCSCs.52 The CCSCs of immunodeficient mice express high expression of the tyrosine protein kinase receptor EphB2, which exhibits strong tumor production and long-term self-renewal potential, but gradually silences in cell differentiation.53
The high mobility group protein A1 (HMGA1) is a key transcription factor in the molecular mechanism of colon cancer metastasis.54-57 Previous study found that HMGAI-induced CCSCs express Twist 1 protein in transgenic mice, inhibit the expression of E-cadherin, and promote tumor metastasis and development; while knockout of HMGA1 blocks anchorage-independent growth and transplantation of tumor cells.58 The phenomenon of liver metastasis of colon cancer is also related to the involvement of transforming growth factor β (TGF-β).59-61 Transforming growth factor β promotes the growth of primary tumor and the occurrence of liver metastasis, which may be related to the activation of Smad molecules in CCSCs induced by TGF-β and the enhancement of cell invasion and transendothelial migration.62 Other studies have found that CCSCs of colon cancer cell lines HT29 and HCT-116 secrete high levels of plasminogen activator inhibitor 1 (PAI-1), which can significantly stimulate the migration of these 2 cell lines, while antibodies against PAI-1 can block this effect.63
Colorectal cancer stem cells are closely related to the resistance of colon cancer to chemotherapy drugs.64-66 Acetaldehyde dehydrogenase 1-positive HT29 and HT29-taxol cell lines lowly expressed miR-125a/b and highly expressed ALDHIA3 and Mcl1. The downregulation of miR-125a/b caused overexpression of ALDH1A3 and Mcl1 gene expression, increased cancer cell viability and stemness, and decreased cell apoptosis to play a key role in the resistance of chemotherapy drugs.67 Studies have found that BMP4 can attenuate the chemical resistance of CCSCs to treatment and enhance the antitumor effect of 5-fluorouracil (5-Fu) and oxaliplatin.68 The apoptotic protein inhibitor BIRC6 is an important regulator of the resistance of tumor stem cells to oxaliplatin and cisplatin in patients with CRC with liver metastasis, and the treatment with BIRC6 may help eliminate CCSCs and enhance the drug sensitivity of CRC cells to oxaliplatin and cisplatin.69 In another study, the expression of miR-451 in CRC cells with stem cell characteristics was downregulated in patients with CRC who did not respond to irinotecan-based first-line drug therapy, while the recovery of miR-451 reduced the expression of adenosine triphosphate-binding cassette B1.70 The expression of miR-451 leads to the sensitivity of cancer cells to irinotecan, suggesting that downregulation of miR-451 expression in CCSCs is an important factor in drug resistance for patients with CRC.
Targeting the Treatment of CRCSCs
The results of research on CSCs indicate that clinically used chemotherapy drugs and radiation therapy cannot effectively eliminate CSCs. Colorectal cancer stem cells are not sensitive to the treatment of 5-Fu and oxaliplatin, which may be the key factors leading to tumor metastasis and recurrence after chemotherapy.71 Therefore, research on targeting CCSCs has gradually become a research hotspot in the treatment of CRC in recent years (Table 2).
Table 2.
Trial | Target | Status |
---|---|---|
NCT02753127 | BBI-608 (STAT3 inhibitor) + FOLFIRI I metastatic colorectal cancer | Enrolling phase 3 |
NCT01553851 | GSK1120212 (MEK1/2 inhibitor) in oral cavity squamous cell cancer | Phase 2 complete |
NCT01190345 | Bevacizumab (anti-VEGF) + conventional therapy in breast cancer | Phase 2 |
NCT01579812 | Metformin (type 2 antidiabetic) + conventional therapy in ovarian, fallopian tube, and primary peritoneal cancer | Phase 2 |
NCT01624090 | Mithramycin (RNA synthesis inhibitor) in lung, esophageal, mesothelioma, breast cancer | Phase 2 |
NCT01861054 | Reparixin (inhibitor of CXCR1 and CXCR2) in breast cancer | Phase 2 |
NCT01195415 | Vismodegib (Hedgehog inhibitor) + Gemcitabine in advanced pancreatic cancer | Phase 2 |
NCT00645333 | MK-0752 (γ-secretase inhibitor) + Docetaxel in metastatic breast cancer | Phase 2 |
NCT01088815 | GDC-0449 (Hedgehog inhibitor) + conventional therapy in metastatic pancreatic cancer | Phase 2 |
NCT02370238 | Paclitaxel + Reparixin in metastatic triple negative breast cancer | Phase 2 |
NCT02279719 | BBI608 (STAT3 inhibitor) + Sorafenib or BBI503 (Nanog inhibitor) + Sorafenib in advanced hepatocellular carcinoma | Phase 2 |
NCT01951690 | VS-6063 (FAK inhibitor) in KRAS mutant non-small cell lung cancer | Phase 2 complete |
NCT02541370 | Cocktail CD133 CAR-T and CART-EGFR | Phase 2 |
NCT02049489 | ICT-121DC vaccine | Phase 2 |
NCT02915445 | EpCAM-targeted CAR-T cells | Phase 1 |
NCT03013712 | EpCAM-targeted CAR-T cells | Phase 2 |
NCT02216409 | Hu5F9-G4 (monoclonal antibody) | Phase 1 |
NCT02953782 | Combination Hu5F9-G4 and Cetuximab | Phase 2 |
NCT01567202 | Dendritic cell-based vaccine | Phase 2 |
Abbreviations: CSC, cancer stem cell; EGFR, epidermal growth factor receptor; EpCAM, epithelial cell adhesion factor molecule; STAT3, signal transducer and activator of transcription 3; VEGF, vascular endothelial growth factor.
Targeting the Cellular Signal Pathway of CCSCs
The signal pathways of CSCs mainly refer to their self-renewal pathways, including Wnt, Hedgehog, and Notch pathways.72-74 Almost all intestinal tumors have mutations in the Wnt pathway, involving changes in the tumor suppressor genes APC and Axin2, or the oncogene β-catenin.75 The activation of Wnt pathway in CRC has made it a target for molecular intervention. The use of small molecule inhibitors to inhibit β-catenin activity in the nucleus or to block β-catenin aggregation has been confirmed by in vitro experiments.68 In addition, treatment with β-catenin antisense primers reduced β-catenin expression and cancer cell stemness, and significant inhibition of tumor growth was observed in animal models of CRC.76
Hedgehog can activate the Hedgehog signaling pathway by binding to its receptor, and a highly expressed transcription factor Gli1 is found in CD133-positive CRC cells, which acts to activate Hedgehog, but not in CD133-negative cells.77 By knocking out the SMO gene and blocking the Hedgehog pathway in CRC cell lines, it is possible to inhibit the stemness of CCSCs in vitro and promote apoptosis of CRC cells.78 In addition, blocking the Hedgehog pathway of CCSCs can also inhibit tumor growth in vivo.79
The Notch pathway has been shown to be one of the important pathways for CCSCs to maintain stem cell characteristics.80 In an animal model of CRC, the polyclonal antibody 21M18 was used to block the role of DLL4 in the Notch pathway, and the proportion of tumor stem cells was reduced, and tumor growth was significantly inhibited.81 Further studies have shown that in the CRC metastasis model, although the treatment of imatinib can inhibit tumor growth, the proportion of ESA+CD44+CD166+ CCSCs is significantly increased.56 Combined application of imatinib and hDLL4 antibodies can significantly inhibit tumor growth and reduce the proportion of CCSCs, which is superior to antibody therapy alone.82
Promote the Differentiation of Stem Cells in CRC
Chemotherapy drugs for CRC are mainly directed to cells that are rapidly proliferating and isolated, while tumor stem cells are in a resting state in the cell cycle, which may be the main reason for their escape chemotherapy drugs.83-85 Therefore, it has become another major research direction to promote the further differentiation of CSCs to make them sensitive to traditional chemotherapy.86,87 Bone morphogenetic protein 4 can cause undifferentiated tumor cells to lose tumorigenicity, and tumors that lose tumorigenicity will undergo apoptosis, change cell proliferation cycle, or cell morphology changes.88
Study on the Microenvironment of CSCs
The stem cell microenvironment affects the ability of stem cells to proliferate, migrate, and invade. The results of previous study showed that both malignant tumors were congenital and acquired matrix changes.89 Treatment for the microenvironment of tumor cells may achieve significant results. CD133-positive CCSCs are capable of autocrine IL-4 and alter the microenvironment of CCSCs, making them insensitive to the chemotherapy drug 5-Fu or oxaliplatin.90 Conversely, when IL-4 antibodies are administered, tumor growth is significantly inhibited and sensitivity to chemotherapeutic drugs is increased. This phenomenon was also confirmed in animal models, and chemotherapy with IL-4 antibody treatment can significantly inhibit tumor growth.91
Colorectal Cancer Stem Cells and Tumor Immunotherapy
Tumor immunotherapy controls tumor recurrence and metastasis by improving the body’s antitumor immunity.92 After years of research, solid tumor immunotherapy has made some progress, but its clinical efficacy has not yet reached the goal of radical tumor treatment. The reason may be that the antigen applied by tumor immunotherapy is directed against tumor-differentiated cells, but the role of CSCs in tumor recurrence and metastasis is not significant.93 Therefore, further efforts to improve the efficacy of tumor immunotherapy should focus on treatment of CSCs. Studies have found that CSCs express high tumor-associated antigens and are used as antigen-induced DC vaccines to treat tumors, which can stimulate the action of antigen-specific T cells and prolong survival.94 Moreover, studies have shown that CSC antigens are superior to tumor cells in inducing antitumor immunity.95-97
If the tumor originates from the canceration of normal stem cells, treatment of CSCs is bound to result in damage to normal tissue stem cells.98-101 This contradictory state seems to indicate that treatment of CSCs can cause damage to the entire body.8,100,102 However, due to the different sensitivity of normal tissue stem cells and CSCs to treatment, the treatment of CSCs can kill tumor stem cells to the greatest extent.70,103-105 Just as the current chemotherapy drugs, although it causes serious side damage to the body, its role in controlling tumors is still positive.
Outlook
Traditional treatments for CRC include surgery, preoperative or postoperative adjuvant radiotherapy, or chemotherapy. After years of research on tumors, we have realized that tumors are a systemic chronic disease. Surgical treatment can remove tumors, and the adjuvant radiotherapy and chemotherapy can reduce the volume of the solid tumor to a certain extent and increase the survival rate of the patient. However, the theory of CSCs makes us more aware that chemoradiotherapy can only target differentiated tumor cells, but has little effect on CSCs that cause tumor recurrence and metastasis. In particular, recent studies have shown that changes in the tumor microenvironment can cause differentiated tumor cells to return to a state similar to stem cells. Tumor cells may have a phenomenon of returning to the ancestors, and the treatment of CSCs alone cannot cure the tumor. Therefore, future treatment methods should adopt a comprehensive treatment mode that minimizes tumor burden. By combining drugs targeting CCSCs with traditional chemotherapeutic drugs, both tumor metastasis and drug resistance are prevented, which will improve the efficacy of treating colon cancer at this stage. Colorectal cancer stem cells targeted inhibitors as an emerging treatment for colon cancer; although there are still many unclear mechanisms yet to be discovered, it is expected that these drugs will prevent colon cancer recurrence and treat colon cancer metastasis in the future.
Abbreviations
- 5-Fu
5-fluorouracil
- AKT
silk/threonine protein kinase
- ALDH
acetaldehyde dehydrogenase
- BMP
bone morphogenetic protein
- CCSCs
colorectal cancer stem cells
- CRC
colorectal cancer
- CSC
cancer stem cell
- EGF
epidermal growth factor
- EGFR1
EGF receptor 1
- EpCAM
epithelial cell adhesion factor molecule
- HMGA1
high mobility group protein A1
- IL-6
interleukin 6
- PAI-1
plasminogen activator inhibitor 1
- STAT3
signal transducer and activator of transcription 3
- TERT
telomerase reverse transcriptase
- TGF-β
transforming growth factor β
Footnotes
Authors’ Note: Yu-Shui Ma and Wen Li contributed equally to this work. QLL and DF designed and supervised research. All authors interpreted the data and contributed to the final version of the manuscript. Our study did not require an ethical board approval because it did not contain human or animal trials.
Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This study was supported partly by grants from the National Natural Science Foundation of China (81972214, 81772932, 81472202, 81201535, 81302065, 81671716, 81301993, 81372175, and 81472209), The Fundamental Research Funds for the Central Universities (22120170212 and 22120170117), Shanghai Natural Science Foundation (12ZR1436000), and Shanghai Municipal Commission of Health and Family Planning (201540228).
ORCID iD: Da Fu https://orcid.org/0000-0002-0878-2575
References
- 1. Laissue P. The forkhead-box family of transcription factors: key molecular players in colorectal cancer pathogenesis. Mol Cancer. 2019;18(1):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Cristóbal I, Sanz-Alvarez M, Torrejón B, et al. Potential therapeutic impact of miR-145 deregulation in colorectal cancer. Mol Ther. 2018;26(6):1399–1400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. O’Leary MP, Warner SG, Kim SI, et al. A novel oncolytic chimeric orthopoxvirus encoding luciferase enables real-time view of colorectal cancer cell infection. Mol Ther Oncolytics. 2018;9:13–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Zhu Y, Wang C, Becker SA, et al. miR-145 antagonizes SNAI1-mediated stemness and radiation resistance in colorectal cancer. Mol Ther. 2018;26(3):744–754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Ibarrola-Villava M, Cervantes A, Bardelli A. Preclinical models for precision oncology. Biochim Biophys Acta Rev Cancer. 2018;1870(2):239–246. [DOI] [PubMed] [Google Scholar]
- 6. Xu H, Wang C, Song H, Xu Y, Ji G. RNA-Seq profiling of circular RNAs in human colorectal cancer liver metastasis and the potential biomarkers. Mol Cancer. 2019;18(1):8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Su R, Cao S, Ma J, et al. Knockdown of SOX2OT inhibits the malignant biological behaviors of glioblastoma stem cells via up-regulating the expression of miR-194-5p and miR-12. Mol Cancer. 2018;16(1):171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bai X, Ni J, Beretov J, Graham P, Li Y. Cancer stem cell in breast cancer therapeutic resistance. Cancer Treat Rev. 2018;69:152–163. [DOI] [PubMed] [Google Scholar]
- 9. Bhartiya D, Patel H, Ganguly R, et al. Novel insights into adult and cancer stem cell biology. Stem Cells Dev. 2018;27(22):1527–1539. [DOI] [PubMed] [Google Scholar]
- 10. Toiyama Y, Okugawa Y, Fleshman J, Richard Boland C, Goel A. MicroRNAs as potential liquid biopsy biomarkers in colorectal cancer: a systematic review. Biochim Biophys Acta Rev Cancer. 2018;1870(2):274–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Herrera M, Llorens C, Rodríguez M, et al. Differential distribution and enrichment of non-coding RNAs in exosomes from normal and cancer-associated fibroblasts in colorectal cancer. Mol Cancer. 2018;17(1):114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Li J, Song P, Jiang T, et al. Heat shock factor 1 epigenetically stimulates glutaminase-1-dependent mTOR activation to promote colorectal carcinogenesis. Mol Ther. 2018;26(7):1828–1839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Ribatti D. An historical note on the cell theory. Exp Cell Res. 2018;364(1):1–4. [DOI] [PubMed] [Google Scholar]
- 14. Makino S. Further evidence favoring the concept of the stem cell in ascites tumors of rats. Ann N Y Acad Sci. 1956;63(5):818–830. [DOI] [PubMed] [Google Scholar]
- 15. Tzelepis K, De Braekeleer E, Aspris D, et al. SRPK1 maintains acute myeloid leukemia through effects on isoform usage of epigenetic regulators including BRD4. Nat Commun. 2018;9(1):5378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Escobar G, Barbarossa L, Barbiera G, et al. Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens. Nat Commun. 2018;9(1):2896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445(7123):106–110. [DOI] [PubMed] [Google Scholar]
- 19. Kryeziu K, Bruun J, Guren TK, Sveen A, Lothe RA. Combination therapies with HSP90 inhibitors against colorectal cancer. Biochim Biophys Acta Rev Cancer. 2019;1871(2):240–247. [DOI] [PubMed] [Google Scholar]
- 20. Chen E, Yang F, He H, et al. Alteration of tumor suppressor BMP5 in sporadic colorectal cancer: a genomic and transcriptomic profiling based study. Mol Cancer. 2018;17(1):176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Chen X, Guan H, Liu XD, et al. p53 positively regulates the expression of cancer stem cell marker CD133 in HCT116 colon cancer cells. Oncol Lett. 2018;16(1):431–438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Haraguchi N, Ohkuma M, Sakashita H, et al. CD133+CD44+ population efficiently enriches colon cancer initiating cells. Ann Surg Oncol. 2008;15(10): 2927–2933. [DOI] [PubMed] [Google Scholar]
- 23. Liu D, Sun J, Zhu J, Zhou H, Zhang X, Zhang Y. Expression and clinical significance of colorectal cancer stem cell marker EpCAMhigh/CD44+ in colorectal cancer. Oncol Lett. 2014;7(5):1544–1548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Dalerba P, Dylla SJ, Park IK, et al. Phenotypic characterization of human colorectal cancer stem cells. Proc Natl Acad Sci U S A. 200;104(24):10158–10163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Bekaii-Saab T, El-Rayes B. Identifying and targeting cancer stem cells in the treatment of gastric cancer. Cancer. 2017;123:1303–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Pang R, Law WL, Chu AC, et al. A subpopulation of CD26+ cancer stem cells with metastatic capacity in human colorectal cancer. Cell Stem Cell. 2010;6(6):603–615. [DOI] [PubMed] [Google Scholar]
- 27. Huang EH, Hynes MJ, Zhang T, et al. Aldehyde dehydrogenase 1 is a marker for normal and malignant human colonic stem cells (SC) and tracks SC overpopulation during colon tumorigenesis. Cancer Res. 2009;69(8):3382–3389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Cammareri P, Scopelliti A, Todaro M, et al. Aurora-A is essential for the tumorigenic capacity and chemoresistance of colorectal cancer stem cells. Cancer Res. 2010;70(11):4655–4665. [DOI] [PubMed] [Google Scholar]
- 29. Dessein AF, Stechly L, Jonckheere N, et al. Autocrine induction of invasive and metastatic phenotypes by the MIF-CXCR4 axis in drug-resistant human colon cancer cells. Cancer Res. 2010;70(11):4644–4654. [DOI] [PubMed] [Google Scholar]
- 30. Sun W, Fu S. Role of cancer-associated fibroblasts in tumor structure, composition and the microenvironment in ovarian cancer. Oncol Lett. 2019;18(3):2173–2178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Willis ND, Wilson RG, Hutchison CJ. Lamin A: a putative colonic epithelial stem cell biomarker which identifies colorectal tumours with a more aggressive phenotype. Biochem Soc Trans. 2008;36(pt 6):1350–1353. [DOI] [PubMed] [Google Scholar]
- 32. Lue H, Dewor M, Leng L, Bucala R, Bernhagen J. Activation of the JNK signalling pathway by macrophage migration inhibitory factor (MIF) and dependence on CXCR4 and CD. Cell Signal. 2011;23(1):135–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Bhullar KS, Lagarón NO, McGowan EM, et al. Kinase-targeted cancer therapies: progress, challenges and future directions. Mol Cancer. 2018;17(1):48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Liu Y, Lv J, Liu J, et al. STAT3/p53 pathway activation disrupts IFN-β-induced dormancy in tumor-repopulating cells. J Clin Invest. 2018;128(3):1057–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Lin L, Liu Y, Li H, et al. Targeting colon cancer stem cells using a new curcumin analogue, GO-Y030. Br J Cancer. 2011;105(2):212–220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Aleksakhina SN, Kashyap A, Imyanitov EN. Mechanisms of acquired tumor drug resistance. Biochim Biophys Acta Rev Cancer. 2019;1872(2):188310. [DOI] [PubMed] [Google Scholar]
- 37. Jin F, Yang R, Wei Y, et al. HIF-1α-induced miR-23a∼27a∼24 cluster promotes colorectal cancer progression via reprogramming metabolism. Cancer Lett. 2019;440-441:211–222. [DOI] [PubMed] [Google Scholar]
- 38. Hou Z, Guo K, Sun X, et al. TRIB2 functions as novel oncogene in colorectal cancer by blocking cellular senescence through AP4/p21 signaling. Mol Cancer. 2018;17(1):172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Yue B, Liu C, Sun H, et al. A positive feed-forward loop between LncRNA-CYTOR and Wnt/β-catenin signaling promotes metastasis of colon cancer. Mol Ther. 2018;26(5):1287–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Shenoy AK, Fisher RC, Butterworth EA, et al. Transition from colitis to cancer: high Wnt activity sustains the tumor-initiating potential of colon cancer stem cell precursors. Cancer Res. 2012;72(19):5091–5100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Pan Y, Tong JHM, Lung RWM, et al. RASAL2 promotes tumor progression through LATS2/YAP1 axis of hippo signaling pathway in colorectal cancer. Mol Cancer. 2018;17(1):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Yu J, Han Z, Sun Z, Wang Y, Zheng M, Song C. LncRNA SLCO4A1-AS1 facilitates growth and metastasis of colorectal cancer through β-catenin-dependent Wnt pathway. J Exp Clin Cancer Res. 2018;37(1):222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Fassunke J, Müller F, Keul M, et al. Overcoming EGFRG724S-mediated osimertinib resistance through unique binding characteristics of second-generation EGFR inhibitors. Nat Commun. 2018;9(1):4655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Tzchori I, Falah M, Shteynberg D, et al. Improved patency of ePTFE grafts as a hemodialysis access site by seeding autologous endothelial cells expressing Fibulin-5 and VEGF. Mol Ther. 2018;26(7):1660–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Tsai YC, Zeng T, Abou-Kheir W, et al. Disruption of ETV6 leads to TWIST1-dependent progression and resistance to epidermal growth factor receptor tyrosine kinase inhibitors in prostate cancer. Mol Cancer. 2018;17(1):42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Vitiello PP, Cardone C, Martini G, et al. Receptor tyrosine kinase-dependent PI3 K activation is an escape mechanism to vertical suppression of the EGFR/RAS/MAPK pathway in KRAS-mutated human colorectal cancer cell lines. J Exp Clin Cancer Res. 2019;38(1):41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Sun J, Luo Q, Liu L, Song G. Low-level shear stress promotes migration of liver cancer stem cells via the FAK-ERK1/2 signalling pathway. Cancer Lett. 2018;427:1–8. [DOI] [PubMed] [Google Scholar]
- 48. Khattar E, Kumar P, Liu CY, et al. Telomerase reverse transcriptase promotes cancer cell proliferation by augmenting tRNA expression. J Clin Invest. 2016;126(10):4045–4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Xu M, Chen X, Lin K, et al. The long noncoding RNA SNHG1 regulates colorectal cancer cell growth through interactions with EZH2 and miR-154-5p. Mol Cancer. 2018;17(1):141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Belhadj S, Moutinho C, Mur P, et al. Germline variation in O6-methylguanine-DNA methyltransferase (MGMT) as cause of hereditary colorectal cancer. Cancer Lett. 2019;447:86–92. [DOI] [PubMed] [Google Scholar]
- 51. Katoh M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity. Int J Oncol. 2018;51(5):1357–1369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Luo Y, Chen JJ, Lv Q, et al. Long non-coding RNA NEAT1 promotes colorectal cancer progression by competitively binding miR-34a with SIRT1 and enhancing the Wnt/β-catenin signaling pathway. Cancer Lett. 2019;440-441:11–22. [DOI] [PubMed] [Google Scholar]
- 53. De Robertis M, Mazza T, Fusilli C, et al. EphB2 stem-related and EphA2 progression-related miRNA-based networks in progressive stages of CRC evolution: clinical significance and potential miRNA drivers. Mol Cancer. 2018;17(1):169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Yagiz K, Rodriguez-Aguirre ME, Lopez Espinoza F, et al. A retroviral replicating vector encoding cytosine deaminase and 5-FC induces immune memory in metastatic colorectal cancer models. Mol Ther Oncolytics. 2017;8:14–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Yamano T, Kubo S, Fukumoto M, et al. Whole cell vaccination using immunogenic cell death by an oncolytic adenovirus is effective against a colorectal cancer model. Mol Ther Oncolytics. 2016;3:16031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Wei F, Zhang T, Deng SC, et al. PD-L1 promotes colorectal cancer stem cell expansion by activating HMGA1-dependent signaling pathways. Cancer Lett. 2019;450:1–13 [DOI] [PubMed] [Google Scholar]
- 57. Wang Y, Hu L, Zheng Y, Guo L. HMGA1 in cancer: cancer classification by location. J Cell Mol Med. 2019;23(4):2293–2302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Parry AJ, Hoare M, Bihary D, et al. NOTCH-mediated non-cell autonomous regulation of chromatin structure during senescence. Nat Commun. 2018;9(1):1840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Ma YS, Huang T, Zhong XM, et al. Proteogenomic characterization and comprehensive integrative genomic analysis of human colorectal cancer liver metastasis. Mol Cancer. 2018;17(1):139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Oshima G, Guo N, He C, et al. In vivo delivery and therapeutic effects of a microRNA on colorectal liver metastases. Mol Ther. 2018;25(7):1588–1595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Gao T, Hu Q, Hu X, et al. Novel selective TOPK inhibitor SKLB-C05 inhibits colorectal carcinoma growth and metastasis. Cancer Lett. 2019;445:11–23. [DOI] [PubMed] [Google Scholar]
- 62. Grassi A, Perilli L, Albertoni L, et al. A coordinate deregulation of microRNAs expressed in mucosa adjacent to tumor predicts relapse after resection in localized colon cancer. Mol Cancer. 2018;17(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Peng Y, Kajiyama H, Yuan H, et al. PAI-1 secreted from metastatic ovarian cancer cells triggers the tumor-promoting role of the mesothelium in a feedback loop to accelerate peritoneal dissemination. Cancer Lett. 2019;442:181–192. [DOI] [PubMed] [Google Scholar]
- 64. Liu T, Han Z, Li H, Zhu Y, Sun Z, Zhu A. LncRNA DLEU1 contributes to colorectal cancer progression via activation of KPNA. Mol Cancer. 2018;17(1):118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Zhang J, Ren P, Xu D, et al. Human UTP14a promotes colorectal cancer progression by forming a positive regulation loop with c-Myc. Cancer Lett. 2019;440-441:106–115. [DOI] [PubMed] [Google Scholar]
- 66. Giordano G, Parcesepe P, D’Andrea MR, et al. JAK/Stat5-mediated subtype-specific lymphocyte antigen 6 complex, locus G6D expression drives mismatch repair proficient colorectal cancer. J Exp Clin Cancer Res. 2019;38(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Uppada SB, Gowrikumar S, Ahmad R, et al. MASTL induces colon cancer progression and chemoresistance by promoting Wnt/β-catenin signaling. Mol Cancer. 2018;17(1):111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Li T, Jian X, He H, et al. MiR-452 promotes an aggressive colorectal cancer phenotype by regulating a Wnt/β-catenin positive feedback loop. J Exp Clin Cancer Res. 2018;37(1):238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Zhang YK, Wang YJ, Lei ZN, et al. Regorafenib antagonizes BCRP-mediated multidrug resistance in colon cancer. Cancer Lett. 2019;442:104–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Deng S, Deng Q, Zhang Y, et al. Non-platelet-derived CXCL4 differentially regulates cytotoxic and regulatory T cells through CXCR3 to suppress the immune response to colon cancer. Cancer Lett. 2019;443:1–12. [DOI] [PubMed] [Google Scholar]
- 71. Jeong S, Yun HK, Jeong YA, et al. Cannabidiol-induced apoptosis is mediated by activation of Noxa in human colorectal cancer cells. Cancer Lett. 2019;447:12–23. [DOI] [PubMed] [Google Scholar]
- 72. Tsai KW, Lo YH, Liu H, et al. Linc00659, a long noncoding RNA, acts as novel oncogene in regulating cancer cell growth in colorectal cancer. Mol Cancer. 2018;17(1):72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Duan S, Huang W, Liu X, et al. IMPDH2 promotes colorectal cancer progression through activation of the PI3K/AKT/mTOR and PI3K/AKT/FOXO1 signaling pathways. J Exp Clin Cancer Res. 2018;37(1):304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Ma YS, Yu F, Zhong XM, et al. miR-30 family reduction maintains self-renewal and promotes tumorigenesis in NSCLC-initiating cells by targeting oncogene TM4SF. Mol Ther. 2018;26(12):2751–2765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Murillo-Garzón V, Gorroño-Etxebarria I, Åkerfelt M, et al. Frizzled-8 integrates Wnt-11 and transforming growth factor-β signaling in prostate cancer. Nat Commun. 2018;9(1):1747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Krishnamurthy N, Kurzrock R. Targeting the Wnt/beta-catenin pathway in cancer: update on effectors and inhibitors. Cancer Treat Rev. 2018;62:50–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Feng HC, Lin JY, Hsu SH, et al. Low folate metabolic stress reprograms DNA methylation-activated sonic hedgehog signaling to mediate cancer stem cell-like signatures and invasive tumour stage-specific malignancy of human colorectal cancers. Int J Cancer. 2017;141(12):2537–2550. [DOI] [PubMed] [Google Scholar]
- 78. Chang SC, Ding JL. Ubiquitination and SUMOylation in the chronic inflammatory tumor microenvironment. Biochim Biophys Acta Rev Cancer. 2018;1870(2):165–175. [DOI] [PubMed] [Google Scholar]
- 79. Ylä-Herttuala S. The pharmacology of gene therapy. Mol Ther. 2017;25(8):1731–1732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Wang D, Hu G, Du Y, et al. Aberrant activation of hedgehog signaling promotes cell proliferation via the transcriptional activation of forkhead box M1 in colorectal cancer cells. J Exp Clin Cancer Res. 2017;36(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Smith DC, Eisenberg PD, Manikhas G, et al. A phase I dose escalation and expansion study of the anticancer stem cell agent demcizumab (anti-DLL4) in patients with previously treated solid tumors. Clin Cancer Res. 2014;20(24):6295–6303. [DOI] [PubMed] [Google Scholar]
- 82. Imperial R, Ahmed Z, Toor OM, et al. Comparative proteogenomic analysis of right-sided colon cancer, left-sided colon cancer and rectal cancer reveals distinct mutational profiles. Mol Cancer. 2018;17(1):177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Fletcher R, Wang YJ, Schoen RE, Finn OJ, Yu J, Zhang L. Colorectal cancer prevention: immune modulation taking the stage. Biochim Biophys Acta Rev Cancer. 2018;1869(2):138–148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Wang X, Yu H, Sun W, et al. The long non-coding RNA CYTOR drives colorectal cancer progression by interacting with NCL and Sam68. Mol Cancer. 2018;17(1):110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Hatano Y, Semi K, Hashimoto K, et al. Reducing DNA methylation suppresses colon carcinogenesis by inducing tumor cell differentiation. Carcinogenesis. 2015;36(7):719–729. [DOI] [PubMed] [Google Scholar]
- 86. Ordóñez-Morán P, Dafflon C, Imajo M, Nishida E, Huelsken J. HOXA5 counteracts stem cell traits by inhibiting Wnt signaling in colorectal cancer. Cancer Cell. 2015;28(6):815–829. [DOI] [PubMed] [Google Scholar]
- 87. Wang Y, Wu M, Lei Z, et al. Dysregulation of miR-6868-5p/FOXM1 circuit contributes to colorectal cancer angiogenesis. J Exp Clin Cancer Res. 2018;37(1):292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Hu JL, Wang W, Lan XL, et al. CAFs secreted exosomes promote metastasis and chemotherapy resistance by enhancing cell stemness and epithelial-mesenchymal transition in colorectal cancer. Mol Cancer. 2019;18(1):91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Wu G, Ma Z, Cheng Y, et al. Targeting Gas6/TAM in cancer cells and tumor microenvironment. Mol Cancer. 2018;17(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Zhang B, Fu D, Xu Q, Cong X, Wu C, Zhong X, The senescence-associated secretory phenotype is potentiated by feedforward regulatory mechanisms involving Zscan4 and TAK. Nat Commun. 2018;9(1):1723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Dong R, Gong Y, Meng W, et al. The involvement of M2 macrophage polarization inhibition in fenretinide-mediated chemopreventive effects on colon cancer. Cancer Lett. 2017;388:43–53. [DOI] [PubMed] [Google Scholar]
- 92. Rezvani K, Rouce R, Liu E, Shpall E. Engineering natural killer cells for cancer immunotherapy. Mol Ther. 2017;25(8):1769–1781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Zhang Y, Guo L, Li Y, et al. MicroRNA-494 promotes cancer progression and targets adenomatous polyposis coli in colorectal cancer. Mol Cancer. 2018;17(1):1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Cullis PR, Hope MJ. Lipid nanoparticle systems for enabling gene therapies. Mol Ther. 2017;25(7):1467–1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Abou-El-Enein M, Grainger DW, Kili S. Registry contributions to strengthen cell and gene therapeutic evidence. Mol Ther. 2018;26(5):1172–1176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Durham NM, Mulgrew K, McGlinchey K, et al. Oncolytic VSV primes differential responses to immuno-oncology therapy. Mol Ther. 2017;25(8):1917–1932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Deng S, Zhang G, Kuai J, et al. Lentinan inhibits tumor angiogenesis via interferon γ and in a T cell independent manner. J Exp Clin Cancer Res. 2018;37(1):260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Matsuyama T, Ishikawa T, Takahashi N, et al. Transcriptomic expression profiling identifies ITGBL, an epithelial to mesenchymal transition (EMT)-associated gene, is a promising recurrence prediction biomarker in colorectal cancer. Mol Cancer. 2019;18(1):19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Mur P, De Voer RM, Olivera-Salguero R, Rodríguez-Perales S, Pons T, Setién F. Germline mutations in the spindle assembly checkpoint genes BUB1 and BUB3 are infrequent in familial colorectal cancer and polyposis. Mol Cancer. 2018;17(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Nimmakayala RK, Batra SK, Ponnusamy MP. Unraveling the journey of cancer stem cells from origin to metastasis. Biochim Biophys Acta Rev Cancer. 2019;1871(1):50–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Ma YS, Lv ZW, Yu F, et al. MiRNA-302a/d inhibits the self-renewal capability and cell cycle entry of liver cancer stem cells by targeting the E2F7/AKT axis. J Exp Clin Cancer Res. 2018;37(1):252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Yu F, Liu JB, Wu ZJ, et al. Tumor suppressive microRNA-124a inhibits stemness and enhances gefitinib sensitivity of non-small cell lung cancer cells by targeting ubiquitin-specific protease 1. Cancer Lett. 2018;427:74–84. [DOI] [PubMed] [Google Scholar]
- 103. Ma J, Kala S, Yung S, et al. Blocking stemness and metastatic properties of ovarian cancer cells by targeting p70S6K with dendrimer nanovector-based siRNA delivery. Mol Ther. 2018;26(1):70–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Zhang C, Wang Z, Yang Z, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA+ metastatic colorectal cancers. Mol Ther. 2017;25(5):1248–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Hombach AA, Rappl G, Abken H. Blocking CD30 on T cells by a dual specific CAR for CD30 and colon cancer antigens improves the CAR T cell response against CD30− tumors. Mol Ther. 2019;27(10):1825–1835. [DOI] [PMC free article] [PubMed] [Google Scholar]