Metastatic components in colorectal cancer
Marjan Hajimoradi Javarsiani1, Shagayegh Haghjooy Javanmard2, Francesca Colonna3
1 Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran 2 Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran 3 Department of General Pathology, Cattolica del Sacro Cuore Largo Francesco University, Rome, Italy
Date of Submission | 16-Jan-2019 |
Date of Decision | 05-Mar-2019 |
Date of Acceptance | 29-May-2019 |
Date of Web Publication | 28-Aug-2019 |
Correspondence Address: Dr. Marjan Hajimoradi Javarsiani Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan Iran
 Source of Support: None, Conflict of Interest: None  | Check |
DOI: 10.4103/jrms.JRMS_957_18
Recent experiments have shown that cells with different genetic mutations can give rise to cancer transformation, both in vitro and in vivo, supported by the crosstalk between cancer cells and stroma. The stroma and the complex set of involved cells make up the tumor microenvironment that supports the engraftment of metastatic cells. In fact, environmental factors support colorectal cancer arise by formation and maintenance of cancer stem cells (CSCs). In this review, we discuss interactions between CSCs and their microenvironment that can provide better therapeutic opportunities in the metastatic cancer.
Keywords: Cancer stem cells, colorectal cancer, metastasis, tumor microenvironment
How to cite this article: Javarsiani MH, Javanmard SH, Colonna F. Metastatic components in colorectal cancer. J Res Med Sci 2019;24:75 |
Introduction | |  |
One of the most frequent types of cancer worldwide is colorectal cancer (CRC).[1] The intestine epithelium has a higher self-renewal rate than any other tissues in the body.[2] Classically, CRC pathogenesis has been characterized as the stepwise progression of cancerous lesions from potentially malignant precursors, predominantly tubular adenomas.[3]
Surgery, CRC therapeutic gold standard, is effective if tumor cells have not spread to distant organs. Colorectal-cancer stem cells (CR-CSCs) can metastasize in liver and lungs. Around the 30% of patients have a too late diagnosis of CRC.[4],[5] In fact, in 20%–25% of patients with colon cancer, and in 18% of patients with rectal cancer, metastases are present at the time of the first diagnosis.[6],[7] The most common metastasis localization in CRC patients is liver, but 2.1% of these patients show lung metastases,[8] with frequency about three times higher for patients with rectal cancer than for patients with colon cancer. In order to control and eradicate cancer, we need to focus not only on malignant cancer cells but also on the benign stromal cells useful indeed to cancer cells.
Metastasis, spreading of cancer cells to other organs, is an ability to regenerate a tumor at a distant site.[9] It is now recognized that the neogenesis of the tumor in a foreign organ is tightly bound to the gain of stem-like phenotype of cancer cells.[10] This feature critically depends on their interaction with microenvironment to migrate, survive in circulation, take root, and proliferate in a new organ [Figure 1].[9]
Colorectal Cancer Pathogenesis | |  |
The chromosomal instability pathway
The chromosomal instability pathway is known as the adenoma-carcinoma sequence, that establish a predictable progression of genetic mutations that involve histologic consequences and changes.[11],[12] Colon cancer pathogenesis is carried out by mutations called “drivers,” necessary but not sufficient, and mutations called “passengers” contributing to tumor progression and that are witness to the multiple intra- and inter-patient heterogeneity. The genomic changes include inactivation of at least three tumor suppression genes (loss of adenomatous polyposis coli [APC], p53 and loss of heterozygosity for chromosome 18 [18q LOH]) and activation of proto-oncogenes (K-Ras).
Driver's mutations are:
- Inactivation of tumor suppressor APC (chromosome region 5q21, germline in FAP syndrome, or somatic in sporadic cases)
- Activation of proto-oncogene K-Ras
- Loss of p53
- 18q LOH, consequently loss of Deleted in colon carcinoma (DCC) and of some SMAD, transforming growth factor beta pathway signal transducer proteins.
Recently, mutations in the genes of tumor growth factor beta receptor (TGFβR) and PIK3CA has been also described for the adenoma-carcinoma sequence model.[11],[13]
Adenomatous polyposis coli and Wnt signaling pathway
Familial adenomatous polyposis (FAP) and others familial syndromes make APC-associated polyposis conditions. FAP is a CRC-predisposition syndrome that leads to the development of very numerous precancerous lesions or polyps in the last intestine tract. It represents <1% of all cases of colon cancer. In this disease, it is absolutely necessary to undergo a prophylactic colectomy to avoid the tumor, which occurs in 100% of cases.
Mutations in APC/Wnt/β-catenin are the main actresses in CRC carcinogenesis in both sporadic (80% APC mutated) and hereditary cases. Moreover, in sporadic tumors, where the APC gene is not mutated, mutations in other pathway components, such as β-catenin, regulated by APC, are found.[14],[15]
Normally, APC blocks the transition from G to S stages of the cell cycle. In the cytoplasm, APC binds some proteins such as axin, glycogen synthase kinase-3β and β-catenin; the latter is so phosphorylated, ubiquitinated, and degraded through the proteasome. If there is no APC complex,β-catenin can move into the nucleus. Here, it activates the transcription of proliferation's genes such as Myc and Cyclin D1. Furthermore, β-catenin stabilizes intercellular junctions and contact inhibition, important factors involved in the metastatic process.[11],[16]
Epidermal growth factor receptor pathway and K-Ras
In the epithelium, epidermal growth factor receptor (EGFR) represents a transmembrane tyrosine kinase that performs its activity of cellular proliferation and survival through two intracellular pathways. Its ligand, epidermal growth factor (EGF), binds the extracellular domain of the receptor leading to its dimerization. Hence, the intracellular domain is autophosphorylated and activates some proteins such as Ras/Raf/Mitogen-activated protein kinases (MAPK) for one pathway and PI3K/AKT for the other pathway [Figure 2]. The First pathway regulates cell proliferation, differentiation, senescence, and apoptosis. The gene that encodes for Raf protein is an oncogene that includes also the H-Ras, N-Ras, and K-Ras isoforms.[17] The last one was the most mutated in 40% of sporadic CRCs. Furthermore, BRAF, a member of Raf family, acts in this pathway and is mutated in sporadic CRCs. After the bond between ligand and receptor, the downstream mechanism includes the activation of cascade signal through PI3K mediated by Ras. This activation inhibits apoptosis, whereas RAF activation stimulates cellular proliferation. For this reason, is clear that K-Ras mutations increase cell proliferation and survival independently of EGFR receptor activation.[11],[18] | Figure 2: Epidermal growth factor receptor Pathway and its two pathways: Ras/Raf/Mitogen-activated protein kinases or PI3K/AKT
Click here to view |
TP53 mutation
TP53 is the guardian of the genome, involved in the control of apoptosis and cell cycle. It is often mutated in several neoplastic diseases included CRC. P53 induces G stage cell-cycle arrest when there are some DNA damages, so enables DNA repair before the DNA replication process. Unsuccessful repair induces cell apoptosis. In general, TP53 mutation occurs at the time of transition from adenoma to cancer lesion where it is overexpressed in 60% of the tumors.[11]
Loss of heterozygosity for chromosome 18
LOH represents the loss of one of the two gene's copies. This LOH in 18q21 is seen in advanced CRC and involves some proteins such as SMAD proteins or DCC. The DCC is a transmembrane protein encoded by a “conditional tumor suppressor gene.” This one, in the absence of netrin-1, its ligand, can block cell growth. This ligand is produced in the colon crypts where its concentration decrease when cell differentiates and moves toward the surface. This gradient contributes to the physiological process of apoptosis and sloughing of epithelial cells. So when the gene is mutated, in the 70% of CRCs cases, netrin-1 will not bind its receptor resulting in abnormal cell survival.[11],[19]
PI3K/AKT pathway and tumor growth factor beta receptor II
As mentioned earlier, EGFR activates 2 pathways. The second one involves PI3K/AKT/mTOR. PI3K phosphorylates AKT that, once activated, phosphorylates several proteins such as mTOR. PI3KCA gene encodes the catalytic subunit of PI3K and is mutated in over 25% of CRCs.
Another protagonist is the TGFβ family that includes several multifactorial proteins involved in many cellular processes. TGFβ receptors have some isoforms: type I (I), type II (RII), and type III (RIII). The type II is mutated in 90% of CRCs with microsatellite instability. In early stages of carcinogenesis, this receptor mediates some suppressive effects, whereas in late stages, it promotes tumor development by inhibiting tumor cell death. It participates also in epithelial-to-mesenchymal transition (EMT) that induces tumor invasion and metastasis. It has been shown that TGFβ RII activates downstream PI3K/AKT. The activation of this pathway can make cells resistant to growth factor deprivation and stress-induced apoptosis and promote cell motility.[11]
Tumor-stroma Interaction | |  |
Nonmalignant cells and tumor microenvironment (TME) interact with each other. Immune cells,[20] endothelial cells and fibroblasts [21] are the main factors of the TME that support the engraftment and self-renewal of CR-CSCs. Therefore, the different tumor-associated cells together with the extracellular matrix (ECM) support tumor formation. On the other hand, tumor cells strongly transform the nature and content of the stroma and TME in general. For these reasons, TME is an important factor in the cancer development and so in metastatic colonization. The understanding of the mechanisms established between tumor cells and TME is of strong scientific interest and could provide potential targets for the development of new anticancer therapies [Figure 3].
Cancer-associated fibroblasts
Cancer-associated fibroblasts (CAFs) have a morphology similar to myofibroblasts. They are large spindle-shaped cells activated during the wound healing process.[22] CRC-associated fibroblasts show specific markers including fibroblast surface protein-1, alpha-smooth muscle actin, vimentin, prolyl-4-hydroxylase, chondroitin sulfate proteoglycan neuron-glial antigen-2, fibroblast-activated protein (FAP), and platelet-derived growth factor receptor.[23] CAFs origin is heterogeneous and still under study. This is to be found in different cell types, such as mesenchymal stem cells derived from bone marrow, resident tissue fibroblasts, hematopoietic stem cells, endothelial cells (mesothelium-endothelial transition [EndMT]) and epithelial cells (EMT).[24],[25] In the liver, for example, activated CAFs are responsible for the production of different factors such as hepatic growth factor (HGF), some cytokines and chemokines, EGF, IGF1 and 2, prostaglandin E2 (PGE2) and the vascular endothelial growth factor (VEGF) that contribute to the tumor metastasization.[26] CAFs increase self-renewal of CR-CSCs and their invasion by specific cytokine and chemokine secretion after chemotherapy treatment such as the interleukin-17 (IL-17A).[21]
Some factors secreted by stromal cells such as HGF, OPN, and stromal-cell-derived factor-1 (SDF-1)[27] increase the CD44-v6 protein required for cell migration. The remodeling of the ECM takes place with the intervention of the matrix metalloprotein family members (MMP). MMP-2 and MMP-9 constitute the basal membrane and contribute to tumor proliferation by degrading type IV collagen and laminin. MMP-9 is activated by MMP-3 and MMP-13.[28] The expression of MMP-9 in fibroblasts is induced by tumor necrosis factor-alpha (TNF-alpha) and TGF-β.[29] CAFs activated with TGF-β secrete IL-11, which leads to increased STAT3-dependent survival [23] thus promoting the onset of liver metastases.[30] For this reason, TGF-β signaling may be therapeutically relevant in the context of CAFs and liver metastases.[31]
Furthermore, CAFs can alter their energy metabolism, which can promote tumor growth. They use aerobic glycolysis as an energy source. This metabolism generates lactate and ketones which, when secreted in the intracellular space, act as paracrine oncometabolites that feed the oxidative mitochondrial metabolism in cancer cells.[32] This aerobic glycolysis is referred to as “Warburg's inverse effect.” The acid environment generated by this metabolic switch not only activates the MMPs, but it also prevents the attack of the growing tumor mass by the immune cells.
Tumour growth factor-beta
Endothelial cells may be involved in the formation of the metastatic niche transforming into cells similar to CAF, through a mechanism driven by TGF-β called EndMT.[31] TGF-β is an important factor for the interactions between metastatic tumor cells and the microenvironment in CRC. This acts as a tumor suppressor during initial tumor formation and instead has a predominant oncogenic role during tumor progression. This switch appears to occur after the acquisition of further mutations involving, for example, P53[33] or SMAD4.[34] In advanced tumors, the increase in TGF-β levels is induced by a prometastatic program activated by cross-talk between tumor cells and stromal cells. Indeed, the TGF-β level increases the survival of metastatic cells and therefore, their organ colonization.[23] CRC cells capable of initiating metastases possess the ability to raise TGF-β levels in the environment by secreting TGF-β or recruiting TGF-β-producing cells such as macrophages, CAFs, or platelets.[23],[24],[25],[26],[27]
Endothelial cell
The transformation of endothelial cells leads to pathological disorders. In normal tissues, these form a continuous and uniform monolayer but, in TME, endothelial cells have an atypical shape and size with ruffled margins and promote a stem cell phenotype.
Numerous cell types referred to as TME.[35] Angiogenesis is one of the hallmarks of cancer. It is necessary for the development of malignant tumors and is mediated by a group of angiogenic factors such as VEGF and the placental growth factor. Many types of tumor cells in a hypoxic environment secrete VEGF.[36],[37] Furthermore, CAFs upregulate the release of IL6 in the presence of tumor cells which increases angiogenesis.[38] Angiogenesis induced with VEGF and IL-6 is further enhanced by the production of VEGF by the same fibroblasts.[38]
Other angiogenic factors are PDGF, FGF, and SDF-1; in human colon carcinomas, among these factors, PDGF-R is mainly expressed by tumor-associated stromal cells and by pericytes of tumor vasculature.[39] The expression of PDGF-R on CAFs, in a dependent stanniocalcin-1 manner, is associated with metastasis and poor prognosis in CRC.[40] A particular cytokine, IL-33, is secreted by endothelial and epithelial cells for the activation of NF-kβ and MAPK signaling and it may have therapeutic interest in the reprogramming of endothelial cells and in the normalization of tumor vasculature in CRC. Most data showed that IL-33 increases endothelial cell proliferation, migration, and differentiation in blood vessels improving neo-angiogenesis.[41]
Immune cells
The immune system acts as an effective protector against cancer and has a dual role, in fact, it is able both to promote and suppress the development of tumor.[42] The immune system counteracts tumor cells with innate immunity cells (e.g. macrophages, neutrophils, and dendritic cells) and adaptive immunity cells (B and T lymphocytes). This system recognizes specific tumor antigens (neo-antigens) expressed by malignant cells. Cancer cells produce PGE2 which plays a key role in the tumor escape phase as it suppresses immunity and induces inflammation. This mechanism is necessary for tumor growth in immunocompetent hosts. This effect has been observed in breast, melanoma, and CRC.[43] Chemokine signaling plays an important role in the recruitment and communication of CRC cells with other TME residents. Finally, it often promotes both the progression of cancer and the spread of metastases.[44] As previously described, tumor and stromal cells secrete chemokines, cytokines, and angiogenic factors. These act as TME immunological modulators. For example, microbial products and interferon-γ attract monocytes to tumor sites that differentiate into alternative mature macrophages (M2 instead of the classic macrophage, M1). Macrophages, one of the most abundant tumor infiltrating cell types, have many tumor-suppressor roles in CRC.[45] The macrophage polarization depends on TME and is a process involving TGF-β, ILs, and chemokines that convert them into alternative macrophages M2.[46] These release IL12, IL23, and nitric oxide increase the expression of the major histocompatibility complex (MHC) which leads to T-cell differentiation and promotion of angiogenesis. During inflammation, the environment stimulates neutrophil migration to the site. In particularly, N1 neutrophils have antitumor effects, and N2 neutrophils have pro-tumor effects. In addition, in this case, TGF-β plays an important role in converting N1 neutrophils to N2 and produces arginase to inactivate the effector functions of T cells. Nevertheless, tumor-associated macrophages and dendritic cells are all sources of CAF for stromal TGF-β.[31]
Regarding T cells, there are three types – CD8+ T cells (cytotoxic T lymphocytes [CTLs]), CD4+ T cells (helper T cells), and regulatory T cells. CTLs recognize specific antigens by mediating immune surveillance and are effective in CRC metastasis.[31] T helper lymphocytes interact directly with MHC Class II molecules on cells, which can influence the behavior of some types of cells, namely macrophages, CTLs, and B cells. All immune cells, in particular, CD4+ T cells, influence self-renewal of CR-CSCs through IL-22 secretion. They in fact activate the methyltransferase DOT1 L responsible for the transcription of genes associated with stem cells.[20] Finally, B lymphocytes differentiate into plasma cells by recognizing the antigen directly or interacting with T helper cells. They release cytokines such as IL10/17/35, IFN-γ and TNF-α for immune system signaling. They also make up long-lived memory B cells. The accumulation of plasma cells or memory B cells is a specific antitumor immune response.
Conclusions | |  |
In this review, we try to explain the CRC metastatic microenvironment. Although many factors and signaling pathways have been described as responsible for tumor progression, a better understanding of microenvironmental factors is essential to improve or design new therapeutic approaches that allow good prognosis even in the CRC advanced stages.
Acknowledgments
The author would like to thank the professor Micol E. Fiori and her research team at the Department of Oncology and Molecular Medicine, Istituto Superiore di Sanitø, Rome, Italy, for assistance learning.
Financial support and sponsorship
Nil.
Conflicts of interest
There are no conflicts of interest.
References | |  |
1. | Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin 2015;65:87-108. |
2. | Vogelstein B, Fearon ER, Hamilton SR, Kern SE, Preisinger AC, Leppert M, et al. Genetic alterations during colorectal-tumor development. N Engl J Med 1988;319:525-32. |
3. | Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. Cell 1990;61:759-67. |
4. | Kolfschoten NE, Wouters MW, Gooiker GA, Eddes EH, Kievit J, Tollenaar RA, et al. Nonelective colon cancer resections in elderly patients: Results from the dutch surgical colorectal audit. Dig Surg 2012;29:412-9. |
5. | Smothers L, Hynan L, Fleming J, Turnage R, Simmang C, Anthony T. Emergency surgery for colon carcinoma. Dis Colon Rectum 2003;46:24-30. |
6. | Kekelidze M, D'Errico L, Pansini M, Tyndall A, Hohmann J. Colorectal cancer: Current imaging methods and future perspectives for the diagnosis, staging and therapeutic response evaluation. World J Gastroenterol 2013;19:8502-14. |
7. | Leufkens AM, van den Bosch MA, van Leeuwen MS, Siersema PD. Diagnostic accuracy of computed tomography for colon cancer staging: A systematic review. Scand J Gastroenterol 2011;46:887-94. |
8. | Mitry E, Guiu B, Cosconea S, Jooste V, Faivre J, Bouvier AM. Epidemiology, management and prognosis of colorectal cancer with lung metastases: A 30-year population-based study. Gut 2010;59:1383-8. |
9. | Massagué J, Obenauf AC. Metastatic colonization by circulating tumour cells. Nature 2016;529:298-306. |
10. | Amjadi F, Mehdipoor R, Zarkesh-Esfahani H, Javanmard SH. Leptin serves as angiogenic/mitogenic factor in melanoma tumor growth. Adv Biomed Res 2016;5:127.  [ PUBMED] [Full text] |
11. | Hamilton SR. Colorectal carcinoma in patients with Crohn's disease. Gastroenterology 1985;89:398-407. |
12. | Javanmard SH, Dana N. The effect of interferon γ on endothelial cell nitric oxide production and apoptosis. Adv Biomed Res 2012;1:69.  [ PUBMED] [Full text] |
13. | Ewing I, Hurley JJ, Josephides E, Millar A. The molecular genetics of colorectal cancer. Frontline Gastroenterol 2014;5:26-30. |
14. | Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell 2013;154:274-84. |
15. | Najdi R, Holcombe RF, Waterman ML. Wnt signaling and colon carcinogenesis: Beyond APC. J Carcinog 2011;10:5.  [ PUBMED] [Full text] |
16. | Polakis P. Wnt signaling and cancer. Genes Dev 2000;14:1837-51. |
17. | Scaltriti M, Baselga J. The epidermal growth factor receptor pathway: A model for targeted therapy. Clin Cancer Res 2006;12:5268-72. |
18. | Hallberg B, Rayter SI, Downward J. Interaction of ras and raf in intact mammalian cells upon extracellular stimulation. J Biol Chem 1994;269:3913-6. |
19. | Pancione M, Remo A, Colantuoni V. Genetic and epigenetic events generate multiple pathways in colorectal cancer progression. Patholog Res Int 2012;2012:509348. |
20. | Kryczek I, Lin Y, Nagarsheth N, Peng D, Zhao L, Zhao E, et al. IL-22(+)CD4(+) T cells promote colorectal cancer stemness via STAT3 transcription factor activation and induction of the methyltransferase DOT1L. Immunity 2014;40:772-84. |
21. | Lotti F, Jarrar AM, Pai RK, Hitomi M, Lathia J, Mace A, et al. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A. J Exp Med 2013;210:2851-72. |
22. | De Wever O, Demetter P, Mareel M, Bracke M. Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer 2008;123:2229-38. |
23. | Calon A, Espinet E, Palomo-Ponce S, Tauriello DV, Iglesias M, Céspedes MV, et al. Dependency of colorectal cancer on a TGF-β-driven program in stromal cells for metastasis initiation. Cancer Cell 2012;22:571-84. |
24. | Orimo A, Weinberg RA. Heterogeneity of stromal fibroblasts in tumors. Cancer Biol Ther 2007;6:618-9. |
25. | Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther 2006;5:1640-6. |
26. | Yamamura Y, Asai N, Enomoto A, Kato T, Mii S, Kondo Y, et al. Akt-girdin signaling in cancer-associated fibroblasts contributes to tumor progression. Cancer Res 2015;75:813-23. |
27. | Todaro M, Gaggianesi M, Catalano V, Benfante A, Iovino F, Biffoni M, et al. CD44v6 is a marker of constitutive and reprogrammed cancer stem cells driving colon cancer metastasis. Cell Stem Cell 2014;14:342-56. |
28. | Knauper V, Smith B, Lopez-Otin C, Murphy G, Activation of progelatinase b (proMMP-9) by active collagenase-3 (MMP-13). Eur J Biochem 1997; 248, 369–373. |
29. | Stuelten CH, DaCosta Byfield S, Arany PR, Karpova TS, Stetler-Stevenson WG, Roberts AB. Breast cancer cells induce stromal fibroblasts to express MMP-9 via secretion of TNF-alpha and TGF-beta. J Cell Sci 2005;118:2143-53. |
30. | González HE, Eugenín EA, Garcés G, Solís N, Pizarro M, Accatino L, et al. Regulation of hepatic connexins in cholestasis: Possible involvement of kupffer cells and inflammatory mediators. Am J Physiol Gastrointest Liver Physiol 2002;282:G991-1001. |
31. | Tauriello DV, Calon A, Lonardo E, Batlle E. Determinants of metastatic competency in colorectal cancer. Mol Oncol 2017;11:97-119. |
32. | Seifabadi S, Vaseghi G, Javanmard SH, Omidi E, Tajadini M, Zarrin B. The cytotoxic effect of memantine and its effect on cytoskeletal proteins expression in metastatic breast cancer cell line. Iran J Basic Med Sci 2017;20:41-5. |
33. | Adorno M, Cordenonsi M, Montagner M, Dupont S, Wong C, Hann B, et al. A mutant-p53/Smad complex opposes p63 to empower TGFbeta-induced metastasis. Cell 2009;137:87-98. |
34. | Zhang B, Halder SK, Kashikar ND, Cho YJ, Datta A, Gorden DL, et al. Antimetastatic role of smad4 signaling in colorectal cancer. Gastroenterology 2010;138:969-800. |
35. | Hanahan D, Coussens LM. Accessories to the crime: Functions of cells recruited to the tumor microenvironment. Cancer Cell 2012;21:309-22. |
36. | Hlatky L, Tsionou C, Hahnfeldt P, Coleman CN. Mammary fibroblasts may influence breast tumor angiogenesis via hypoxia-induced vascular endothelial growth factor up-regulation and protein expression. Cancer Res 1994;54:6083-6. |
37. | Ferrara N, Adamis AP. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov 2016;15:385-403. |
38. | Nagasaki T, Hara M, Nakanishi H, Takahashi H, Sato M, Takeyama H. Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: Anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour-stroma interaction. Br J Cancer 2014;110:469-78. |
39. | Kitadai Y, Sasaki T, Kuwai T, Nakamura T, Bucana CD, Hamilton SR, et al. Expression of activated platelet-derived growth factor receptor in stromal cells of human colon carcinomas is associated with metastatic potential. Int J Cancer 2006;119:2567-74. |
40. | Peña C, Céspedes MV, Lindh MB, Kiflemariam S, Mezheyeuski A, Edqvist PH, et al. STC1 expression by cancer-associated fibroblasts drives metastasis of colorectal cancer. Cancer Res 2013;73:1287-97. |
41. | Zhang Y, Davis C, Shah S, Hughes D, Ryan JC, Altomare D, et al. IL-33 promotes growth and liver metastasis of colorectal cancer in mice by remodeling the tumor microenvironment and inducing angiogenesis. Mol Carcinog 2017;56:272-87. |
42. | Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Integrating immunity's roles in cancer suppression and promotion. Science 2011;331:1565-70. |
43. | Fiori ME, Villanova L, De Maria R. Cancer stem cells: At the forefront of personalized medicine and immunotherapy. Curr Opin Pharmacol 2017;35:1-1. |
44. | Itatani Y, Kawada K, Fujishita T, Kakizaki F, Hirai H, Matsumoto T, et al. Loss of SMAD4 from colorectal cancer cells promotes CCL15 expression to recruit CCR1+myeloid cells and facilitate liver metastasis. Gastroenterology 2013;145:1064-75.e11. |
45. | Braster R, Bögels M, Beelen RH, van Egmond M. The delicate balance of macrophages in colorectal cancer; their role in tumour development and therapeutic potential. Immunobiology 2017;222:21-30. |
46. | Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009;16:183-94. |
[Figure 1], [Figure 2], [Figure 3]
This article has been cited by | 1 |
Ferroptosis open a new door for colorectal cancer treatment |
|
| Hong Liang, Xia He, Yitong Tong, Niuniu Bai, Yushu Pu, Ke Han, Yi Wang | | Frontiers in Oncology. 2023; 13 | | [Pubmed] | [DOI] | | 2 |
Thymidylate Synthase Overexpression Drives the Invasive Phenotype in Colon Cancer Cells |
|
| Wojciech M. Ciszewski, Malgorzata Chmielewska-Kassassir, Lucyna A. Wozniak, Katarzyna Sobierajska | | Biomedicines. 2022; 10(6): 1267 | | [Pubmed] | [DOI] | | 3 |
Platismatia glauca
—Lichen species with suppressive properties on migration and invasiveness of two different colorectal carcinoma cell lines
|
|
| Dragana S. Šeklic, Milena M. Jovanovic | | Journal of Food Biochemistry. 2022; | | [Pubmed] | [DOI] | | 4 |
Systematic review of tumour budding and association with common mutations in patients with colorectal cancer |
|
| Phimmada Hatthakarnkul,Jean A. Quinn,Amna Ahmed Mohemmd Matly,Aula Ammar,Hester C. van Wyk,Donald C. McMillan,Joanne Edwards | | Critical Reviews in Oncology/Hematology. 2021; 167: 103490 | | [Pubmed] | [DOI] | | 5 |
Specific causes of recurrence after surgery and mortality in patients with colorectal cancer: A competing risks survival analysis |
|
| Malihe Safari,Hossein Mahjub,Habib Esmaeili,Mohammad Abbasi,Ghodratollah Roshanaei | | Journal of Research in Medical Sciences. 2021; 26(1): 13 | | [Pubmed] | [DOI] | | 6 |
Expression level of long noncoding RNA NKILA-miR103-miR107 inflammatory axis and its clinical significance as potential biomarker in patients with colorectal cancer |
|
| Davood Jafari,Farshid Noorbakhsh,Alireza Delavari,Javad Tavakkoli-Bazzaz,Samad Farashi-Bonab,Rasoul Abdollahzadeh,Nima Rezaei | | Journal of Research in Medical Sciences. 2020; 25(1): 41 | | [Pubmed] | [DOI] | |
|
 |
 |
|