Abstract
The morbidity of colorectal cancer (CRC) has risen to third place among malignant tumors worldwide. In addition, CRC is a common cancer in China whose incidence increases annually. Angiogenesis plays an important role in the development of tumors because it can bring the nutrients that cancer cells need and take away metabolic waste. Various mechanisms are involved in the formation of neovascularization, and vascular endothelial growth factor is a key mediator. Meanwhile, angiogenesis inhibitors and drug resistance (DR) are challenges to consider when formulating treatment strategies for patients with different conditions. Thus, this review will discuss the molecules, signaling pathways, microenvironment, treatment, and DR of angiogenesis in CRC.
Keywords: angiogenesis, colorectal cancer, drug resistance, therapy, VEGF
Various mechanisms are involved in the formation of neovascularization, and vascular endothelial growth factor is a key mediator. Meanwhile, angiogenesis inhibitors and drug resistance are challenges to consider when formulating treatment strategies for patients with different conditions.

1. INTRODUCTION
As the third most common malignancy worldwide, colorectal cancer (CRC) refers to the malignant transformation of colorectal cells. The global incidence of CRC has increased in recent decades. 1 The mortality rate of CRC ranks second next to lung cancer. When diagnosed, approximately one‐quarter of patients present with metastatic disease. 2 Thus, a cure for cancer based on the mechanism of tumor formation is necessary. Complex networked systems are involved in tumor cell proliferation, survival, angiogenesis, invasion, and metastasis. Given that malignant tumor cells need nutrients, including oxygen and growth factors, to support their growth, they require sufficient blood circulation around them. Tumor vessel formation is an important promoter of tumor growth and metastasis. Folkman proposed that tumor angiogenesis is the beginning of tumor development. 3 However, tumor‐promoting angiogenesis was not demonstrated until vascular endothelial growth factor (VEGF)‐A was identified and its monoclonal antibody was manufactured, which ultimately clarified the preliminary mechanism between neovascularization and tumorigenesis. 3 Angiogenesis indicates that neonatal blood vessels take shape from existing blood vessels. This process is related to the proliferation, migration, and differentiation of endothelial cells (ECs), and multiple procedures during its process are rigorously regulated. 4 In addition, angiogenesis is primarily regulated by growth factors, cytokines, oncogenes, and other modulators. 5 Considering its key role in tumor formation and development, it might be a promising target in tumor treatment. 6 Although various successful studies have been conducted, its mechanism remains unclear, and the effects of targeted therapy are also controversial. These issues will be briefly discussed below.
2. ANGIOGENESIS IN TUMORS
Angiogenesis is rather prominent in various aspects under normal physiological conditions, including tissue growth, wound healing, menstrual cycle, and placental implantation. 7 Regular occurrence of angiogenesis heavily depends on the dynamic equilibrium between the promoting and inhibiting function systems. 8 Angiogenesis is the process of new blood vessel formation. This process plays a key role in the progression of all types of cancers. Tumor masses have high nutritional requirements to support their need for growth; thus, new blood vessels are gradually formed around them to persistently supply oxygen and glucose. 9 , 10 In malignant tumors, angiogenic processes are sustainably maintained in a pro‐angiogenic milieu, 11 similar to a wound that never heals.
2.1. Characteristics of tumor vasculature
Tumor blood vessels are structurally and functionally abnormal compared with normal blood vessels. Besides being heterogeneous, the diameters of most tumor blood vessels are wide and their shapes are curved in different sizes, with few pericytes and defective basement membranes (BMs). 12 Tumor blood vessels are also hyp erpermeable, partly because of the overproduction of VEGF. The loose intercellular junction of tumor endothelial cells (TECs), lack of pericytes, and smooth muscle cells lead to extravasation of plasma fluid and proteins. 13 TECs with altered phenotypic release factors promote tumor progression and downregulate suppressing factors, finally promoting tumor formation adjacent to tumor cells in space. 14 Furthermore, tumor blood vessels are molecularly different because of their responses to environmental cues through transcriptional regulation. 15 In adenoma with dysplasia, pre‐malignant lesions of the colon, VEGF (p < 0.0005), and microvessel density (p < 0.0005) significantly increase. 16 Thus, tumor blood vessels have different characteristics compared with normal blood vessels (Figure 1).
FIGURE 1.

Full‐text mechanism diagram. In normal blood vessels, endothelial cells have tight junctions with a normal number of surrounding pericytes and intact basement membranes (BMs). The diameters of most tumor blood vessels are wide, and their shapes are curved in different sizes, with few pericytes around and defective BMs. Tumor blood vessels are also hyperpermeable. The tumor microenvironment consists of stromal cells, ECM components, and exosomes. Many growth factors that promote angiogenesis and their signaling pathways are involved in these changes. Multiple anti‐angiogenesis agents target different molecules. Drugs targeting immune checkpoints are also illustrated in this figure.
2.2. Types of vascular formation
Tumor blood vessels are formed in various patterns, such as vasculogenesis, sprouting angiogenesis, intussusceptive angiogenesis, and vascular mimicry. Vasculogenesis and angiogenesis are the two main types of vascular formation. In addition, tumor angiogenesis is the most extensively studied pattern of new vessel generation. 17 Vasculogenesis is the process in which endothelial and hematopoietic progenitor cells in the blood cycle are recruited by cytokines and chemokines to create new vessels. 18 Sprouting angiogenesis is the sprouting of neoplastic capillaries from existing ECs and tumor blood vessels; it might be the most characteristic way for the tumor to obtain oxygen and nutrients. 19 , 20 VEGF is the key player driving this process, which occurs in pathological situations in malignant tumors. 21 However, studies on intussusceptive angiogenesis are limited; in this type of angiogenesis, transluminal tissue pillars grow in pre‐existing vessels and then integrate to recreate the vascular system. 22 It is also referred to as a “complementary method” to sprouting angiogenesis. 23 Contrary to canonical tumor angiogenesis, vasculogenic mimicry (VM) supplies independent blood perfusion to tumor cells. 20 The phenomenon of vascular mimicry exists in various kinds of tumor masses, including CRC. 24 The mechanism is a complex process and has not been distinctly distinguished. In particular, CRC stem cells (CRCSCs) are transdifferentiated to create vascular tube structures that assist tumor blood supply independent of tumor angiogenesis. 25 Nevertheless, epithelial–mesenchymal transition (EMT) and tumor microenvironment (TME) are related to sprouting angiogenesis. 20 As VM cells directly come into contact with blood flow, shed cells can be easily transported. Therefore, VM is related to advanced malignant tumors with high invasion and high metastasis, which are indicators of poor patient prognosis. 26 Moreover, vessel co‐option indicates that tumor tissue receives oxygen and nutrients from pre‐existing vessels rather than creating new vascellums. 27 , 28 , 29 Vessel co‐option is a possible cause of anti‐angiogenic drug resistance (DR). Frentzas showed that vessel co‐option might be involved in an undesirable response to anti‐angiogenic agents in patients with CRC and hepatic metastases. 28 Utilization of numerous angiogenesis types, together with some normal physiological balance disruptions, can eventually lead to the unbridled growth of tumor cells.
3. ANGIOGENESIS AND COLORECTAL CANCER
Similar to other observed solid tumors, CRC development, progression, and metastasis depend heavily on angiogenesis. 30 , 31 Numerous molecules participate in the process, such as growth factors (VEGF and epidermal growth factors [EGFs]), fibroblast growth factor (FGF)‐2, transforming growth factor (TGF)‐α and TGF‐β, angiopoietins (Angs), platelet‐derived growth factor (PDGF), membrane‐bound factors (integrins, ephrins, cadherins, matrix metalloproteinases [MMPs], and hypoxia‐inducible factor‐1 [HIF‐1]). 32 Figure 1 illustrates some prominent pro‐angiogenic factors.
3.1. Molecules in angiogenesis
Vascular endothelial growth factor plays a crucial role in all forms of solid tumor growth and development, including CRC, by causing the formation of new blood vessels. Its high serum level is closely associated with CRC and its clinical stages. 33 Derived from tumor cells and the surrounding microenvironment, it is upregulated by hypoxia, growth factors, and cytokines such as IL‐1, EGFs, PDGFs, and tumor necrosis factor (TNF)‐α. 34 , 35 , 36 Furthermore, VEGF‐mediated pathogenic effects on vascular permeability are primarily caused by junction remodeling, fenestra induction, and vesiculo‐vascular organelles (VVOs). 37 As a survival factor, VEGF can accelerate the growth and prevent apoptosis of vascular ECs. 38 The seven members of the VEGF family are VEGF‐A to VEGF‐F and placenta growth factor (PlGF). 35 Among them, VEGF‐A, VEGF‐B, and PIGF stimulate angiogenesis, whereas VEGF‐C and VEGF‐D are responsible for lymphangiogenesis. 39 VEGF‐A undergoes alternative splicing of pre‐mRNA from eight exons that leads to multiple isoforms, including VEGF121, VEGF165, VEGF189, and VEGF206. 34 , 40 The overexpressed VEGF165 increases the expansion of the tumor by promoting the recruitment of smooth muscle cells and vessel maturation in CRC. 41 Three VEGF receptors exist in CRC: VEGFR‐1, VEGFR‐2, and VEGFR‐3. VEGFR‐1 is related to tumor grade, Dukes’ stage, and lymph node involvement; VEGFR‐2 is correlated with lymph node involvement; and VEGFR‐3 is not associated with any of the clinicopathological variables. 42 Activated VEGFR‐2 leads to the activation of various signaling pathways, including the PLCγ and RAS/RAF/MEK/ERK (MAPK) pathways, by which the growth of EC is promoted, and the PI3K/AKT pathway, by which cells might avoid apoptosis. 43 VEGF might suggest metastasis and poor prognosis of CRC. 44 Overexpressed VEGF was detected in 70% of the cases in a study where 50 patients with CRC were enrolled, thereby revealing patients’ poor prognosis as its tight relationship with tumor size, grade, and advanced tumor stage (p = 0.006, p < 0.001, p < 0.001, respectively). 45 Meanwhile, overexpression of VEGF‐A can reverse the inhibition of CRC cell proliferation, migration, invasion, and angiogenesis caused by HIF‐1miR‐150‐5p. 46 Moreover, VEGF‐VEGFR activity might be altered by HIF‐1, COX‐2, mutated K‐RAS, and p53, which can promote the malignant phenotype of tumor cells such as proliferation and migration. 47
Binding with EGFR and EGF can activate several crucial downstream signaling pathways, including PI3K/AKT/mTOR, MAPK, and Janus kinase (JAK)/Signal Transducer and Activator of Transcription 3 (STAT3), in regulating cell growth survival, invasion, and migration. 48 , 49 , 50 Dysregulation of the EGFR signaling pathway often occurs in human cancers, including CRC. 51 The erythroblastosis oncogene B (ErbB)/human epidermal growth factor receptor (HER) family is composed of tyrosine kinase receptors (RTKs), which contain ErbB1 (EGFR/HER1), ErbB2 (Neu/HER2), ErbB3 (HER3), and ErbB4 (HER4). 52 , 53 HER2 is a special member of the EGFR family, which is activated by heterodimerizing with other ligand‐bound receptors but not ligands. 54 Gene amplification and missense mutation are the most common alterations in HER2 in 7%–8% CRC. 55 Approximately 5% of metastatic CRC tumors are facilitated by amplification or mutation of HER2, which might be a cancer‐promoting factor, a prognostic and diagnostic biomarker, and a promising treatment target in CRC. 56 EGFR was found to be highly expressed in 85.2% of the 54 patients with cancer and correlated with tumor sizes and invasion depth (p = 0.043 and p = 0.05, respectively). 57 In addition, angiogenic‐associated cytokines including VEGFA and IL‐8, produced by the EGFR/Akt/NF‐κB pathway, can be activated by highly dry human CRC cells, thereby inducing angiogenesis. 58 Heparin‐binding EGF‐like growth factor (HB‐EGF), a member of the EGF family, and EGF are both pro‐angiogenesis mediators that work by activating signaling pathways including PI3K, MAPK, and eNOS. 59
Twenty‐three small heparin‐binding growth factor members contained in the FGF family are highly conserved and bind to one or more of the four highly affine FGFRs (FGFR1–FGFR4). 6 FGF2 is also a basic FGF (bFGF) that plays dual roles not only as a stimulator of VEGF‐A expression in ECs or stromal cells but also as a regulator of VEGFR‐2 signaling; thus, it is the most distinctive mediator in regulating angiogenesis. 60 In addition, high expression of FGFs and FGFRs in cancers including CRC and its pathway functional mechanism is related to tumor growth and invasion. 61 The MAPK, PI3K, PLCγ, or STAT pathways, activated by the binding of FGFs and FGFRs, participate in regulating cell proliferation, survival, differentiation, migration, and angiogenesis. 62 Knuchel et al. showed that fibroblasts induce cell contact‐dependent CRC cell migration and invasion in vitro under 2D and 3D conditions by FGF‐2. They are located on the fibroblast cell surface, activate SRC, and are mediated by FGFR, and the adhesion of cancer cell to fibroblast is dependent on αvβ5 integrin. 63
The PDGF family harbors four heparin‐binding polypeptide growth factors: A, B, C, and D. PDGF has a wide range of sources, such as activated platelets, ECs, epithelial cells, inflammatory cells, and glial cells. Moreover, its targets contain a broad category of cell types, including fibroblasts, pericytes, and smooth muscle cells. 64 All members in the PDGF family show strong angiogenic capacity in vivo, but the PDGF‐B/PDGFRβ axis is the most representative. 65 PDGFs and PDGFRs are expressed in a large range of malignant tumors, and their activation is relevant to tumor growth, metastasis, invasion, and angiogenesis by stimulating downstream signaling pathways. 66 Pericytes and vascular smooth muscle cells (VSMCs) are the major targets of the PDGF/PDGFR signaling pathway, and they are known to promote angiogenesis. Higher preoperative intraplatelet VEGF and PDGF levels were detected in patients with CRC than in controls. 67 Higher expression of PDGF‐BB was found in patients with CRC than in patients with adenoma. Consistently, the high levels of PDGFR α/β in patients with CRC were related to tumor invasion and metastasis. 68 PDGF‐BB can elicit different downstream cascade pathways, including PI3K, PLCγ, MAPK, and JAK/STAT. 69
Similar to VEGFs, Angs are one of the growth factors that can regulate vascular homeostasis and tissue repair. Three members are included in the human Angs family (Ang‐1, Ang‐2, and Ang‐4), whereas their receptors are Tie‐1 and ‐2, which are highly expressed transmembrane tyrosine kinases in ECs. Ang‐1 and ‐2 are the main ligands of Tie‐2; upon their activation, several downstream signaling pathways are stimulated, including PI3K/AKT, MARK, caspase‐9, eNOS, Bad, and survivin. 70 , 71 , 72 Ang‐1 promotes the recruitment of smooth muscle cells and pericytes to maintain the stabilization of mature vasculature. 73 However, as a prognostic factor in metastatic CRC, 74 Ang‐2 guides vasculature conversion to an unstable state, which is easier to induce under the effect of VEGF. 75 Thus, Ang‐2 might be highly expressed in tumor cells, whereas Ang‐1 is the opposite. This deduction was verified by Ahmad et al. 76 Ang‐2 was found to be an oncogene, whose malignant biological functions were investigated with shRNA in the LoVo CRC cell line, and its expression was convoluted with clinicopathological parameters in CRC. 77 Similar to VEGF, Ang‐2 expression is induced by hypoxia in CRC. 78
3.2. Signaling pathway in angiogenesis
The phosphatidylinositol‐3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling pathway overexpression has been identified in various malignant tumors, including primary and metastatic CRCs. 79 Common in CRC, overactivation of mTOR signaling is strongly related to cancer progression, initiation, and DR. In CRC, mTOR significantly regulates proliferation, survival, growth, differentiation, and autophagy. 80 The PI3K/AKT/mTOR pathway is an intracellular signaling pathway composed of different kinases. Dysregulation of the PI3K signaling pathway is caused by the amplification and genetic mutation of the PI3K gene, encoding a catalytic and regulatory subunit of PI3K isoforms, and various protein mutations described in this pathway, which is responsible for cell growth, proliferation, survival, and angiogenesis. 81 , 82 For angiogenesis, mTOR is important for energy metabolism in ECs. It receives signals from PI3K‐Akt, which can be upregulated by growth factors (VEGF, insulin‐like growth factor‐1 [IGF‐1], and EGF) and their respective receptors. 83 , 84 PI3K/Akt signal transduction, which is essential for proliferation, metastasis, and survival, is one of the classic ways to increase blood vessel quantity and vascular permeability. Blood vessel reconstruction can be achieved by VSMC phenotype transformation. 13 , 85 Increased EGF upregulates hydrogen peroxide (H2O2) synthesis, which stimulates ribosomal protein S6 kinase beta‐1 (S6K1) or p70S6K1 via the PI3K/AKT/mTOR pathway, eventually activating VEGF. 86 In CRC, other angiogenic factors can also be regulated by the PI3K/AKT pathway, including angiopoietins and nitric oxide (NO). 84
The human TGF‐β family consists of 33 identified members, including three TGF‐β isoforms, three activins, growth and differentiation factors, and nodal and bone morphogenetic protein. 87 In addition, the SMAD family is the downstream effector of this signaling pathway. 88 Nevertheless, the absence of SMAD4 upregulates VEGF expression, increases the vascular quantity, and promotes tumor metastasis in the CRC HCT116 cell line. 89 The combination of TGF‐β and its type II TGF‐β receptors (TGFBR2) initially activates the TGF‐β signaling pathway. 90 Under normal conditions, TGF‐β suppresses normal intestinal epithelial cell proliferation and induces apoptosis and differentiation. 91 However, highly expressed TGF‐β plays a pro‐tumor role in the late stage of CRC, thereby increasing the production of several mitogenic growth factors, including TGF‐α, FGF, and EGF. 92 Recently, CRC was suggested to be classified into four consensus molecular subtypes (CMS) based on transcriptomic properties and the following molecular characteristics: CMS1 (microsatellite instability [MSI] immune), with DNA damage repair and defective DNA mismatch repair, BRAF mutations, and diffuse immune infiltrate; CMS2 (canonical), with more oncogene expression with absent tumor suppressor gene expression, epithelial differentiation, Wnt and MYC pathway upregulation; CMS3 (metabolic), with KRAS activating mutations and metabolic dysregulation; and CMS4 (mesenchymal), with angiogenesis, EMT, TGF‐β signaling activating, and matrix remodeling. 93 The TGFBR2 gene, containing microsatellite sequence, accumulates MSI mutations to a high level in nearly all CRC cells. Nonetheless, TGF‐β signaling activation remains functional to promote the tumor–stromal interaction, which has a positive correlation with malignant cell phenotype and poor prognosis. 94 , 95 Secreted by TGF‐β‐stimulated cancer‐associated fibroblasts (CAFs), IL11 acts on GP130/STAT3 signaling in tumor cells. 96
The NF‐κB family contains five members that interact with one another to homodimerize or heterodimerize: NF‐κB1 (p50), NF‐κB1 (p52), RelA (p65), RelB, and c‐Rel. 97 The IKK kinase complex consisting of catalytic subunits IKKα and IKKβ and regulatory subunit NF‐κB (an essential modulator) is the key element of the NF‐κB signaling cascade. 98 NF‐κB signaling has two different but interactive pathways: the canonical (activated by TNF‐α, lipopolysaccharide, and IL‐1) and non‐canonical (activated by BAFF, CD40, receptor‐activated NF‐κB ligand, and lymphotoxin β) pathways. 99 Their activation is positively associated with cell survival, angiogenesis, apoptosis, and metastasis in CRC. 100 Moreover, in CRC, the expression of NF‐κB (p65) is directly associated with the expression level of HIF‐1α, VEGF, and vascular invasion. 101 The NF‐κB pathway promotes the expression of multiple angiogenic factors, including VEGF, PDGF‐BB, CXCL1, CXCL8, MMP‐2, MMP‐9, COX‐2, and IL‐8, ulteriorly inducing tumor angiogenesis. Highly expressed B7‐H3 in the tissues of patients with CRC promotes VEGFA expression through the NF‐κB pathway. In other words, the B7‐H3/NF‐κB/VEGFA axis plays a role in CRC angiogenesis. 102 As one of the immune checkpoint proteins, such as star molecules CD80 and CD86, B7‐H3 (CD276) was detected in various malignant masses, including CRC. 103 B7‐H3 has immunomodulating effects and participates in regulating angiogenesis. 104 In the study of tumor‐bearing mice, highly expressed B7‐H3 was found to be correlated with increases in TGF‐β and interleukin 10 (IL‐10). 105 This phenomenon might activate the JAK–STAT pathway and promote VEGF expression, thereby inducing angiogenesis. 106
As another critical signaling pathway, JAK/STAT occupies a place in the list of regulating angiogenesis and other pathological processes in CRC. JAK and STAT are two key tyrosine kinase‐associated receptors in this pathway. They become activated after being coupled. Four proteins constitute the JAK family: JAK1, 2, 3, and TYK2. 107 Meanwhile, the STAT family has seven members: STAT1 to STAT7. 108 Binding to their ligands, RTK, cytokine receptors, or G‐protein‐coupled receptors (GPCRs) recruit JAK, which facilitates signal transduction and STAT3 phosphorylation and homodimerization. 109 By promoting gene expression and release of inflammatory mediators to activate the STAT3 signaling pathway, STAT2 is considered a factor in promoting the development of CRC. 110 In a hypoxia micro‐environment, activated by reactive oxygen species (ROS), a mechanistic target of rapamycin complex 1, and/or IL‐6 in cancer cells, STAT3 finally induces HIF‐1a expression. This transcription factor promotes the transcription of VEGF. These events act on surrounding ECs to reactivate STAT3 via VEGFR. In ECs, together with HIF‐1a and specificity protein 1 (sp1), STAT3 upregulates gene expression that induces EC growth, survival, and migration, resulting in angiogenesis. 111 Otherwise, several promoting factors have been reported, including IL‐6, EGF, IL‐11, and solute carrier family 6 member 14 (SLC6A14). 112 , 113 , 114 , 115
The aberrant activation of the Wnt signaling pathway is another major molecular mechanism in CRC development and progression. 116 Humans have 19 Wnts, suggesting it is a complicated signaling and biological process. 117 The Wnt signaling pathway has two branches: canonical and non‐canonical pathways. 100 β‐catenin is a crucial member of the canonical pathway as a switch, and its expression is promoted by activated Wnt signaling. 118 Mutations of the β‐catenin gene associated with nuclear localization of the protein have been mainly detected in microsatellite unstable CRC. 116 Wnt/β‐catenin signaling upregulates angiogenic factors such as VEGFs, chemokines, and MMPs. 116 Wnt2 is primarily produced by CAFs, thereby inducing invasive and metastatic CRC cell phenotypes. 119 Wnt2 levels are closely correlated with the expression of angiogenesis‐associated regulators, including Ang‐2, PlGF, granulocyte colony‐stimulating factor (G‐CSF), IL‐6, and extracellular matrix components. 120 The expression levels of transglutaminase 2 (TGM2) and transmembrane‐4 L‐six family member‐1 (TM4SF1) are both higher in CRC tissues than in normal tissues. TGM2 upregulates the expression of Wnt3a and β‐catenin, whereas TM4SF1 activates the Wnt/β‐catenin/c‐Myc/SOX2 signaling pathway, thereby promoting angiogenesis in CRC. 100
Moreover, the COX‐2/PGE2/EP4 axis, 121 notch signaling pathway, 100 c‐MET/hepatocyte growth factor (HGF) signaling pathway, 122 and EPH/ephrin signaling system 123 are all related to angiogenesis in CRC. These various factors and pathways are connected and cross each other like points and lines. Therefore, angiogenesis is stimulated by multiple points to achieve promotion. The complex network they form is like a map but with landmarks, which can be inferred as targets for diagnosis, prediction, or treatment.
3.3. Tumor microenvironment
The TME is a complex system formed by the interaction of different cells and their products, which can be described as “soil” cultivating tumor cells. 124 It is important for tumor progression and metastasis and is functionally immunosuppressive. 125 , 126 It is a compound comprising stromal cells (including pericytes, ECs, fibroblasts, macrophages, regulatory T cells, myeloid‐derived suppressor cells, and platelets), ECM components (including inflammatory cytokines, chemokines, MMPs, integrins, and other secreted molecules), and exosomes. 127 TECs are the innermost cells of tumor blood vessels, which are mostly derived from normal vascular ECs or directly differentiated from tumor cells. Their irregular cell morphology and phenotype lead to the defective barrier function of blood vessels, which can be beneficial for tumor survival. 125 , 128 In the processes of tumorigenesis and angiogenesis, multifunctional pericytes, together with ECs, play a key role in BM remodeling in the TME. 12 , 129 Immune functions of pericytes might be involved in the exit of innate leukocytes in inducing angiogenesis, regulating lymphocyte activation, and controlling phagocytic activity. 130 CAFs, including fibroblasts or myofibroblasts, take effect in the ECM through intercellular contact, soluble growth factor (FGF2 and VEGFA) secretion, and promotion of ECs’ malignant phenotype transformation. 131 Autophagy of CAFs induced by oxidative stress has a positive effect on tumor proliferation and metabolism. 132 Exosomes secreted by CAFs promote CRC metastasis and chemotherapy resistance. 133 Expressed in CAFs, endoglin is involved in CAF‐mediated invasion and metastasis through TGF‐b signaling pathway activation. 134 Tumor‐associated macrophages (TAMs) are indispensable in TME and control angiogenesis. 135 TAMs were detected both in vivo and in human tissues as a main source of multiple kinds of pro‐angiogenic and ECM remodeling regulators, including VEGF, EGF, PDGF, TGF‐α, TGF‐β, Ang‐1 and 2, and MMPs (e.g., MMP2, MMP9, and MMP12). 136 The interaction between TAMs and CRC cells mediated by cytokines or exosomes can jointly promote the metastasis of CRC by regulating the EMT of tumor cells and the M2‐type polarization of TAMs. 137 Tumor‐associated neutrophils (TANs) promote tumor invasion and angiogenesis by upregulating MMP9, VEGF, and HGF in the primary and metastatic sites. The neutrophil‐to‐lymphocyte ratio is a promising predictive marker of CRC. 138 As a member of various proteases, the MMP family plays a fundamental role in angiogenesis. 116 It is pivotal in degrading ECM, which makes the blood vessel permeable and allows cancer cells to dissociate from the tumor mass and be easily transferred. 139 In colon cancer, the upregulated MMPs are correlated with tumor progression, and some of them influence tumor invasion, metastasis, or poor outcomes. In Buttacavoli et al., MMP2 and MMP9 were found to be expressed more in colon cancer tissues than in normal adjacent tissues. 140 Moreover, the MMP family is regulated by circRNAs in ECM remodeling. 141 Citrullination of ECM, expression of peptidylarginine deiminase 4 (PAD4, a member of the PAD family), and expression of tenascin C (TNC, a glycoprotein in ECM) were found to promote liver metastasis in human CRC. 142
Inflammation has been well established as a hallmark of CRC. Pro‐inflammatory factors promote tumor growth and angiogenesis while inhibiting apoptosis and suppressing antitumor activities. 143 Myeloid‐derived suppressor cells (MDSCs) in the TME have dual functions. They promote the metastasis of tumor cells by infiltrating primary tumors and promoting tumor angiogenesis. MDSCs also inhibit the immune response to accelerate tumor progression. MDSCs maintain the survival and proliferation of tumor cells by releasing inflammatory cytokines (IL‐1, IL‐6, IL‐23, and IL‐17A) or possibly induce adaptive anti‐tumoral immunity by IL‐12, interferon‐gamma (IFN‐γ). 144 Overexpressed CD33+ CD11b+ HLA‐DR−MDSCs were found in primary CRC tissues, which suggested advanced TNM stage and lymph node metastasis. 145 IL‐1α is one of the dominant inflammatory mediators influencing the pathogenesis of inflammation‐associated CRC. It enhances angiogenesis, metastasis, DR, and inhibition of tumor‐suppressive genes in CRC. 146 IL‐17A is a pro‐inflammatory cytokine that contributes to the pathogenesis of inflammatory and autoimmune diseases. 147 High levels of IL‐17 in serum and tissues of patients with CRC are important in the metastasis and prognosis of CRC. 148 IL‐17A activates the ERK, p38 MAPK, and NF‐κB signaling pathways within transformed enterocytes, thereby inducing early tumor development in mice. 149 As a tumor prompter, IL‐17A also relies on stromal cells in the microenvironment. In murine models, tumor‐infiltrating Th17 cells and IL‐17 can stimulate TAF to release G‐CSF, which, in turn, recruits MDSCs to the tumor parenchyma. 150 MDSCs produce VEGF, prokineticin 2/Bv8, MMP9, and pro‐inflammatory S100A8/9 molecules (calprotectin). 151 Chemokines are GPCRs binding small peptides, which mediate angiogenesis, inflammation, and chemoattraction. 152 Tumor cells often secrete a few inflammatory chemokines, such as neutrophil‐attracting CXC‐chemokines. After combining with CXCR1 and/or CXCR2, CXC‐chemokines can induce the migration of TANs. In the background of the TME, increasing evidence has revealed that by infiltrating into tumor tissues, neutrophils play a prominent role in promoting tumors and embracing growth, invasion, angiogenesis, and metastasis in various cancers, including CRC, although they were originally thought to be antitumor cells. 138
Continuously driven by cancer‐promoting factors in the tumor, novel vascular networks might be incapable of maturing and pruning, vascular diameters are not of uniform size, and blood flow through the poorly organized and malformed vessels is possibly chaotic. 153 Along with high tumor cell density, hypoxia is induced within the tumor mass. Hypoxia is not only a hallmark of cancer that influences cancer cells’ function but also an important component in the TME because it alters the extracellular matrix, modulates the tumor‐immune response (immune cell infiltration, immune checkpoint [IC] expression, and secretion of immune molecules) in the TME 154 and increases angiogenesis. Tumor hypoxia promotes the recruitment of ECs and pericytes to stimulate angiogenesis by inducing VEGF, particularly VEGF‐A, 155 and hastens the recruitment of bone marrow‐derived cells (BMDCs). Recruited stromal cells heighten tumorigenesis through extracellular matrix remodeling, growth factor signaling, and evasion of the antitumor immune response. 126 A hypoxic environment inhibits immune response by promoting ICs such as PD‐L1 expression, stimulating immunosuppressive cells including TAMs, MDSCs, and regulatory T cells (Treg cells), as well as inhibiting tumor‐infiltrating lymphocyte (TIL) infiltration. 156 , 157
Tumor hypoxia is primarily attuned by a transcription factor family, described as HIFs. 158 HIFs are heterodimeric transcription factors consisting of one of three possible isoforms of an O2‐labile α subunit (HIF‐1α, 2α, and 3α) and a HIF‐1β subunit. Expression of HIF‐1α induced by hypoxia promotes abnormal angiogenesis formation and enhances CRC metastasis. 154 Evidence from Arabsorkhi et al. showed that the level of HIF‐1α expression in CRC is related to different MSI classifications. 159 Overexpressed HIF‐1 acts as a master regulator of oxygen‐regulated gene expression; in addition, its target genes are particularly relevant to cancer‐encoding angiogenic factors, proliferation/survival factors, glucose transporters, and glycolytic enzymes. 160 In the hypoxic environment, ubiquitin‐mediated HIF‐1 degradation is disabled, leading to the accumulation of HIF‐1, 161 which plays an active role in promoting the expression of angiogenesis‐associated target genes, including HIF1, VEGF, PLGF, Ang‐2, TGF‐β, and hypoxaMIRs. 162 The supposed Warburg effect indicates that regardless of oxygen level, tumor cells preferentially utilize glycolysis to produce lactic acid for energy supply. This process has extremely high efficiency in energy generation to meet the nutritional needs of tumor cells’ rapid growth. 163 The increased HIF‐1α can augment the Warburg effect. 162
As a gaseous free radical, NO acts as a signal that is relatively stable in biological systems. 164 It is involved in angiogenesis and stimulates the EGF‐R signaling pathway. NO mediates stimulatory effects on tyrosine phosphorylation of EGF‐R. BK‐mediated angiogenesis in ECs involves the induction of the expression of VEGF associated with the activation of the NO/EGF‐R/p21Ras/ERK1/2 MAP kinase signaling pathway, which indicates that NO generation plays a role in the expression of VEGF. 165 Nearly all cells, whether normal or malignant, produce NO by taking advantage of heme enzymes from the family of NADPH cytochrome P450 reductases, referred to as NO synthase (NOS). 166 Three isoforms have been identified in mammals: NOS1 (neuronal) and NOS3 (endothelial), which are persistently expressed, and NOS2, which needs to be induced and was initially characterized in macrophages. All isoforms require l‐arginine as a substrate for NO synthesis. NOS2 and NOS3 play key roles in angiogenesis with the regulation of VEGF. 164 Endogenous NO promotes colon neoplasms. Moreover, NO is a crucial factor in many signaling pathways in CRC, including the Wnt/β‐catenin and ERK pathway, which are relevant to cancer initiation, metastasis, inflammation, and chemoresistance/radioresistance. Thus, NO/NOS is expected to be a promising target for the treatment of CRC. 167
Exosomes are membrane‐bound extracellular vehicles (EVs) that can transmit bioactive molecules between different cells in vivo. 168 These signaling molecules have been identified as miRNAs, mRNAs, lncRNAs, and proteins. 169 Exosomes released by various cell types perform various biological functions, primarily mediating communication between different cells, particularly those active in cancer, including CRC. 170 The ncRNAs released from exosomes play a pivotal role in multiple processes of tumor formation and development, including proliferation, differentiation, angiogenesis, migration, and apoptosis. 171 , 172 The quantity and contents of exosomes are significantly different in tumors or normal tissues; thus, exosomes might be diagnostic indicators of CRC. 168 Numerous EV‐miRNAs are not only possible diagnostic markers in CRC, including miR‐1246, miR‐21, miR‐92A, and various others, 173 but also useful prognostic markers, such as miR‐27a and miR‐130a, which indicate poor prognosis. 174 Exosomal miR‐21 derived from transformed cells regulates VEGF and angiogenesis in recipient cells. 175 In addition, cancer‐derived exosomes activate the angiogenic properties of macrophages, such as producing VEGF. Meanwhile, exosomes from macrophages are thought to disturb the adhesion, morphology, and apoptosis of tumor cells, thereby contributing to their migration, invasion, and metastasis. 176
3.4. RNA: Long non‐coding RNAs, miRNA, and circRNA
Long non‐coding RNAs (lncRNAs), miRNA, and circRNA belong to the family of non‐coding RNAs (ncRNAs). Acting as oncogenes or tumor suppressor genes in CRC, they might be potential diagnostic biomarkers. 177
Long non‐coding RNAs are longer than 200 nts non‐coding transcripts, which recently became one of the largest and most significantly diverse RNA families. As small and long evolutionarily conserved ncRNA families, lncRNAs activate and repress genes through various mechanisms at both transcriptional and translational levels. 172 They have dual effects on tumor cell proliferation, angiogenesis, and DR by promoting or inhibiting them. Thus, DR affected by lncRNA appears in various aspects of tumor treatment strategies involving chemotherapy, targeted therapy, and immunotherapy. 178 , 179 Recently discovered lncRNA SET binding factor 2 antisense RNA 1 (lncRNA SBF2‐AS1), an oncogenic antisense RNA to SBF2, is located at 11p15.1 locus and is 2708 nt long. Furthermore, lncRNA SBF2‐AS1 participates in the progression of various tumors, including CRC. 180 lncRNA and miRNA interact with each other. On the one hand, lncRNA can regulate diverse functions and expression levels of miRNA as endogenous regulators. On the other hand, miRNA affects the stability of lncRNA after combining with it. lncRNA can competitively bind to the target mRNA of miRNA to isolate it, thereby inhibiting its function. Multiple pairs of interacting lncRNA and miRNA have been identified in angiogenesis, such as H19/miR‐let‐7 and NFIA/miR‐382‐5p. 181 , 182 Interactions among lncRNA/miRNA/mRNA have been found in liver metastasis, EMT, inflammation formation, and chemoresistance/radioresistance in CRC. In summary, lncRNAs play an important role in CRC growth and metastasis. 183
miRNAs are highly conserved short single‐stranded ncRNAs (18–22 nucleotides). They promote regulatory effects via the 3′‐untranslated binding region (3′‐UTR) of target messenger RNA in the posttranscriptional regulation of genes, 184 which indicates that miRNAs modulate protein‐coding gene expression primarily through mRNA degradation or silencing. 172 In addition, miRNAs have two contradictory functions in regulating angiogenesis in CRC. Some miRNAs directly affect VEGF or inhibit angiogenesis through certain signaling pathways (PI3K/AKT and HIF‐1a). By contrast, some other RNAs, particularly exocrine‐derived ones, can promote angiogenesis. Along with angiogenesis, miRNAs also have effects on cancer genesis, invasion, and metastasis as diverse functions. 182 , 185 miRNAs that promote or inhibit angiogenesis are shown in Table 1.
TABLE 1.
| miRNA | Function | Targets | Result |
|---|---|---|---|
| miR‐145, miR‐206, miR‐148a, miR‐195‐5p, miR‐107 | Inhibit | HIF | Inhibit angiogenesis |
| miR‐622, miR‐590‐5p, miR‐520a, miR‐126, miR‐27b, miR‐150‐5p, miR‐1249 | Inhibit | VEGF | |
| miR‐218 | Inhibit | Connective tissue growth factor | |
| miR‐125a‐3p, miR‐143 | Inhibit | PI3K/AKT | |
| miR‐7, miR‐375 | Inhibit | EGFR | |
| miR‐181a‐5p | Inhibit | MMP‐14 | |
| miR‐181a | Activate | SRC (increased VEGF secretion) | Promote angiogenesis |
| miR‐1229 | Inhibit | HIPK2 (inhibits VEGF angiogenic gene) | |
| miR‐194 | Inhibit | Platelet‐reactive protein 1 (TSP‐1 inhibits VEGF) | |
| miR‐25‐3p | Inhibit | KLF2/KLF4 (inhibit promoter activity of VEGFR2) |
Abbreviations: HIF, hypoxia‐inducible factor; VEGF, vascular endothelial growth factor.
Previous studies showed that circRNAs also play multiple roles in regulating the TME. 187 circRNAs are a family of single‐stranded closed‐circle molecules that lack 5′ and 3′ ends and poly(A) tails, which make them capable of resisting RNase R, leading to high stability. 188 As an indispensable factor inhibiting angiogenesis, circRNAs can conversely promote VEGFA and the expression of other pro‐angiogenic molecules to positively regulate angiogenesis. 187 For example, the high expression of circ‐Erbin was found in CRC, thereby promoting the miR‐125a‐5p‐5p/miR‐138‐5p/4E binding protein 1 axis to elevate the expression of HIF‐1α and finally inducing angiogenesis. 189 By contrast, in glioblastoma multiforme cells, circSMARCA5 was confirmed to downregulate the expression of VEGFA through alternative splicing of its pre‐mRNA to limit the increase of blood vessel density. 190 Several studies showed similar results; the expression levels of different circRNAs might be upregulated or downregulated in CRC. Collectively, these research findings revealed that circRNA is related to the progression and pathogenesis of CRC. 191 As they can be both oncogenic and anti‐oncogenic, circRNAs can potentially be utilized in the treatment and prognosis of CRC. 192
Therefore, by enlarging our perspective to the level of the microenvironment, the participation of different functions of various cells and the specific release of different types of signals make this mixture similar to soil with abundant nutrients, which is greatly different from the normal tissue environment. Such an environment is more conducive to the growth and development of tumors than a normal tissue environment.
3.5. Gut microbiota
Being parasitic in the human intestinal tract, the microflora is a huge organism that can interact with the host. As for CRC, the gut microbiota plays a special role. Gut microflora is important for host survival and health because of its antitumor capability, and it can alleviate tissue damage by reducing the level of oxygen free radicals. A significant abundance variation of intestinal flora was found in patients with CRC compared with healthy people. 193 The abundance of intestinal flora is affected by hypoxia in the microenvironment. 194 Alterations in specific flora abundance might accelerate the formation and development of CRC. In addition, gut microbiota is involved in modulating immunity, transforming metabolome, and modifying the therapeutic effect. 154 , 195 Disturbance of the category and quantity of intestinal flora activates the NF‐κB pathway by stimulating intestinal epithelial cells that can trigger an inflammation stage. 193 Furthermore, chronic inflammation is the key element in inducing CRC formation. Several pro‐tumorigenic and anti‐tumorigenic bacterial species and their respective products of metabolism disturb major signaling pathways such as Wnt, PI3K‐Akt, MAPK, TGF‐β, EGFR, mTOR, and p53. 196 Intestinal microflora also participates in the formation of the TME by mediating angiogenesis. 197 In general, bacterial toxins are related to pro‐inflammatory processes, activation of angiogenesis, and cellular proliferation pathways. 198 Notably, among multitudinous intestinal microflora metabolites, bile acid can promote CRC progression through multiple mechanisms, including inhibiting apoptosis and enhancing cancer cell proliferation, invasion, and angiogenesis. 199 Some scholars have proposed the application of drugs for intestinal flora in combination with chemotherapy and immunotherapy to improve treatment response and tolerance. 198
4. TREATMENT STRATEGY
In recent decades, with the research progress on cancer etiology, treatment methods for CRC have been constantly updated and improved. Besides surgery, which is the most common treatment method, other approaches such as chemotherapy, targeted therapy, immunotherapy, and radiotherapy are used to improve therapeutic effectiveness and prognosis and extend the survival period and quality of life of patients with metastatic CRC (mCRC). For patients with neoadjuvant chemotherapy indications, 5‐fluorouracil (5‐FU), 5‐FU plus leucovorin (LV), single‐agent capecitabine, reduced‐dose capecitabine plus oxaliplatin (XELOX), or oxaliplatin plus 5‐FU plus LV (FOLFOX) are considered optimal treatment schemes. 200 On the basis of the staging of patients with CRC, postoperative adjuvant chemotherapy, including 5‐FU plus LV, capecitabine, mFOLFOX6, or XELOX, might be selected. Among these plans, combination with oxaliplatin has the best treatment effect and the greatest benefit for patients. 201 , 202 , 203 FOLFIRI (doublet cytotoxic combinations of fluorouracil, leucovorin, and irinotecan) and FOLFOX are the two first‐line treatment prescriptions for patients with mCRC, and they are presently recommended by European Society for Medical Oncology (ESMO) guidelines and the Pan‐Asia adaptation of the guidelines. 2 , 204
Clinical evidence has shown that combination with targeted therapy and chemotherapy significantly improves progression‐free survival (PFS) and overall survival (OS) compared to chemotherapy alone in patients with mCRC. 200 Among various aspects, angiogenesis has been validated to be a key element in the pathogenesis of malignancy, and it has provided biological insights and subsequent therapeutic options. 10 Regarding its central status in tumor angiogenesis, VEGFs and their receptors are the major targets in anti‐angiogenesis treatment. 205 Tumors highly dependent on VEGF‐induced angiogenesis, such as CRC, renal cell carcinoma, and neuroendocrine tumors, might have a relatively satisfactory response to anti‐VEGF drugs. 206 In addition, targeted drugs are recommended as first‐line treatment drugs for most patients, unless contraindications exist. 30
4.1. Targeting agents
Inhibition of angiogenesis by blocking VEGF is a major focus of targeted cancer therapy. The important functions of pro‐angiogenesis molecules, which have strong relations with tumor growth, invasion, and metastasis, make them ideal targets in suppressing tumors including CRC. 32 VEGF is one of the most decisive factors that promote angiogenesis. Several different strategies have been applied to block VEGF, such as neutralizing anti‐VEGF monoclonal antibodies, monoclonal antibodies that block VEGFRs, and small‐molecule tyrosine kinase inhibitors (TKIs) that block VEGFR activation and downstream signaling Figure 1. 207
Anti‐VEGF: Bevacizumab is an IgG1 humanized monoclonal antibody (MoAB) against VEGF‐A. Initially, it was recommended as a first‐line treatment because it showed good results in phases I and II clinical trials. Bevacizumab plus chemotherapy, compared with chemotherapy alone, showed advantages for PFS (10.6 months vs. 6.2 months, p < 0.001) and OS (20.3 months vs. 15.6 months, p < 0.001). 208 Nevertheless, bevacizumab combined with standard first‐line treatment did not show the expected advantages of treatment with ras mutation in a phase III clinical trial. 209 As a second‐line treatment, bevacizumab plus FOLFOX had a better outcome than FOLFOX alone: 7.3 months versus 4.7 months (p < 0.001) in PFS and 12.9 months versus 10.8 months (p = 0.0011) in OS. 210 Therefore, it was recommended that bevacizumab be combined with a chemotherapy regimen. In addition, other researchers have proposed that bevacizumab can eliminate RAS mutant clones to convert RAS gene mutant, which is more dependent on angiogenesis compared with wild‐type (WT) RAS genes. 211
Aflibecept: Aflibecept is a fusion protein targeting VEGF‐A, VEGF‐B, and PlGF. It might exhibit a more comprehensive inhibition effect on angiogenesis because of its multiple targets compared with bevacizumab or ramucirumab. 30 VELOUR results 212 revealed that the OS median survival was 13.50 months in the FOLFIRI/aflibercept group versus 12.06 months in the control group. Moreover, FOLFIRI/aflibercept showed a remarkable improvement in the median PFS of 6.90 months versus 4.67 in the placebo (p < 0.0001). The response rate (RR) was 19.8% in the aflibercept group but 11.1% in the placebo group (p < 0.0001). 213 Strongly supported by existing research results, aflibercept combined with FOLFIRI can be an option for patients with DR or progression after oxaliplatin‐containing treatment. Meanwhile, ESMO guidelines explicitly recommend aflibercept as an alternative second‐line treatment agent for RAS WT and RAS mutant patients. 30
Anti‐VEGF receptors: Ramucirumab is a humanized monoclonal antibody targeting the extracellular domain of VEGF receptor 2 (VEGFR2). It received US Food and Drug Administration (FDA) approval in 2015 for the second‐line treatment of mCRC 214 combined with FOLFIRI, based on the results of the RAISE trial. Ramucirumab is a newcomer among the antiangiogenic agents that can improve overall survival with a safe and manageable toxicity profile. 215 Fruquintinib (HMPL‐013), a long‐term small‐molecule, which can selectively inhibit VEGFR (VEGFR1, VEGFR2, and VEGFR3), has demonstrated several advantages, such as low off‐target toxicity, good drug tolerance, and a remarkable effect in clinical studies. Thus, it is recommended as a third‐line agent for the treatment of patients with CRC via other targeted therapy drugs. 216
Numerous small‐molecule TKIs exist. 217 Regorafenib is a multi‐targeting kinase inhibitor (TKI) approved for the treatment of patients with mCRC as a third‐line treatment for advanced CRC in comparison with standard chemotherapy. 218 This agent was originally developed as a RAF1 inhibitor. The dual blockade of VEGFRs and TIE2 can lead to accessional anti‐angiogenesis effects and the distinctive regulation of vessel stability. In addition, it is a TKI of the VEGF signaling pathways, 219 enabling its continuous antiangiogenic effect even in tumors resistant to VEGF inhibitors. Moreover, regorafenib has the important effect of enhancing anti‐tumor immunity via macrophage modulation. 220 Therefore, preliminary evidence suggested that this multi‐kinase inhibitor might be an optimal combination partner for immune checkpoint inhibitors (ICIs). 221
However, prolonged VEGF blockade enhances tumor hypoxia, causes resistance to hypoxia‐induced apoptosis, and increases VEGF expression, thereby promoting tumor aggressiveness. 222 , 223 Aside from VEGFR, EGFR is another commonly used target in treating the anti‐angiogenesis of CRC. 224 The treatment strategy often has two directions: monoclonal antibodies that block EFGR and inhibitors targeting intracellular tyrosine kinase. As a chimeric IgG antibody, cetuximab leads to the internalization and degradation of EGFR after binding to its external domain. 225 However, immunity reactions might occur as cetuximab is a murine‐human chimeric antibody. Panitumumab is the perfect solution to this issue; it is an antibody that is fully humanized and does not induce cytotoxicity mediated by antibody‐dependent cells. 226 Cetuximab and panitumumab are approved as first‐line treatment agents for CRC by the FDA. However, anti‐EGFR drugs are not recommended for priority use in second‐ or third‐line treatment of CRC because they do not show good statistics in PFS or OS. 227 Approximately 45% of colon cancer cells present RAS mutations. 228 In patients with mCRC, the proportions of KRAS, NRAS, and HRAS mutations are 40%–50%, 2%–9%, and 1%–2%, respectively. 229 The KRAS proto‐oncogene encodes a GTPase protein (KRAS) that is crucial in copious molecular pathways including the EGFR pathway. 230 With high‐frequency occurrence in CRC KRAS mutations, KRAS G12V is related to multiple aspects of tumor clinical pathology, such as invasion and poor prognosis. Moreover, it is linked to undesirable therapeutic effects of anti‐EGFR agents. There are interactions between KRAS G12V and HIF‐1α. Furthermore, KRAS G12V promotes the expression of HIF‐1α, whereas overexpressed hypoxia or HIF‐1α activates KRAS G12V. Only patients with WT RAS tumors receive a clinical benefit from anti‐EGFR antibody therapy. 231 Hypoxia is related to anti‐EGFR therapy resistance. Thus, adding HIF‐1α inhibitor PX‐478 to it might achieve a good therapeutic effect. 232 Moreover, KRAS mutations can represent the response to EGFR inhibitors as a negative predictive factor. 231 With the introduction of anti‐EGFR in the treatment of RAS WT mCRC, the optimal sequencing between anti‐VEGFs and anti‐EGFRs in this population of patients has been a matter of intense debate. For example, data from Francesca showed that using anti‐EGFRs in the first‐line setting for the right CRC (RC) should be avoided when other therapeutic alternatives are available. 233 Meanwhile, for RAS WT metastatic CRC patients with left‐side colon tumors, chemotherapy plus anti‐EGFR agents are recommended as first‐line treatment. 43
Ingredients from traditional Chinese medicine, such as several phenolic compounds (e.g., flavones, phenolcarboxylic acids, and ellagitannins), 234 hyperforin, 235 Raddeanin A, 236 and matrine 237 suppress CRC by inhibiting angiogenesis and other mechanisms. However, the details are not clearly explained. These promising pharmaceutical ingredients have not been approved as standard treatment drugs, which might be used as an adjuvant treatment.
4.2. Drug resistance
Drug resistance cannot be ignored in the treatment of CRC. Data from Bardelli et al. reported that DR occurs in approximately 80% of cases during treatment. 238 The mechanisms of DR are divided into the following three aspects: transformation in VEGF dependence, alternative pathways, and stromal cell interactions. 32 As described, anti‐angiogenic therapies might cause hypoxia and increase HIF‐1a expression, which are known as drivers of EMT. Moreover, HIF‐1 has multiple functions to promote cancer cell survival in the hypoxic environment. Resistance to anti‐angiogenesis agents involves several different but related mechanisms: recruiting various BMDCs, which differentiate into ECs, pericytes, and pro‐angiogenic monocytes, such as TAM; enhancing and increasing pericyte coverage, which safeguards tumor blood vessels; and increasing invasiveness of tumor cells, thereby leading to vessel co‐option. 28 , 239 , 240
Compensatory pathways in angiogenesis are nonnegligible in DR. TGF‐β, FGF 2, PDGF, Ang‐2, and IL‐1 are assumed to be highly relevant to anti‐VEGF resistance in cancer. 241 Alternative angiogenic factors and their pathways are shown in Table 2. Others involve EGF, G‐CSF, PlGF, HGF, stromal cell‐derived factor‐1, and IGF. 242 The table below shows that Ras/Raf/MEK/ERK and PI3K/Akt are the major downstream signaling pathways. Interestingly, these two pathways are also the dominant cascades of EGFR activation, 199 which indirectly validates the crosstalk between VEGF and EFGR. Other mechanisms might include EGF overexpression, EGFR alteration, RAS/RAF/PI3K gene mutations, ERBB2/MET/IGF‐1R activation, metabolic remodeling, MSI, and autophagy. 243
TABLE 2.
Alternative angiogenic factors and their pathways 241
| Ligands→ | Ang‐2 | Bv8 | bFGF | IL‐1 | PDGF | PlGF | TGF ‐β1 |
|---|---|---|---|---|---|---|---|
| Receptors→ | Tie2 | PROKR2 | FGFR | IL‐1RI, IL‐1RAcP | PDGFR | VEGFR1, NRP1/2 | TGF‐βRII, TGF ‐βRI |
| Pathways↓ | |||||||
| Ras/Raf/MEK/ERK | ▲ | ▲ | ▲ | ||||
| PI3K/Akt | ▲ | ▲ | ▲ | ▲ | |||
| JAK/STAT | ▲ | ||||||
| PLCγ | ▲ | ▲ | |||||
| NF‐κB | ▲ | ||||||
| JNK | ▲ | ||||||
| P38/MAPK | ▲ | ▲ | |||||
| Smad | ▲ |
Note: ▲: regulating downstream pathways.
Abbreviation: VEGF, vascular endothelial growth factor.
Molecular and biochemical mechanisms are related to the phenotypic changes that support carcinogenesis, including apoptosis inhibition, reinforced tumor cell proliferation, increased invasiveness, cell adhesion perturbations, angiogenesis promotion, and immune surveillance inhibition. 231 Tumor immune escape implies that tumor cells escape from immune surveillance and inhibit the immune response of the host. 244 Tumor cells have developed several mechanisms to avoid detection by immune cells. Secretion of soluble immunosuppressive factors, such as TGF‐β and IL‐10, or downregulation of major histocompatibility complex (MHC 1) expression might all be related to immune escape. 245 IC modulation is another well‐known mechanism by which tumor cells suppress the local immune response. IC receptors, including programmed death 1 (PD‐1), cytotoxic T lymphocyte antigen 4 (CTLA‐4), and CD279, contribute to inactivating and exhausting tumor‐related T cells. 246
ICIs represent a new time for cancer treatment. They selectively combine with immunosuppressive molecules on the surface of immunocytes (e.g., CTLA‐4) or on tumor cells, such as PD‐1 or their ligand (PD‐L1), to block tumor cells’ immune escape. 247 , 248 Ipilimumab, pembrolizumab, and nivolumab are three types of ICIs approved by the FDA in 2015 for CRC treatment in patients with mismatch repair defects or microsatellite instability. 221 , 249 Nivolumab and pembrolizumab are anti‐PD1 drugs that competitively bind with PD‐1, thereby blocking tumor immune evasion mediated by the combination of PD‐1 and its ligands, PD‐L1 or PD‐L2. By contrast, ipilimumab inhibits the combination of CTLA‐4 and a cluster of differentiation 80/86 (CD80/CD86), which attenuates T‐cell activation. Nevertheless, the application of ICIs remains limited because of the lack of sensitive markers and inevitable DR. 154
5. DISCUSSION
To date, although anti‐angiogenesis agents, with their highlighted advantages such as improving oxygen levels and drug delivery through vascular normalization, carry weight in the treatment of CRC, their therapeutic efficacy remains far from satisfactory. However, ensuring that every patient receiving treatment achieves satisfactory results in one unified plan is never an easy task. With the increase in knowledge about CRC, novel targets have been identified. Nevertheless, therapy resistance and unresponsiveness to immunotherapy remain major treatment obstacles. However, the treatment scheme selection based on biomarkers for patients with CRC remains limited because of the incomplete accuracy of conventional biomarkers in diagnosis, prediction, and prognosis. Thus, further studies are necessary to develop clinically applicable biomarkers. However, developing appropriate therapeutic programs to increase ICI activity and efficacy through the regulation of gut microbiota in patients with CRC is another clinical challenge. Artificial intelligence shows promise and will takes us into a new era to persistently improve molecular prediction algorithms from the sea of usable data. It might help us explore new methods to overcome the predicaments of the current antitumor strategy.
AUTHOR CONTRIBUTIONS
Zhenni Yang: Writing – original draft; writing – review and editing. XuQian Zhang: Writing – review and editing. Xiaozhe Bai: Writing – review and editing. Xiaonan Xi: Supervision. Wentian Liu: Supervision. Weilong Zhong: Formal analysis; project administration; supervision.
FUNDING INFORMATION
The authors would like to thank the National Natural Science Foundation of China (grant nos. 82000511 and 82303509), the Scientific and Technological Projects of Tianjin (21JCQNJC01120), the Health Science and Technology Project of Tianjin (TJWJ2021QN006), the Scientific Research Project of Tianjin Education Commission (2019KJ197), and the National Key Research and Development Program (2022YFC2504004) for financially supporting this study.
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no competing interests.
ETHICS STATEMENT
Approval of the research protocol by an Institutional Review Board: If not applicable.
Informed Consent: If not applicable.
Registry and the Registration No. of the study/trial: If not applicable.
Animal Studies: If not applicable.
ACKNOWLEDGMENTS
Not applicable.
Yang Z, Zhang X, Bai X, Xi X, Liu W, Zhong W. Anti‐angiogenesis in colorectal cancer therapy. Cancer Sci. 2024;115:734‐751. doi: 10.1111/cas.16063
Zhenni Yang and Xuqian Zhang contributed equally to this work.
Contributor Information
Xiaonan Xi, Email: 9820210009@nankai.edu.cn.
Wentian Liu, Email: tliu01@tmu.edu.cn.
Weilong Zhong, Email: zhongweilong@tmu.edu.cn.
DATA AVAILABILITY STATEMENT
No new data has been used or generated.
REFERENCES
- 1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. doi: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
- 2. Van Cutsem E, Cervantes A, Adam R, et al. ESMO consensus guidelines for the Management of Patients with metastatic colorectal cancer. Ann Oncol. 2016;27(8):1386‐1422. doi: 10.1093/annonc/mdw235 [DOI] [PubMed] [Google Scholar]
- 3. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285(21):1182‐1186. doi: 10.1056/NEJM197111182852108 [DOI] [PubMed] [Google Scholar]
- 4. Otrock ZK, Mahfouz RAR, Makarem JA, Shamseddine AI. Understanding the biology of angiogenesis: review of the Most important molecular mechanisms. Blood Cells Mol Dis. 2007;39(2):212‐220. doi: 10.1016/j.bcmd.2007.04.001 [DOI] [PubMed] [Google Scholar]
- 5. Peng Y, Wang Y, Zhou C, Mei W, Zeng C. PI3K/Akt/mTOR pathway and its role in cancer therapeutics: are we making headway? Front Oncol. 2022;12:819128. doi: 10.3389/fonc.2022.819128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Angelucci A, Delle Monache S, Cortellini A, Di Padova M, Ficorella C. “Vessels in the storm”: searching for prognostic and predictive Angiogenic factors in colorectal cancer. Int J Mol Sci. 2018;19(1):299. doi: 10.3390/ijms19010299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Daly ME, Makris A, Reed M, Lewis CE. Hemostatic regulators of tumor angiogenesis: a source of antiangiogenic agents for cancer treatment? J Natl Cancer Inst. 2003;95(22):1660‐1673. doi: 10.1093/jnci/djg101 [DOI] [PubMed] [Google Scholar]
- 8. Kazerounian S, Lawler J. Integration of pro‐ and anti‐Angiogenic signals by endothelial cells. J Cell Commun Signal. 2018;12(1):171‐179. doi: 10.1007/s12079-017-0433-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Nishida N, Yano H, Nishida T, Kamura T, Kojiro M. Angiogenesis in cancer. Vasc Health Risk Manag. 2006;2(3):213‐219. doi: 10.2147/vhrm.2006.2.3.213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Prager GW, Poettler M. Angiogenesis in cancer. Basic mechanisms and therapeutic advances. Hamostaseologie. 2012;32(2):105‐114. doi: 10.5482/ha-1163 [DOI] [PubMed] [Google Scholar]
- 11. El‐Kenawi AE, El‐Remessy AB. Angiogenesis inhibitors in cancer therapy: mechanistic perspective on classification and treatment rationales. Br J Pharmacol. 2013;170(4):712‐729. doi: 10.1111/bph.12344 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Baluk P, Morikawa S, Haskell A, Mancuso M, McDonald DM. Abnormalities of basement membrane on blood vessels and endothelial sprouts in tumors. Am J Pathol. 2003;163(5):1801‐1815. doi: 10.1016/S0002-9440(10)63540-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Phung TL, Ziv K, Dabydeen D, et al. Pathological angiogenesis is induced by sustained Akt signaling and inhibited by rapamycin. Cancer Cell. 2006;10(2):159‐170. doi: 10.1016/j.ccr.2006.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Maishi N, Hida K. Tumor endothelial cells accelerate tumor metastasis. Cancer Sci. 2017;108(10):1921‐1926. doi: 10.1111/cas.13336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. St Croix B, Rago C, Velculescu V, et al. Genes expressed in human tumor endothelium. Science. 2000;289(5482):1197‐1202. doi: 10.1126/science.289.5482.1197 [DOI] [PubMed] [Google Scholar]
- 16. Staton CA, Chetwood ASA, Cameron IC, Cross SS, Brown NJ, Reed MWR. The Angiogenic switch occurs at the adenoma stage of the adenoma carcinoma sequence in colorectal cancer. Gut. 2007;56(10):1426‐1432. doi: 10.1136/gut.2007.125286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Zhao J, Li L, Han ZY, Wang ZX, Qin LX. Long noncoding RNAs, emerging and versatile regulators of tumor‐induced angiogenesis. Am J Cancer Res. 2019;9(7):1367‐1381. doi: 10.2217/fon-2017-0149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Rafii DC, Psaila B, Butler J, Jin DK, Lyden D. Regulation of Vasculogenesis by platelet‐mediated recruitment of bone marrow‐derived cells. Arterioscler Thromb Vasc Biol. 2008;28(2):217‐222. doi: 10.1161/ATVBAHA.107.151159 [DOI] [PubMed] [Google Scholar]
- 19. Haas G, Fan S, Ghadimi M, De Oliveira T, Conradi LC. Different forms of tumor vascularization and their clinical implications focusing on vessel Co‐option in colorectal cancer liver metastases. Front Cell Dev Biol. 2021;9:612774. doi: 10.3389/fcell.2021.612774 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Luo Q, Wang J, Zhao W, et al. Vasculogenic mimicry in carcinogenesis and clinical applications. J Hematol Oncol. 2020;13(1):19. doi: 10.1186/s13045-020-00858-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Melincovici CS, Boşca AB, Şuşman S, et al. Vascular endothelial growth factor (VEGF) – key factor in Normal and pathological angiogenesis. Rom J Morphol Embryol. 2018;59(2):455‐467. [PubMed] [Google Scholar]
- 22. Patan S, Alvarez MJ, Schittny JC, Burri PH. Intussusceptive microvascular growth: a common alternative to capillary sprouting. Arch Histol Cytol. 1992;55(Suppl):65‐75. doi: 10.1679/aohc.55.suppl_65 [DOI] [PubMed] [Google Scholar]
- 23. Karthik S, Djukic T, Kim JD, et al. Synergistic interaction of sprouting and Intussusceptive angiogenesis during zebrafish caudal vein plexus development. Sci Rep. 2018;8(1):9840. doi: 10.1038/s41598-018-27791-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Ge H, Luo H. Overview of advances in Vasculogenic mimicry – a potential target for tumor therapy. Cancer Manag Res. 2018;10:2429‐2437. doi: 10.2147/CMAR.S164675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Lizárraga‐Verdugo E, Avendaño‐Félix M, Bermúdez M, Ramos‐Payán R, Pérez‐Plasencia C, Aguilar‐Medina M. Cancer stem cells and its role in angiogenesis and vasculogenic mimicry in gastrointestinal cancers. Front Oncol. 2020;10:413. doi: 10.3389/fonc.2020.00413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Yang JP, Liao YD, Mai DM, et al. Tumor vasculogenic mimicry predicts poor prognosis in cancer patients: a meta‐analysis. Angiogenesis. 2016;19(2):191‐200. doi: 10.1007/s10456-016-9500-2 [DOI] [PubMed] [Google Scholar]
- 27. van Dam PJ, van der Stok EP, Teuwen LA, et al. International consensus guidelines for scoring the histopathological growth patterns of liver metastasis. Br J Cancer. 2017;117(10):1427‐1441. doi: 10.1038/bjc.2017.334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Frentzas S, Simoneau E, Bridgeman VL, et al. Vessel co‐option mediates resistance to anti‐Angiogenic therapy in liver metastases. Nat Med. 2016;22(11):1294‐1302. doi: 10.1038/nm.4197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Coelho AL, Gomes MP, Catarino RJ, et al. Angiogenesis in NSCLC: is vessel co‐option the trunk that sustains the branches? Oncotarget. 2017;8(24):39795‐39804. doi: 10.18632/oncotarget.7794 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. González Astorga B, Salvà Ballabrera F, Aranda Aguilar E, et al. Patient profiles as an aim to optimize selection in the second line setting: the role of Aflibercept. Clin Transl Oncol. 2021;23(8):1520‐1528. doi: 10.1007/s12094-021-02568-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Wiśniewska W, Kopka M, Siemiątkowska K, Fudalej MM, Sobiborowicz A, Badowska‐Kozakiewicz AM. The complexity of tumour angiogenesis based on recently described molecules. Contemp Oncol. 2021;25(1):33‐44. doi: 10.5114/wo.2021.105075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Haibe Y, Kreidieh M, El Hajj H, et al. Resistance mechanisms to anti‐Angiogenic therapies in cancer. Front Oncol. 2020;10:221. doi: 10.3389/fonc.2020.00221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Coşkun Ö, Öztopuz Ö, Özkan ÖF. Determination of IL‐6, TNF‐α and VEGF levels in the serums of patients with colorectal cancer. Cell Mol Biol (Noisy‐le‐Grand). 2017;63(5):97‐101. doi: 10.14715/cmb/2017.63.5.18 [DOI] [PubMed] [Google Scholar]
- 34. Apte RS, Chen DS, Ferrara N. VEGF in signaling and disease: beyond discovery and development. Cell. 2019;176(6):1248‐1264. doi: 10.1016/j.cell.2019.01.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Otrock ZK, Makarem JA, Shamseddine AI. Vascular endothelial growth factor family of ligands and receptors: review. Blood Cells Mol Dis. 2007;38(3):258‐268. doi: 10.1016/j.bcmd.2006.12.003 [DOI] [PubMed] [Google Scholar]
- 36. Sharma T, Dhingra R, Singh S, et al. Aflibercept: a novel VEGF targeted agent to explore the future perspectives of anti‐Angiogenic therapy for the treatment of multiple tumors. Mini Rev Med Chem. 2013;13(4):530‐540. doi: 10.2174/1389557511313040006 [DOI] [PubMed] [Google Scholar]
- 37. Weis SM, Cheresh DA. Pathophysiological consequences of VEGF‐induced vascular permeability. Nature. 2005;437(7058):497‐504. doi: 10.1038/nature03987 [DOI] [PubMed] [Google Scholar]
- 38. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9(6):669‐676. doi: 10.1038/nm0603-669 [DOI] [PubMed] [Google Scholar]
- 39. Lee Y‐J, Karl DL, Maduekwe UN, et al. Differential effects of VEGFR‐1 and VEGFR‐2 inhibition on tumor metastases based on host organ environment. Cancer Res. 2010;70(21):8357‐8367. doi: 10.1158/0008-5472.CAN-10-1138 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 40. Neufeld G, Cohen T, Gitay‐Goren H, et al. Similarities and differences between the vascular endothelial growth factor (VEGF) splice variants. Cancer Metastasis Rev. 1996;15(2):153‐158. doi: 10.1007/BF00437467 [DOI] [PubMed] [Google Scholar]
- 41. Dakowicz D, Zajkowska M, Mroczko B. Relationship between VEGF family members, their receptors and cell death in the neoplastic transformation of colorectal cancer. Int J Mol Sci. 2022;23(6):3375. doi: 10.3390/ijms23063375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Rmali KA, Puntis MCA, Jiang WG. Tumour‐associated angiogenesis in human colorectal cancer. Colorect Dis. 2007;9(1):3‐14. doi: 10.1111/j.1463-1318.2006.01089.x [DOI] [PubMed] [Google Scholar]
- 43. Xie YH, Chen YX, Fang JY. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct Target Ther. 2020;5(1):22. doi: 10.1038/s41392-020-0116-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Canavese M, Ngo DTM, Maddern GJ, Hardingham JE, Price TJ, Hauben E. Biology and therapeutic implications of VEGF‐A splice isoforms and single‐nucleotide polymorphisms in colorectal cancer. Int J Cancer. 2017;140(10):2183‐2191. doi: 10.1002/ijc.30567 [DOI] [PubMed] [Google Scholar]
- 45. Mohamed SY, Mohammed HL, Ibrahim HM, Mohamed EM, Salah M. Role of VEGF, CD105, and CD31 in the prognosis of colorectal cancer cases. J Gastrointest Cancer. 2019;50(1):23‐34. doi: 10.1007/s12029-017-0014-y [DOI] [PubMed] [Google Scholar]
- 46. Chen X, Xu X, Pan B, et al. miR‐150‐5p suppresses tumor progression by targeting VEGFA in colorectal cancer. Aging (Albany NY). 2018;10(11):3421‐3437. doi: 10.18632/aging.101656 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26(5):605‐622. doi: 10.1016/j.ccell.2014.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Khan K, Valeri N, Dearman C, et al. Targeting EGFR pathway in metastatic colorectal cancer‐ tumour Heterogeniety and convergent evolution. Crit Rev Oncol Hematol. 2019;143:153‐163. doi: 10.1016/j.critrevonc.2019.09.001 [DOI] [PubMed] [Google Scholar]
- 49. Ah G, Ws S. Epidermal growth factor receptor pathway mutations and colorectal cancer therapy. Arch Pathol Lab Med. 2011;135:127‐132. doi: 10.5858/arpa.2011-0047-RA [DOI] [PubMed] [Google Scholar]
- 50. Roskoski R. Small molecule inhibitors targeting the EGFR/ErbB family of protein‐tyrosine kinases in human cancers. Pharmacol Res. 2019;139:395‐411. doi: 10.1016/j.phrs.2018.11.014 [DOI] [PubMed] [Google Scholar]
- 51. Daveri E, Adamo AM, Alfine E, Zhu W, Oteiza PI. Hexameric procyanidins inhibit colorectal cancer cell growth through both redox and non‐redox regulation of the epidermal growth factor signaling pathway. Redox Biol. 2021;38:101830. doi: 10.1016/j.redox.2020.101830 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Custodio A, Feliu J. Prognostic and predictive biomarkers for epidermal growth factor receptor‐targeted therapy in colorectal cancer: beyond KRAS mutations. Crit Rev Oncol Hematol. 2013;85(1):45‐81. doi: 10.1016/j.critrevonc.2012.05.001 [DOI] [PubMed] [Google Scholar]
- 53. Arteaga CL, Engelman JA. ERBB receptors: from oncogene discovery to basic science to mechanism‐based cancer therapeutics. Cancer Cell. 2014;25(3):282‐303. doi: 10.1016/j.ccr.2014.02.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Hynes NE, Lane HA. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer. 2005;5(5):341‐354. doi: 10.1038/nrc1609 [DOI] [PubMed] [Google Scholar]
- 55. Ivanova M, Venetis K, Guerini‐Rocco E, et al. HER2 in metastatic colorectal cancer: pathology, somatic alterations, and perspectives for novel therapeutic schemes. Life (Basel). 2022;12(9):1403. doi: 10.3390/life12091403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Siena S, Sartore‐Bianchi A, Marsoni S, et al. Targeting the human epidermal growth factor receptor 2 (HER2) oncogene in colorectal cancer. Ann Oncol. 2018;29(5):1108‐1119. doi: 10.1093/annonc/mdy100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Lawan AI, Ogunbiyi JO. Epidermal growth factor receptor expression of colorectal carcinoma in Nigerian patients. West Afr J Med. 2020;37(2):100‐105. [PubMed] [Google Scholar]
- 58. Chung SY, Chao TC, Su Y. The Stemness‐high human colorectal cancer cells promote angiogenesis by producing higher amounts of Angiogenic cytokines via activation of the Egfr/Akt/Nf‐κB pathway. Int J Mol Sci. 2021;22(3):1355. doi: 10.3390/ijms22031355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Mehta VB, Besner GE. HB‐EGF promotes angiogenesis in endothelial cells via PI3‐kinase and MAPK signaling pathways. Growth Factors. 2007;25(4):253‐263. doi: 10.1080/08977190701773070 [DOI] [PubMed] [Google Scholar]
- 60. Seghezzi G, Patel S, Ren CJ, et al. Fibroblast growth factor‐2 (FGF‐2) induces vascular endothelial growth factor (VEGF) expression in the endothelial cells of forming capillaries: an autocrine mechanism contributing to angiogenesis. J Cell Biol. 1998;141(7):1659‐1673. doi: 10.1083/jcb.141.7.1659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Matsuda Y, Ueda J, Ishiwata T. Fibroblast growth factor receptor 2: expression, roles, and potential as a novel molecular target for colorectal cancer. Patholog Res Int. 2012;2012:574768. doi: 10.1155/2012/574768 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Liu G, Chen T, Ding Z, Wang Y, Wei Y, Wei X. Inhibition of FGF‐FGFR and VEGF‐VEGFR signalling in cancer treatment. Cell Prolif. 2021;54(4):e13009. doi: 10.1111/cpr.13009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Knuchel S, Anderle P, Werfelli P, Diamantis E, Rüegg C. Fibroblast surface‐associated FGF‐2 promotes contact‐dependent colorectal cancer cell migration and invasion through FGFR‐SRC signaling and integrin Αvβ5‐mediated adhesion. Oncotarget. 2015;6(16):14300‐14317. doi: 10.18632/oncotarget.3883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Heldin CH, Westermark B. Mechanism of action and in vivo role of platelet‐derived growth factor. Physiol Rev. 1999;79(4):1283‐1316. doi: 10.1152/physrev.1999.79.4.1283 [DOI] [PubMed] [Google Scholar]
- 65. Betsholtz C. Insight into the physiological functions of PDGF through genetic studies in mice. Cytokine Growth Factor Rev. 2004;15(4):215‐228. doi: 10.1016/j.cytogfr.2004.03.005 [DOI] [PubMed] [Google Scholar]
- 66. Zou X, Tang XY, Qu Z, et al. Targeting the PDGF/PDGFR signaling pathway for cancer therapy: a review. Int J Biol Macromol. 2022;202:539‐557. doi: 10.1016/j.ijbiomac.2022.01.113 [DOI] [PubMed] [Google Scholar]
- 67. Chater C, Bauters A, Beugnet C, M'Ba L, Rogosnitzky M, Zerbib P. Intraplatelet vascular endothelial growth factor and platelet‐derived growth factor: new biomarkers in carcinoembryonic antigen‐negative colorectal cancer? Gastrointest Tumors. 2018;5(1–2):32‐37. doi: 10.1159/000486894 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Manzat Saplacan RM, Balacescu L, Gherman C, et al. The role of PDGFs and PDGFRs in colorectal cancer. Mediators Inflamm. 2017;2017:4708076. doi: 10.1155/2017/4708076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Andrae J, Gallini R, Betsholtz C. Role of platelet‐derived growth factors in physiology and medicine. Genes Dev. 2008;22(10):1276‐1312. doi: 10.1101/gad.1653708 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Parikh SM. The angiopoietins and Tie2 in vascular inflammation. Curr Opin Hematol. 2017;24(5):432‐438. doi: 10.1097/MOH.0000000000000361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Jones N, Master Z, Jones J, et al. Identification of Tek/Tie2 binding partners. Binding to a multifunctional docking site mediates cell survival and migration. J Biol Chem. 1999;274(43):30896‐30905. doi: 10.1074/jbc.274.43.30896 [DOI] [PubMed] [Google Scholar]
- 72. Jones N, Chen SH, Sturk C, et al. A unique autophosphorylation site on Tie2/Tek mediates Dok‐R Phosphotyrosine binding domain binding and function. Mol Cell Biol. 2003;23(8):2658‐2668. doi: 10.1128/MCB.23.8.2658-2668.2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Suri C, Jones PF, Patan S, et al. Requisite role of Angiopoietin‐1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell. 1996;87(7):1171‐1180. doi: 10.1016/S0092-8674(00)81813-9 [DOI] [PubMed] [Google Scholar]
- 74. Jary M, Hasanova R, Vienot A, et al. Molecular description of ANGPT2 associated colorectal carcinoma. Int J Cancer. 2020;147(7):2007‐2018. doi: 10.1002/ijc.32993 [DOI] [PubMed] [Google Scholar]
- 75. Fiedler U, Krissl T, Koidl S, et al. Angiopoietin‐1 and Angiopoietin‐2 share the same binding domains in the Tie‐2 receptor involving the first Ig‐like loop and the epidermal growth factor‐like repeats. J Biol Chem. 2003;278(3):1721‐1727. doi: 10.1074/jbc.M208550200 [DOI] [PubMed] [Google Scholar]
- 76. Ahmad SA, Liu W, Jung YD, et al. Differential expression of Angiopoietin‐1 and Angiopoietin‐2 in colon carcinoma. A possible mechanism for the initiation of angiogenesis. Cancer. 2001;92(5):1138‐1143. doi: 10.1002/1097-0142(20010901)92 [DOI] [PubMed] [Google Scholar]
- 77. Kim H, Ahn TS, Kim CJ, et al. Oncogenic function of Angiopoietin‐2 in vitro and its modulation of tumor progression in colorectal carcinoma. Oncol Lett. 2017;14(1):553‐560. doi: 10.3892/ol.2017.6203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Gu J, Yamamoto H, Ogawa M, et al. Hypoxia‐induced up‐regulation of Angiopoietin‐2 in colorectal cancer. Oncol Rep. 2006;15(4):779‐783. doi: 10.3892/or.15.4.779 [DOI] [PubMed] [Google Scholar]
- 79. Narayanankutty A. PI3K/ Akt/ mTOR pathway as a therapeutic target for colorectal cancer: a review of preclinical and clinical evidence. Curr Drug Targets. 2019;20(12):1217‐1226. doi: 10.2174/1389450120666190618123846 [DOI] [PubMed] [Google Scholar]
- 80. Zhong J, Ding S, Zhang X, et al. To investigate the occurrence and development of colorectal cancer based on the PI3K/AKT/mTOR signaling pathway. FBL. 2023;28(2):37. doi: 10.31083/j.fbl2802037 [DOI] [PubMed] [Google Scholar]
- 81. Coutte L, Dreyer C, Sablin M‐P, Faivre S, Raymond E. PI3K‐AKT‐mTOR pathway and cancer. Bull Cancer. 2012;99(2):173‐180. doi: 10.1684/bdc.2011.1384 [DOI] [PubMed] [Google Scholar]
- 82. Qazi AK, Hussain A, Hamid A, et al. Recent development in targeting PI3K‐Akt‐mTOR signaling for anticancer therapeutic strategies. Anticancer Agents Med Chem. 2013;13(10):1552‐1564. doi: 10.2174/1871520613666131125123241 [DOI] [PubMed] [Google Scholar]
- 83. Shaw RJ, Bardeesy N, Manning BD, et al. The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell. 2004;6(1):91‐99. doi: 10.1016/j.ccr.2004.06.007 [DOI] [PubMed] [Google Scholar]
- 84. Karar J, Maity A. PI3K/AKT/mTOR pathway in angiogenesis. Front Mol Neurosci. 2011;4:51. doi: 10.3389/fnmol.2011.00051 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Fang JY, Richardson BC. The MAPK Signalling pathways and colorectal cancer. Lancet Oncol. 2005;6(5):322‐327. doi: 10.1016/S1470-2045(05)70168-6 [DOI] [PubMed] [Google Scholar]
- 86. Aggarwal V, Tuli HS, Varol A, et al. Role of reactive oxygen species in cancer progression: molecular mechanisms and recent advancements. Biomolecules. 2019;9(11):735. doi: 10.3390/biom9110735 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Morikawa M, Derynck R, Miyazono K. TGF‐β and the TGF‐β family: context‐dependent roles in cell and tissue physiology. Cold Spring Harb Perspect Biol. 2016;8(5):a021873. doi: 10.1101/cshperspect.a021873 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Attisano L, Wrana JL. Signal transduction by the TGF‐Beta superfamily. Science. 2002;296(5573):1646‐1647. doi: 10.1126/science.1071809 [DOI] [PubMed] [Google Scholar]
- 89. Papageorgis P, Cheng K, Ozturk S, et al. Smad4 inactivation promotes malignancy and drug resistance of colon cancer. Cancer Res. 2011;71(3):998‐1008. doi: 10.1158/0008-5472.CAN-09-3269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Jung B, Staudacher JJ, Beauchamp D. Transforming growth factor β superfamily signaling in development of colorectal cancer. Gastroenterology. 2017;152(1):36‐52. doi: 10.1053/j.gastro.2016.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Jiang Z, Cao Q, Dai G, et al. Celastrol inhibits colorectal cancer through TGF‐Β1/Smad signaling. Onco Targets Ther. 2019;12:509‐518. doi: 10.2147/OTT.S187817 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Xu Y, Pasche B. TGF‐Beta Signaling Alterations and Susceptibility to Colorectal Cancer. Hum Mol Genet. 2007;16:R14‐20. doi: 10.1093/hmg/ddl486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Guinney J, Dienstmann R, Wang X, et al. The consensus molecular subtypes of colorectal cancer. Nat Med. 2015;21(11):1350‐1356. doi: 10.1038/nm.3967 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. de Miranda NFCC, van Dinther M, van den Akker BEWM, van Wezel T, ten Dijke P, Morreau H. Transforming growth factor β signaling in colorectal cancer cells with microsatellite instability despite Biallelic mutations in TGFBR2. Gastroenterology. 2015;148(7):1427‐1437.e8. doi: 10.1053/j.gastro.2015.02.052 [DOI] [PubMed] [Google Scholar]
- 95. Itatani Y, Kawada K, Sakai Y. Transforming growth factor‐β signaling pathway in colorectal cancer and its tumor microenvironment. Int J Mol Sci. 2019;20(23):5822. doi: 10.3390/ijms20235822 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. 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. doi: 10.1016/j.ccr.2012.08.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Napetschnig J, Wu H. Molecular basis of NF‐κB signaling. Annu Rev Biophys. 2013;42:443‐468. doi: 10.1146/annurev-biophys-083012-130338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Israël A. The IKK complex, a central regulator of NF‐kappaB activation. Cold Spring Harb Perspect Biol. 2010;2(3):a000158. doi: 10.1101/cshperspect.a000158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Patel M, Horgan PG, McMillan DC, Edwards J. NF‐κB pathways in the development and progression of colorectal cancer. Transl Res. 2018;197:43‐56. doi: 10.1016/j.trsl.2018.02.002 [DOI] [PubMed] [Google Scholar]
- 100. Cao M, Wang Y, Lu G, et al. Classical Angiogenic signaling pathways and novel anti‐Angiogenic strategies for colorectal cancer. Curr Issues Mol Biol. 2022;44(10):4447‐4471. doi: 10.3390/cimb44100305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Yu HG, Zhong X, Yang YN, et al. Increased expression of nuclear factor‐kappaB/RelA is correlated with tumor angiogenesis in human colorectal cancer. Int J Colorectal Dis. 2004;19(1):18‐22. doi: 10.1007/s00384-003-0494-z [DOI] [PubMed] [Google Scholar]
- 102. Wang R, Ma Y, Zhan S, et al. B7‐H3 promotes colorectal cancer angiogenesis through activating the NF‐κB pathway to induce VEGFA expression. Cell Death Dis. 2020;11(1):55. doi: 10.1038/s41419-020-2252-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Ni L, Dong C. New B7 family checkpoints in human cancers. Mol Cancer Ther. 2017;16(7):1203‐1211. doi: 10.1158/1535-7163.MCT-16-0761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Son Y, Kwon SM, Cho JY. CD276 (B7‐H3) maintains proliferation and regulates differentiation in Angiogenic function in late endothelial progenitor cells. Stem Cells. 2019;37(3):382‐394. doi: 10.1002/stem.2944 [DOI] [PubMed] [Google Scholar]
- 105. Han S, Wang Y, Shi X, et al. Negative roles of B7‐H3 and B7‐H4 in the microenvironment of cervical cancer. Exp Cell Res. 2018;371(1):222‐230. doi: 10.1016/j.yexcr.2018.08.014 [DOI] [PubMed] [Google Scholar]
- 106. Freise N, Burghard A, Ortkras T, et al. Signaling mechanisms inducing Hyporesponsiveness of phagocytes during systemic inflammation. Blood. 2019;134(2):134‐146. doi: 10.1182/blood.2019000320 [DOI] [PubMed] [Google Scholar]
- 107. Haan C, Kreis S, Margue C, Behrmann I. Jaks and cytokine receptors–an intimate relationship. Biochem Pharmacol. 2006;72(11):1538‐1546. doi: 10.1016/j.bcp.2006.04.013 [DOI] [PubMed] [Google Scholar]
- 108. Fan Y, Mao R, Yang J. NF‐κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell. 2013;4(3):176‐185. doi: 10.1007/s13238-013-2084-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Gargalionis AN, Papavassiliou KA, Papavassiliou AG. Targeting STAT3 signaling pathway in colorectal cancer. Biomedicine. 2021;9(8):1016. doi: 10.3390/biomedicines9081016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Gamero AM, Young MR, Mentor‐Marcel R, et al. STAT2 contributes to promotion of colorectal and skin carcinogenesis*. Cancer Prev Res (Phila). 2010;3(4):495‐504. doi: 10.1158/1940-6207.CAPR-09-0105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Gao P, Niu N, Wei T, et al. The roles of signal transducer and activator of transcription factor 3 in tumor angiogenesis. Oncotarget. 2017;8(40):69139‐69161. doi: 10.18632/oncotarget.19932 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Zhu L‐Q, Zhang L, Zhang J, et al. Evodiamine inhibits high‐fat diet‐induced colitis‐associated cancer in mice through regulating the gut microbiota. J Integr Med. 2021;19(1):56‐65. doi: 10.1016/j.joim.2020.11.001 [DOI] [PubMed] [Google Scholar]
- 113. Zhao F‐L, Qin C‐F. EGF promotes HIF‐1α expression in colorectal cancer cells and tumor metastasis by regulating phosphorylation of STAT3. Eur Rev Med Pharmacol Sci. 2019;23(3):1055‐1062. doi: 10.26355/eurrev_201902_16993 [DOI] [PubMed] [Google Scholar]
- 114. Heichler C, Scheibe K, Schmied A, et al. STAT3 activation through IL‐6/IL‐11 in cancer‐associated fibroblasts promotes colorectal tumour development and correlates with poor prognosis. Gut. 2020;69(7):1269‐1282. doi: 10.1136/gutjnl-2019-319200 [DOI] [PubMed] [Google Scholar]
- 115. Gupta N, Miyauchi S, Martindale RG, et al. Upregulation of the amino acid transporter ATB0,+ (SLC6A14) in colorectal cancer and metastasis in humans. Biochim Biophys Acta. 2005;1741(1–2):215‐223. doi: 10.1016/j.bbadis.2005.04.002 [DOI] [PubMed] [Google Scholar]
- 116. Kasprzak A. Angiogenesis‐related functions of Wnt signaling in colorectal carcinogenesis. Cancers (Basel). 2020;12(12):3601. doi: 10.3390/cancers12123601 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Tufail M, Wu C. Wnt3a is a promising target in colorectal cancer. Med Oncol. 2023;40(3):86. doi: 10.1007/s12032-023-01958-2 [DOI] [PubMed] [Google Scholar]
- 118. Nusse R, Clevers H. Wnt/β‐catenin signaling, disease, and emerging therapeutic modalities. Cell. 2017;169(6):985‐999. doi: 10.1016/j.cell.2017.05.016 [DOI] [PubMed] [Google Scholar]
- 119. Kramer N, Schmöllerl J, Unger C, et al. Autocrine WNT2 signaling in fibroblasts promotes colorectal cancer progression. Oncogene. 2017;36(39):5460‐5472. doi: 10.1038/onc.2017.144 [DOI] [PubMed] [Google Scholar]
- 120. Unterleuthner D, Neuhold P, Schwarz K, et al. Cancer‐associated fibroblast‐derived WNT2 increases tumor angiogenesis in colon cancer. Angiogenesis. 2020;23(2):159‐177. doi: 10.1007/s10456-019-09688-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Karpisheh V, Joshi N, Zekiy AO, et al. EP4 receptor as a novel promising therapeutic target in colon cancer. Pathol Res Pract. 2020;216(12):153247. doi: 10.1016/j.prp.2020.153247 [DOI] [PubMed] [Google Scholar]
- 122. Parizadeh SM, Jafarzadeh‐Esfehani R, Fazilat‐Panah D, et al. The potential therapeutic and prognostic impacts of the C‐MET/HGF signaling pathway in colorectal cancer. IUBMB Life. 2019;71(7):802‐811. doi: 10.1002/iub.2063 [DOI] [PubMed] [Google Scholar]
- 123. Papadakos SP, Petrogiannopoulos L, Pergaris A, Theocharis S. The EPH/Ephrin system in colorectal cancer. Int J Mol Sci. 2022;23(5):2761. doi: 10.3390/ijms23052761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Mendoza M, Khanna C. Revisiting the seed and soil in cancer metastasis. Int J Biochem Cell Biol. 2009;41(7):1452‐1462. doi: 10.1016/j.biocel.2009.01.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Baghban R, Roshangar L, Jahanban‐Esfahlan R, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020;18(1):59. doi: 10.1186/s12964-020-0530-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. LaGory EL, Giaccia AJ. The ever‐expanding role of HIF in tumour and stromal biology. Nat Cell Biol. 2016;18(4):356‐365. doi: 10.1038/ncb3330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Luo Z, Wang Q, Lau WB, et al. Tumor microenvironment: the culprit for ovarian cancer metastasis? Cancer Lett. 2016;377(2):174‐182. doi: 10.1016/j.canlet.2016.04.038 [DOI] [PubMed] [Google Scholar]
- 128. Salazar N, Zabel BA. Support of tumor endothelial cells by chemokine receptors. Front Immunol. 2019;10:147. doi: 10.3389/fimmu.2019.00147 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kloc M, Kubiak JZ, Li XC, Ghobrial RM. Pericytes, Microvasular dysfunction, and chronic rejection. Transplantation. 2015;99(4):658‐667. doi: 10.1097/TP.0000000000000648 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Birbrair A. Pericyte biology: development, homeostasis, and disease. Adv Exp Med Biol. 2018;1109:1‐3. doi: 10.1007/978-3-030-02601-1_1 [DOI] [PubMed] [Google Scholar]
- 131. Wang FT, Sun W, Zhang JT, Fan YZ. Cancer‐associated fibroblast regulation of tumor neo‐angiogenesis as a therapeutic target in cancer. Oncol Lett. 2019;17(3):3055‐3065. doi: 10.3892/ol.2019.9973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Zhou W, Xu G, Wang Y, et al. Oxidative stress induced autophagy in cancer associated fibroblast enhances proliferation and metabolism of colorectal cancer cells. Cell Cycle. 2017;16(1):73‐81. doi: 10.1080/15384101.2016.1252882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. 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: 10.1186/s12943-019-1019-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Paauwe M, Schoonderwoerd MJA, Helderman RFCP, et al. Endoglin expression on cancer‐associated fibroblasts regulates invasion and stimulates colorectal cancer metastasis. Clin Cancer Res. 2018;24(24):6331‐6344. doi: 10.1158/1078-0432.CCR-18-0329 [DOI] [PubMed] [Google Scholar]
- 135. Larionova I, Tuguzbaeva G, Ponomaryova A, et al. Tumor‐associated macrophages in human breast, colorectal, lung, ovarian and prostate cancers. Front Oncol. 2020;10:566511. doi: 10.3389/fonc.2020.566511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Larionova I, Kazakova E, Gerashchenko T, Kzhyshkowska J. New Angiogenic regulators produced by TAMs: perspective for targeting tumor angiogenesis. Cancers (Basel). 2021;13(13):3253. doi: 10.3390/cancers13133253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Han L, Wang S, Wei C, et al. Tumour microenvironment: a non‐negligible driver for epithelial‐mesenchymal transition in colorectal cancer. Expert Rev Mol Med. 2021;23:e16. doi: 10.1017/erm.2021.13 [DOI] [PubMed] [Google Scholar]
- 138. Mizuno R, Kawada K, Itatani Y, Ogawa R, Kiyasu Y, Sakai Y. The role of tumor‐associated neutrophils in colorectal cancer. Int J Mol Sci. 2019;20(3):529. doi: 10.3390/ijms20030529 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Nelson AR, Fingleton B, Rothenberg ML, Matrisian LM. Matrix Metalloproteinases: biologic activity and clinical implications. J Clin Oncol. 2000;18(5):1135‐1149. doi: 10.1200/JCO.2000.18.5.1135 [DOI] [PubMed] [Google Scholar]
- 140. Buttacavoli M, Di Cara G, Roz E, Pucci‐Minafra I, Feo S, Cancemi P. Integrated multi‐omics investigations of Metalloproteinases in colon cancer: focus on MMP2 and MMP9. Int J Mol Sci. 2021;22(22):12389. doi: 10.3390/ijms222212389 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Ren S, Liu J, Feng Y, et al. Knockdown of circDENND4C inhibits glycolysis, migration and invasion by up‐regulating miR‐200b/c in breast cancer under hypoxia. J Exp Clin Cancer Res. 2019;38(1):388. doi: 10.1186/s13046-019-1398-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Chen Y, Zheng X, Wu C. The role of the tumor microenvironment and treatment strategies in colorectal cancer. Front Immunol. 2021;12:792691. doi: 10.3389/fimmu.2021.792691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Yu L, Zhang M‐M, Hou J‐G. Molecular and cellular pathways in colorectal cancer: apoptosis, autophagy and inflammation as key players. Scand J Gastroenterol. 2022;57(11):1279‐1290. doi: 10.1080/00365521.2022.2088247 [DOI] [PubMed] [Google Scholar]
- 144. Yin Z, Li C, Wang J, Xue L. Myeloid‐derived suppressor cells: roles in the tumor microenvironment and tumor radiotherapy. Int J Cancer. 2019;144(5):933‐946. doi: 10.1002/ijc.31744 [DOI] [PubMed] [Google Scholar]
- 145. OuYang L‐Y, Wu X‐J, Ye S‐B, et al. Tumor‐induced myeloid‐derived suppressor cells promote tumor progression through oxidative metabolism in human colorectal cancer. J Transl Med. 2015;13:47. doi: 10.1186/s12967-015-0410-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Cheng KJ, Mejia Mohammed EH, Khong TL, Mohd Zain S, Thavagnanam S, Ibrahim ZA. IL‐1α and colorectal cancer pathogenesis: enthralling candidate for anti‐cancer therapy. Crit Rev Oncol Hematol. 2021;163:103398. doi: 10.1016/j.critrevonc.2021.103398 [DOI] [PubMed] [Google Scholar]
- 147. Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity. 2008;28(4):454‐467. doi: 10.1016/j.immuni.2008.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Razi S, Baradaran Noveiry B, Keshavarz‐Fathi M, Rezaei N. IL‐17 and colorectal cancer: from carcinogenesis to treatment. Cytokine. 2019;116:7‐12. doi: 10.1016/j.cyto.2018.12.021 [DOI] [PubMed] [Google Scholar]
- 149. Wang K, Kim MK, Di Caro G, et al. Interleukin‐17 receptor a signaling in transformed enterocytes promotes early colorectal tumorigenesis. Immunity. 2014;41(6):1052‐1063. doi: 10.1016/j.immuni.2014.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Chung AS, Wu X, Zhuang G, et al. An Interleukin‐17‐mediated paracrine network promotes tumor resistance to anti‐Angiogenic therapy. Nat Med. 2013;19(9):1114‐1123. doi: 10.1038/nm.3291 [DOI] [PubMed] [Google Scholar]
- 151. Zhao Y, Wu T, Shao S, Shi B, Zhao Y. Phenotype, development, and biological function of myeloid‐derived suppressor cells. Onco Targets Ther. 2016;5(2):e1004983. doi: 10.1080/2162402X.2015.1004983 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Itatani Y, Kawada K, Inamoto S, et al. The role of chemokines in promoting colorectal cancer invasion/metastasis. Int J Mol Sci. 2016;17(5):643. doi: 10.3390/ijms17050643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci. 2020;77(9):1745‐1770. doi: 10.1007/s00018-019-03351-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Zheng Z, Bian C, Wang H, et al. Prediction of immunotherapy efficacy and immunomodulatory role of hypoxia in colorectal cancer. Ther Adv Med Oncol. 2022;14:17588359221138383. doi: 10.1177/17588359221138383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Rasheed S, McDonald PJ, Northover JM, Guenther T. Angiogenesis and hypoxic factors in colorectal cancer. Pathol Res Pract. 2008;204(7):501‐510. doi: 10.1016/j.prp.2008.03.005 [DOI] [PubMed] [Google Scholar]
- 156. Barsoum IB, Smallwood CA, Siemens DR, Graham CH. A mechanism of hypoxia‐mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665‐674. doi: 10.1158/0008-5472.CAN-13-0992 [DOI] [PubMed] [Google Scholar]
- 157. Noman MZ, Desantis G, Janji B, et al. PD‐L1 is a novel direct target of HIF‐1α, and its blockade under hypoxia enhanced MDSC‐mediated T cell activation. J Exp Med. 2014;211(5):781‐790. doi: 10.1084/jem.20131916 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Semenza GL. Hypoxia‐inducible factors in physiology and medicine. Cell. 2012;148(3):399‐408. doi: 10.1016/j.cell.2012.01.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Arabsorkhi Z, Sadeghi H, Gharib E, Rejali L, Asadzadeh‐Aghdaei H, Nazemalhosseini‐Mojarad E. Can hypoxia‐inducible factor‐1α overexpression discriminate human colorectal cancers with different microsatellite instability? Genes Genet Syst. 2021;96(4):193‐198. doi: 10.1266/ggs.21-00026 [DOI] [PubMed] [Google Scholar]
- 160. Lee JW, Bae SH, Jeong JW, Kim SH, Kim KW. Hypoxia‐inducible factor (HIF‐1)alpha: its protein stability and biological functions. Exp Mol Med. 2004;36(1):1‐12. doi: 10.1038/emm.2004.1 [DOI] [PubMed] [Google Scholar]
- 161. Krek W. VHL takes HIF's breath away. Nat Cell Biol. 2000;2(7):E121‐E123. doi: 10.1038/35017129 [DOI] [PubMed] [Google Scholar]
- 162. Coronel‐Hernández J, Delgado‐Waldo I, Cantú de León D, et al. HypoxaMIRs: key regulators of hallmarks of colorectal cancer. Cell. 2022;11(12):1895. doi: 10.3390/cells11121895 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Zhong X, He X, Wang Y, et al. Warburg effect in colorectal cancer: the emerging roles in tumor microenvironment and therapeutic implications. J Hematol Oncol. 2022;15(1):160. doi: 10.1186/s13045-022-01358-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Monteiro HP, Rodrigues EG, Amorim Reis AKC, et al. Nitric oxide and interactions with reactive oxygen species in the development of melanoma, breast, and colon cancer: a redox signaling perspective. Nitric Oxide. 2019;89:1‐13. doi: 10.1016/j.niox.2019.04.009 [DOI] [PubMed] [Google Scholar]
- 165. Moraes MS, Costa PE, Batista WL, et al. Endothelium‐derived nitric oxide (NO) activates the NO‐epidermal growth factor receptor‐mediated signaling pathway in bradykinin‐stimulated angiogenesis. Arch Biochem Biophys. 2014;558:14‐27. doi: 10.1016/j.abb.2014.06.011 [DOI] [PubMed] [Google Scholar]
- 166. Knowles RG, Moncada S. Nitric oxide synthases in mammals. Biochem J. 1994;298(Pt 2):249‐258. doi: 10.1042/bj2980249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Wang H, Wang L, Xie Z, et al. Nitric oxide (NO) and NO synthases (NOS)‐based targeted therapy for colon cancer. Cancers (Basel). 2020;12(7):1881. doi: 10.3390/cancers12071881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Xiao Y, Zhong J, Zhong B, et al. Exosomes as potential sources of biomarkers in colorectal cancer. Cancer Lett. 2020;476:13‐22. doi: 10.1016/j.canlet.2020.01.033 [DOI] [PubMed] [Google Scholar]
- 169. Li S, Li Y, Chen B, et al. exoRBase: a database of circRNA, lncRNA and mRNA in human blood exosomes. Nucleic Acids Res. 2018;46(D1):D106‐D112. doi: 10.1093/nar/gkx891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Yang K, Zhou Q, Qiao B, et al. Exosome‐derived noncoding RNAs: function, mechanism, and application in tumor angiogenesis. Mol Ther Nucleic Acids. 2022;27:983‐997. doi: 10.1016/j.omtn.2022.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Esteller M. Non‐coding RNAs in human disease. Nat Rev Genet. 2011;12(12):861‐874. doi: 10.1038/nrg3074 [DOI] [PubMed] [Google Scholar]
- 172. Tang XJ, Wang W, Hann SS. Interactions among lncRNAs, miRNAs and mRNA in colorectal cancer. Biochimie. 2019;163:58‐72. doi: 10.1016/j.biochi.2019.05.010 [DOI] [PubMed] [Google Scholar]
- 173. Desmond BJ, Dennett ER, Danielson KM. Circulating extracellular vesicle MicroRNA as diagnostic biomarkers in early colorectal cancer‐a review. Cancers (Basel). 2019;12(1):52. doi: 10.3390/cancers12010052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Bjørnetrø T, Redalen KR, Meltzer S, et al. An experimental strategy unveiling Exosomal microRNAs 486‐5p, 181a‐5p and 30d‐5p from hypoxic tumour cells as circulating indicators of high‐risk rectal cancer. J Extracell Vesicles. 2019;8(1):1567219. doi: 10.1080/20013078.2019.1567219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Liu Y, Luo F, Wang B, et al. STAT3‐regulated Exosomal miR‐21 promotes angiogenesis and is involved in neoplastic processes of transformed human bronchial epithelial cells. Cancer Lett. 2016;370(1):125‐135. doi: 10.1016/j.canlet.2015.10.011 [DOI] [PubMed] [Google Scholar]
- 176. Wu J, Li H, Xie H, Wu X, Lan P. The malignant role of exosomes in the communication among colorectal cancer cell, macrophage and microbiome. Carcinogenesis. 2019;40(5):601‐610. doi: 10.1093/carcin/bgy138 [DOI] [PubMed] [Google Scholar]
- 177. Sarraf JS, Puty TC, da Silva EM, et al. Noncoding RNAs and colorectal cancer: a general overview. Microrna. 2020;9(5):336‐345. doi: 10.2174/2211536609666201221124608 [DOI] [PubMed] [Google Scholar]
- 178. De Los Santos MC, Dragomir MP, Calin GA. The role of Exosomal long non‐coding RNAs in cancer drug resistance. Cancer Drug Resist. 2019;2(4):1178‐1192. doi: 10.20517/cdr.2019.74 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Chen S, Fang Y, Sun L, He R, He B, Zhang S. Long non‐coding RNA: a potential strategy for the diagnosis and treatment of colorectal cancer. Front Oncol. 2021;11:762752. doi: 10.3389/fonc.2021.762752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Tan F, Chen J, Wang B, et al. LncRNA SBF2‐AS1: a budding star in various cancers. Curr Pharm des. 2022;28(18):1513‐1522. doi: 10.2174/1381612828666220418131506 [DOI] [PubMed] [Google Scholar]
- 181. Zhao Z, Sun W, Guo Z, Zhang J, Yu H, Liu B. Mechanisms of lncRNA/microRNA interactions in angiogenesis. Life Sci. 2020;254:116900. doi: 10.1016/j.lfs.2019.116900 [DOI] [PubMed] [Google Scholar]
- 182. Ballantyne MD, McDonald RA, Baker AH. lncRNA/MicroRNA interactions in the vasculature. Clin Pharmacol Ther. 2016;99(5):494‐501. doi: 10.1002/cpt.355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Wang L, Cho KB, Li Y, Tao G, Xie Z, Guo B. Long noncoding RNA (lncRNA)‐mediated competing endogenous RNA networks provide novel potential biomarkers and therapeutic targets for colorectal cancer. Int J Mol Sci. 2019;20(22):5758. doi: 10.3390/ijms20225758 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Shirmohamadi M, Eghbali E, Najjary S, et al. Regulatory mechanisms of microRNAs in colorectal cancer and colorectal cancer stem cells. J Cell Physiol. 2020;235(2):776‐789. doi: 10.1002/jcp.29042 [DOI] [PubMed] [Google Scholar]
- 185. Tang Y, Zong S, Zeng H, et al. MicroRNAs and angiogenesis: a new era for the Management of Colorectal Cancer. Cancer Cell Int. 2021;21(1):221. doi: 10.1186/s12935-021-01920-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Lun W, Wu X, Deng Q, Zhi F. MiR‐218 regulates epithelial‐mesenchymal transition and angiogenesis in colorectal cancer via targeting CTGF. Cancer Cell Int. 2018;18:83. doi: 10.1186/s12935-018-0575-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Zhang Q, Wang W, Zhou Q, et al. Roles of circRNAs in the tumour microenvironment. Mol Cancer. 2020;19(1):14. doi: 10.1186/s12943-019-1125-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Chen LL, Yang L. Regulation of circRNA biogenesis. RNA Biol. 2015;12(4):381‐388. doi: 10.1080/15476286.2015.1020271 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Chen LY, Wang L, Ren YX, et al. The circular RNA Circ‐ERBIN promotes growth and metastasis of colorectal cancer by miR‐125a‐5p and miR‐138‐5p/4EBP‐1 mediated cap‐independent HIF‐1α translation. Mol Cancer. 2020;19(1):164. doi: 10.1186/s12943-020-01272-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190. Barbagallo D, Caponnetto A, Brex D, et al. CircSMARCA5 regulates VEGFA mRNA splicing and angiogenesis in glioblastoma Multiforme through the binding of SRSF1. Cancers (Basel). 2019;11(2):194. doi: 10.3390/cancers11020194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Tang X, Ren H, Guo M, Qian J, Yang Y, Gu C. Review on circular RNAs and new insights into their roles in cancer. Comput Struct Biotechnol J. 2021;19:910‐928. doi: 10.1016/j.csbj.2021.01.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Ameli‐Mojarad M, Ameli‐Mojarad M, Hadizadeh M, et al. The effective function of circular RNA in colorectal cancer. Cancer Cell Int. 2021;21(1):496. doi: 10.1186/s12935-021-02196-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Han S, Zhuang J, Wu Y, Wu W, Yang X. Progress in research on colorectal cancer‐related microorganisms and metabolites. Cancer Manag Res. 2020;12:8703‐8720. doi: 10.2147/CMAR.S268943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Han N, Pan Z, Liu G, Yang R, Yujing B. Hypoxia: the “invisible pusher” of gut microbiota. Front Microbiol. 2021;12:690600. doi: 10.3389/fmicb.2021.690600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16(11):690‐704. doi: 10.1038/s41575-019-0209-8 [DOI] [PubMed] [Google Scholar]
- 196. Bennedsen ALB, Furbo S, Bjarnsholt T, Raskov H, Gögenur I, Kvich L. The gut microbiota can orchestrate the signaling pathways in colorectal cancer. APMIS. 2022;130(3):121‐139. doi: 10.1111/apm.13206 [DOI] [PubMed] [Google Scholar]
- 197. Schirbel A, Kessler S, Rieder F, et al. Pro‐Angiogenic activity of TLRs and NLRs: a novel link between gut microbiota and intestinal angiogenesis. Gastroenterology. 2013;144(3):613‐623.e9. doi: 10.1053/j.gastro.2012.11.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Bruneau A, Baylatry MT, Joly AC, Sokol H. Gut microbiota: what impact on colorectal carcinogenesis and treatment? Bull Cancer. 2018;105(1):70‐80. doi: 10.1016/j.bulcan.2017.10.025 [DOI] [PubMed] [Google Scholar]
- 199. Nguyen TT, Ung TT, Kim NH, Jung YD. Role of bile acids in colon carcinogenesis. World J Clin Cases. 2018;6(13):577‐588. doi: 10.12998/wjcc.v6.i13.577 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Zhang Y, Chen Z, Li J. The current status of treatment for colorectal cancer in China. Medicine (Baltimore). 2017;96(40):e8242. doi: 10.1097/MD.0000000000008242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Deng Y, Chi P, Lan P, et al. Modified FOLFOX6 with or without radiation versus fluorouracil and Leucovorin with radiation in Neoadjuvant treatment of locally advanced rectal cancer: initial results of the Chinese FOWARC multicenter, open‐label, randomized three‐arm phase III trial. J Clin Oncol. 2016;34(27):3300‐3307. doi: 10.1200/JCO.2016.66.6198 [DOI] [PubMed] [Google Scholar]
- 202. Rödel C, Graeven U, Fietkau R, et al. Oxaliplatin added to fluorouracil‐based preoperative chemoradiotherapy and postoperative chemotherapy of locally advanced rectal cancer (the German CAO/ARO/AIO‐04 Study): final results of the multicentre, open‐label, randomised, phase 3 trial. Lancet Oncol. 2015;16(8):979‐989. doi: 10.1016/S1470-2045(15)00159-X [DOI] [PubMed] [Google Scholar]
- 203. Ku G, Tan IB, Yau T, et al. Management of colon cancer: resource‐stratified guidelines from the Asian oncology summit 2012. Lancet Oncol. 2012;13(11):e470‐e481. doi: 10.1016/S1470-2045(12)70424-2 [DOI] [PubMed] [Google Scholar]
- 204. Yoshino T, Arnold D, Taniguchi H, et al. Pan‐Asian adapted ESMO consensus guidelines for the Management of Patients with metastatic colorectal cancer: a JSMO‐ESMO initiative endorsed by CSCO, KACO, MOS, SSO and TOS. Ann Oncol. 2018;29(1):44‐70. doi: 10.1093/annonc/mdx738 [DOI] [PubMed] [Google Scholar]
- 205. Chen WZ, Jiang JX, Yu XY, et al. Endothelial cells in colorectal cancer. World J Gastrointest Oncol. 2019;11(11):946‐956. doi: 10.4251/wjgo.v11.i11.946 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Sitohy B, Nagy JA, Dvorak HF. Anti‐VEGF/VEGFR therapy for cancer: reassessing the target. Cancer Res. 2012;72(8):1909‐1914. doi: 10.1158/0008-5472.CAN-11-3406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Dvorak HF. Vascular permeability factor/vascular endothelial growth factor: a critical cytokine in tumor angiogenesis and a potential target for diagnosis and therapy. J Clin Oncol. 2002;20(21):4368‐4380. doi: 10.1200/JCO.2002.10.088 [DOI] [PubMed] [Google Scholar]
- 208. Hurwitz HI, Tebbutt NC, Kabbinavar F, et al. Efficacy and safety of bevacizumab in metastatic colorectal cancer: pooled analysis from seven randomized controlled trials. Oncologist. 2013;18(9):1004‐1012. doi: 10.1634/theoncologist.2013-0107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Passardi A, Nanni O, Tassinari D, et al. Effectiveness of bevacizumab added to standard chemotherapy in metastatic colorectal cancer: final results for first‐line treatment from the ITACa randomized clinical trial. Ann Oncol. 2015;26(6):1201‐1207. doi: 10.1093/annonc/mdv130 [DOI] [PubMed] [Google Scholar]
- 210. Giantonio BJ, Catalano PJ, Meropol NJ, et al. Eastern cooperative oncology group study E3200. Bevacizumab in combination with Oxaliplatin, fluorouracil, and Leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the eastern cooperative oncology group study E3200. J Clin Oncol. 2007;25(12):1539‐1544. doi: 10.1200/JCO.2006.09.6305 [DOI] [PubMed] [Google Scholar]
- 211. Cremolini C, Rossini D, Dell'Aquila E, et al. Rechallenge for patients with RAS and BRAF wild‐type metastatic colorectal cancer with acquired resistance to first‐line Cetuximab and Irinotecan. JAMA Oncol. 2019;5(3):343‐350. doi: 10.1001/jamaoncol.2018.5080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212. Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of Aflibercept to fluorouracil, Leucovorin, and Irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an Oxaliplatin‐based regimen. J Clin Oncol. 2012;30(28):3499‐3506. doi: 10.1200/JCO.2012.42.8201 [DOI] [PubMed] [Google Scholar]
- 213. Van Cutsem E, Joulain F, Hoff PM, et al. Aflibercept plus FOLFIRI vs. placebo plus FOLFIRI in second‐line metastatic colorectal cancer: a post hoc analysis of survival from the phase III VELOUR study subsequent to exclusion of patients who had recurrence during or within 6 months of completing adjuvant oxaliplatin‐based therapy. Target Oncol. 2016;11(3):383‐400. doi: 10.1007/s11523-015-0402-9 [DOI] [PubMed] [Google Scholar]
- 214. Tabernero J, Yoshino T, Cohn AL, et al. Ramucirumab versus placebo in combination with second‐line FOLFIRI in patients with metastatic colorectal carcinoma that progressed during or after first‐line therapy with bevacizumab, oxaliplatin, and a fluoropyrimidine (RAISE): a randomised, double‐blind, multicentre, phase 3 study. Lancet Oncol. 2015;16(5):499‐508. doi: 10.1016/S1470-2045(15)70127-0 [DOI] [PubMed] [Google Scholar]
- 215. Debeuckelaere C, Murgioni S, Lonardi S, et al. Ramucirumab: the long and winding road toward being an option for mCRC treatment. Expert Opin Biol Ther. 2019;19(5):399‐409. doi: 10.1080/14712598.2019.1600505 [DOI] [PubMed] [Google Scholar]
- 216. Chen Z, Jiang L. The clinical application of Fruquintinib on colorectal cancer. Expert Rev Clin Pharmacol. 2019;12(8):713‐721. doi: 10.1080/17512433.2019.1630272 [DOI] [PubMed] [Google Scholar]
- 217. Qin S, Li A, Yi M, Yu S, Zhang M, Wu K. Recent advances on anti‐angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J Hematol Oncol. 2019;12(1):27. doi: 10.1186/s13045-019-0718-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218. Fondevila F, Méndez‐Blanco C, Fernández‐Palanca P, González‐Gallego J, Mauriz JL. Anti‐Tumoral activity of single and combined Regorafenib treatments in preclinical models of liver and gastrointestinal cancers. Exp Mol Med. 2019;51(9):1‐15. doi: 10.1038/s12276-019-0308-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Ibrahim S, Girault A, Ohresser M, et al. Monoclonal antibodies targeting the IL‐17/IL‐17RA Axis: an opportunity to improve the efficiency of anti‐VEGF therapy in fighting metastatic colorectal cancer? Clin Colorectal Cancer. 2018;17(1):e109‐e113. doi: 10.1016/j.clcc.2017.10.003 [DOI] [PubMed] [Google Scholar]
- 220. Arai H, Battaglin F, Wang J, et al. Molecular insight of Regorafenib treatment for colorectal cancer. Cancer Treat Rev. 2019;81:101912. doi: 10.1016/j.ctrv.2019.101912 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221. Akin Telli T, Bregni G, Vanhooren M, Saude Conde R, Hendlisz A, Sclafani F. Regorafenib in combination with immune checkpoint inhibitors for mismatch repair proficient (pMMR)/microsatellite stable (MSS) colorectal cancer. Cancer Treat Rev. 2022;110:102460. doi: 10.1016/j.ctrv.2022.102460 [DOI] [PubMed] [Google Scholar]
- 222. Carmeliet P, Jain RK. Principles and mechanisms of vessel normalization for cancer and other Angiogenic diseases. Nat Rev Drug Discov. 2011;10(6):417‐427. doi: 10.1038/nrd3455 [DOI] [PubMed] [Google Scholar]
- 223. Yamagishi N, Teshima‐Kondo S, Masuda K, et al. Chronic inhibition of tumor cell‐derived VEGF enhances the malignant phenotype of colorectal cancer cells. BMC Cancer. 2013;13:229. doi: 10.1186/1471-2407-13-229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Cheng H, Jiang X, Zhang Q, et al. Naringin inhibits colorectal cancer cell growth by repressing the PI3K/AKT/mTOR signaling pathway. Exp Ther Med. 2020;19(6):3798‐3804. doi: 10.3892/etm.2020.8649 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Mendelsohn J, Prewett M, Rockwell P, Goldstein NI. CCR 20th anniversary commentary: a chimeric antibody, C225, inhibits EGFR activation and tumor growth. Clin Cancer Res. 2015;21(2):227‐229. doi: 10.1158/1078-0432.CCR-14-2491 [DOI] [PubMed] [Google Scholar]
- 226. Yarom N, Jonker DJ. The role of the epidermal growth factor receptor in the mechanism and treatment of colorectal cancer. Discov Med. 2011;11(57):95‐105. [PubMed] [Google Scholar]
- 227. Sobrero AF, Maurel J, Fehrenbacher L, et al. EPIC: phase III trial of Cetuximab plus Irinotecan after Fluoropyrimidine and Oxaliplatin failure in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(14):2311‐2319. doi: 10.1200/JCO.2007.13.1193 [DOI] [PubMed] [Google Scholar]
- 228. Wilson CY, Tolias P. Recent advances in cancer drug discovery targeting RAS. Drug Discov Today. 2016;21(12):1915‐1919. doi: 10.1016/j.drudis.2016.08.002 [DOI] [PubMed] [Google Scholar]
- 229. Janssen JBE, Medema JP, Gootjes EC, Tauriello DVF, Verheul HMW. Mutant RAS and the tumor microenvironment as dual therapeutic targets for advanced colorectal cancer. Cancer Treat Rev. 2022;109:102433. doi: 10.1016/j.ctrv.2022.102433 [DOI] [PubMed] [Google Scholar]
- 230. Marais R, Light Y, Paterson HF, Marshall CJ. Ras recruits Raf‐1 to the plasma membrane for activation by tyrosine phosphorylation. EMBO J. 1995;14(13):3136‐3145. doi: 10.1002/j.1460-2075.1995.tb07316.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Oh HH, Joo YE. Novel biomarkers for the diagnosis and prognosis of colorectal cancer. Intest Res. 2020;18(2):168‐183. doi: 10.5217/ir.2019.00080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Wang Y, Lei F, Rong W, Zeng Q, Sun W. Positive feedback between oncogenic KRAS and HIF‐1α confers drug resistance in colorectal cancer. Onco Targets Ther. 2015;8:1229‐1237. doi: 10.2147/OTT.S80017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Battaglin F, Puccini A, Intini R, et al. The role of tumor angiogenesis as a therapeutic target in colorectal cancer. Expert Rev Anticancer Ther. 2018;18(3):251‐266. doi: 10.1080/14737140.2018.1428092 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Costea T, Vlad OC, Miclea L‐C, Ganea C, Szöllősi J, Mocanu M‐M. Alleviation of multidrug resistance by flavonoid and non‐flavonoid compounds in breast, lung, colorectal and prostate cancer. Int J Mol Sci. 2020;21(2):401. doi: 10.3390/ijms21020401 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Menegazzi M, Masiello P, Novelli M. Anti‐tumor activity of Hypericum Perforatum L. and Hyperforin through modulation of inflammatory signaling, ROS generation and proton dynamics. Antioxidants (Basel). 2020;10(1):18. doi: 10.3390/antiox10010018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Naz I, Ramchandani S, Khan MR, Yang MH, Ahn KS. Anticancer potential of Raddeanin a, a natural triterpenoid isolated from anemone Raddeana regel. Molecules. 2020;25(5):1035. doi: 10.3390/molecules25051035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Chen M‐H, Gu Y‐Y, Zhang AL, Sze DM‐Y, Mo S‐L, May BH. Biological effects and mechanisms of Matrine and other constituents of Sophora Flavescens in colorectal cancer. Pharmacol Res. 2021;171:105778. doi: 10.1016/j.phrs.2021.105778 [DOI] [PubMed] [Google Scholar]
- 238. Bardelli A, Siena S. Molecular mechanisms of resistance to Cetuximab and Panitumumab in colorectal cancer. J Clin Oncol. 2010;28(7):1254‐1261. doi: 10.1200/JCO.2009.24.6116 [DOI] [PubMed] [Google Scholar]
- 239. Bergers G, Hanahan D. Modes of resistance to anti‐angiogenic therapy. Nat Rev Cancer. 2008;8(8):592‐603. doi: 10.1038/nrc2442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. van Beijnum JR, Nowak‐Sliwinska P, Huijbers EJM, Thijssen VL, Griffioen AW. The great escape; the hallmarks of resistance to antiangiogenic therapy. Pharmacol Rev. 2015;67(2):441‐461. doi: 10.1124/pr.114.010215 [DOI] [PubMed] [Google Scholar]
- 241. Itatani Y, Kawada K, Yamamoto T, Sakai Y. Resistance to anti‐angiogenic therapy in cancer‐alterations to anti‐VEGF pathway. Int J Mol Sci. 2018;19(4):1232. doi: 10.3390/ijms19041232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242. Falcon BL, Chintharlapalli S, Uhlik MT, Pytowski B. Antagonist antibodies to vascular endothelial growth factor receptor 2 (VEGFR‐2) as anti‐angiogenic agents. Pharmacol Ther. 2016;164:204‐225. doi: 10.1016/j.pharmthera.2016.06.001 [DOI] [PubMed] [Google Scholar]
- 243. Zhou J, Ji Q, Li Q. Resistance to anti‐EGFR therapies in metastatic colorectal cancer: underlying mechanisms and reversal strategies. J Exp Clin Cancer Res. 2021;40(1):328. doi: 10.1186/s13046-021-02130-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Markman JL, Shiao SL. Impact of the immune system and immunotherapy in colorectal cancer. J Gastrointest Oncol. 2015;6(2):208‐223. doi: 10.3978/j.issn.2078-6891.2014.077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Wang J, Reiss KA, Khatri R, Jaffee E, Laheru D. Immune therapy in GI malignancies: a review. J Clin Oncol. 2015;33(16):1745‐1753. doi: 10.1200/JCO.2015.60.7879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246. Callahan MK, Wolchok JD. At the bedside: CTLA‐4‐ and PD‐1‐blocking antibodies in cancer immunotherapy. J Leukoc Biol. 2013;94(1):41‐53. doi: 10.1189/jlb.1212631 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell. 2015;161(2):205‐214. doi: 10.1016/j.cell.2015.03.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248. Chen YP, Zhang Y, Lv JW, et al. Genomic analysis of tumor microenvironment immune types across 14 solid cancer types: immunotherapeutic implications. Theranostics. 2017;7(14):3585‐3594. doi: 10.7150/thno.21471 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Franke AJ, Skelton WP, Starr JS, et al. Immunotherapy for colorectal cancer: a review of current and novel therapeutic approaches. J Natl Cancer Inst. 2019;111(11):1131‐1141. doi: 10.1093/jnci/djz093 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data has been used or generated.
