Abstract
Inactivating mutations of the adenomatous polyposis coli (APC) gene and consequential upregulation of the Wnt signaling pathway are critical initiators in the development of colorectal cancer (CRC), the third most common cancer in the United States for both men and women. Emerging evidence suggests APCmutations are also found in gastric, breast and other cancers. The APC gene, located on chromosome 5q, is responsible for negatively regulating the b-catenin/Wnt pathway by creating a destruction complex with Axin/Axin2, GSK-3b, and CK1. In the event of an APC mutation, b-catenin accumulates, translocates to the cell nucleus and increases the transcription of Wnt target genes that have carcinogenic consequences in gastrointestinal epithelial stem cells. A literature review was conducted to highlight carcinogenesis related to APC mutations, as well as preclinical and clinical studies for potential therapies that target steps in inflammatory pathways, including IL-6 transduction, and Wnt pathway signaling regulation. Although a range of molecular targets have been explored in murine models, relatively few pharmacological agents have led to substantial increases in survival for patients with colorectal cancer clinically. This article reviews a range of molecular targets that may be efficacious targets for tumors with APC mutations.
Key words: APC gene, colon cancer, colorectal cancer
Introduction
Loss of a functional adenomatous polyposis coli (APC) tumor suppressor gene has long been implicated in the initiation of colorectal cancer, a leading cause of cancer mortality worldwide. Aside from CRC, the APC mutation has additionally been linked to gastric, breast, lung, and brain neoplasms.1-4 The APC gene, located on chromosome 5q, negatively regulates the bcatenin/ WNT pathway that facilitates cell growth and differentiation. 5-7 Wnt is a growth stimulatory protein that when activated facilitates an increase in b-catenin levels and transcriptional upregulation of Wnt target genes implicated in invasive pathologies. 5 Mutations affecting the APC gene prohibit the formation of a b-catenin destruction complex composed of the APC protein, Axin/Axin2, glycogen synthase kinase-3b (GSK-3b), and casein kinase 1 (CK1).8-10 Without a functional destruction complex, bcatenin accumulates, leading to carcinogenic proliferation of gastrointestinal epithelial stem cells.6,11 As the bulk of APC mutations are found in colorectal pathology, the majority of this paper will focus on the pathophysiology and associated molecular targets being investigated as they relate to CRC and associated syndromes, though other solid tumors will also be described.
Transition of epithelial cells to a state of chromosomal instability (CIN) occurs through a series of events. In colorectal cancer, the first pathogenic step that leads to loss of APC heterozygosity and CIN has been associated with both DNA hypomethylation, increasing genomic instability, and hypermethylation, which results in silencing of the APC promotor sequence.12-14 Loss of APC gene function and subsequent accumulation of b-catenin is linked to decreased E-cadherin expression and, therefore, decreased intracellular adhesion.15 A subsequent KRAS mutation leads to the formation of colorectal polyps or benign adenomas, and a final loss of either p53 or Deleted in Colorectal Carcinoma (DCC) tumor suppressor genes leads to malignancy.16,17
Several genetic and modifiable factors have been proposed to contribute to the etiology of dysfunctional APC mutations, including hereditary components, dietary components, age, obesity, and colon inflammation (e.g, history of inflammatory bowel disease). For sporadic cases of colorectal cancers with an aberrant APC gene, loss of APC tends to be an initiating event while in colitisassociated cancer (CAC) it tends to be a later event.18 Patients with colitis have a 1.5-2 times greater likelihood of developing cancer, and it is hypothesized that inflammatory cytokine mediators such as IL-6 induce oxidative damage and influence dysplastic and neoplastic progression.19-32 Similarly, obese patients are hypothesized to require fewer somatic mutations to progress to microsatellite instable colon cancer as elevated cytokines are linked to obesity. Women qualifying as obese appear to particularly be at risk for early onset colorectal cancer.24 While more studies are needed to confirm the exact connection between obesity, inflammation, and cancer development, it is currently thought that obese patients have a lower threshold for carcinogenesis and the chronic inflammatory state contributes to over expression of related signaling pathways (e.g. JAK/STAT) that mediate cancer development. 25,26 Familial adenomatous polyposis (FAP) is an autosomal dominant inherited syndrome affecting the APC gene. The most common disease progression without surgical intervention is to colorectal carcinoma, though some patients carry additional risk for other cancers, including of the duodenum, pancreas, thyroid, liver and central nervous system (CNS).27 Phenotypically, patients will develop hundreds of premalignant polyps as early as their teenage years due to loss of heterozygosity of the APC allele. In addition to the original germline mutation most commonly found from codons 1061 to 1309, a second mutation renders both APC alleles nonfunctional.28 Progression to colorectal cancer in these patients is predictable and typically occurs by age 40 to 50, therefore prophylactic colectomies are often required as a preventative measure.29-31 However, most APC mutations are somatic, or noninherited. Approximately 60% of these sporadic cases appear to have mutations localized at the exon 15 region of the APC gene, termed the mutation cluster region (MCR).32 Interestingly, for unknown reasons patients with FAP who have germline mutations outside of codons 1194 and 1392, a region frequently associated with loss of heterozygosity (LOH) mutations, tend to also develop their second tumor-initiating mutation in the MCR.33,34 It is hypothesized that despite the differences in sporadic and hereditary colorectal cancers, genomic sequencing may be beneficial in both cases to identify potential therapeutic targets and interventions. 35
FAP is composed of several minor variants, including attenuated FAP (AFAP), Gardner Syndrome, and Turcot Syndrome, that are related to APC gene malfunction. AFAP is a category of APC mutation associated FAP typically denoted by fewer polyps, later onset of disease, and lower overall risk of development of colorectal cancer.36 This disease in general is thought to be milder and it has been implied that a unique third hit, or mutation, of a germline allele may occur in some instances of polyp formation.36 In turn, Gardner syndrome denotes a subset of FAP that is characterized by premalignant colonic polyps, but also extracolonic features including desmoid tumors, osteomas, fibromas, epidermal cysts, and ocular involvement.37 Finally, Turcot syndrome is both associated with FAP and hereditary non-polyposis colorectal cancer (HNPCC) which is characterized by colonic involvement with the addition of a primary brain tumor, such as glioblastoma or medulloblastoma. 38,39 These syndromes have unique features though each have an underlying propensity to develop colonic adenocarcinoma.
A growing literature base addresses an array of therapeutic regimens that could prove helpful in patients with APC mutated colorectal cancer.40 Typically, colorectal cancer patients receive a combined regimen of leucovorin, 5-fluorouracil, and oxaliplatin (FOLFOX), with survival rates for stage II or III cancer after 3 years reaching approximately 78.2%.41 However, given the high mortality rates with approximately 50,000 deaths per year in the United States alone, current therapies are inadequate.42-45 In cancers with APC inactivation, targeted treatment options are lacking and provide minimal increases in survival rates.
Molecular pathway
The APC gene codes for the APC protein, a multi-domain protein capable of binding to various cellular proteins. The APC protein is involved in many functions of the cell, with roles in development, tumor suppression, cell adhesion, cellular migration, and chromosome segregation through protein-protein interactions.46
APC is thought to play a vital role in fetal development. Its expression has been demonstrated in most fetal tissues.47 Embryonic stem cells without the APC gene are unable to differentiate. 48 In murine studies, complete knock-out in embryos is lethal.49-51 Heterozygous APC mutant mice are able to survive but develop many intestinal polyps.52 These mutant mice, referred to as multiple intestinal neoplasia or “Min” mice, have been used as models for intestinal tumorigenesis as the polyps closely resemble those of FAP patients.46
Wnt signaling and b-catenin regulation
As a tumor suppressor, APC acts to suppress the Wnt signaling pathway, which plays a key role in cellular proliferation and differentiation in several organ systems.46With proper Wnt-receptor/ligand interaction, buildup of b-catenin occurs and translocates to the nucleus, where it complexes with and activates T-cell factor (TCF), a Wnt signaling mediator (Figure 1). This interaction with TCF results in activation of target genes such as cyclin D1 (CCND1), MYC, and EphB, the ultimate targets of the signaling pathway which promote oncogenic proliferation and differentiation.46,53-55 Without proper Wnt signaling, APC inhibits b-catenin, preventing the downstream signaling of TCF and gene activation. APC prevents the accumulation and function of b-catenin by several mechanisms including, forming a complex with GSKb and axin which marks a-catenin for degradation; promoting b-catenin export from the nucleus; directly binding b-catenin, blocking its interaction with TCF; and forming a repressor complex which inhibits TCFdependent transcription.49-51,56
In cell adhesion, APC binds with b-catenin to adhere E-cadherin to both a-catenin and the actin cytoskeleton in the lateral plasma membrane of epithelial cells.57 Loss of this interaction between APC and b-catenin, and consequent loss of cell adhesion, is thought to contribute to tumorigenesis.58
Role in cytoskeleton stability, cell migration, and chromosome segregation
APC is important in the control and function of the actin cytoskeleton, particularly in cell polarity and migration. APC binds and activates Asef1, a guanine nucleotide exchange factor which in turn activates Cdc42, an important control protein in signaling pathways which alter the cytoskeleton and cause cell flattening, membrane ruffling, and cell migration.59 APC also binds with IQmotif- containing GTPase activation protein 1 (IQGAP1) to influence actin filaments and microtubules.60 In migrating cells, IQGAP1 and APC complex with CLIP-170, along with either Rac1 or Cdc42, in order to stabilize microtubules and to aid in polarized migration of the cell.60 Microtubule stabilization and polymerization are further regulated by APC forming a complex with the Rho downstream target EB1.61 Together, APC and EB1 bind the formin protein mDia on the plus-end of microtubules, capping and stabilizing the microtubule.61
APC has been found to be important in chromosome segregation during mitosis. Though its exact mechanism is not known, many different mutations of the APC gene have led to mitotic malfunction and tumor formation.46 During mitosis, APC is found associated with the kinetochores, mitotic spindles, and centrosomes. 62 It is thought that APC’s role in microtubule regulation is important in kinetochore function and chromosome segregation.63 Mutations which cause truncation of APC cause aneuploidy and chromosomal aberrations.64
Cell cycle regulation
The APC protein plays several roles in regulating the cell cycle, namely during the G1/S transition, the G2/M transition, and during base excision repair.65-67 First, with respect to the G1/S transition, APC has been shown to perform regulatory functions upstream of the Rb/E2F complex.66 b-catenin, which is inhibited by APC, activates cyclin D1.66 This in turn phosphorylates Rb, separating it from the E2F transcription factor and allowing E2F to make protein regulators such as cyclin E and cyclin A. These cyclins progress the cell past the G1-S restriction point.68 b-catenin also interacts with and activates c-MYC, which can enhance E2F and progress cells into the S phase as well.66 In this manner, inhibition of APC, which is common in colorectal cancers, can lead to unregulated cell cycle progression through lack of upstream modulation of b-catenin, cyclin D1, and c-MYC. Inhibition of APC and its effects on the G1/S transition may occur through its interaction with hDLG.69 hDLG is a member of the membrane-associated guanylate kinase protein family (MAGUK) and has been shown to form a complex with APC to inhibit further cell cycle progression.69 When hDLG is mutated, APC-regulated cells pass the G1/S restriction point unhindered.69 This response to a hDLG mutation implies that if the APC-hDLG complex cannot be made due to an hDLG mutation, then the G1/S restriction point may become unregulated and the cell cycle will proceed.
Furthermore, APC can regulate the cell cycle at the G2/M transition, though an exact mechanism has not been outlined.65 The APC protein has been shown to contain consensus sequences for phosphorylation via the CDC2 kinase, which usually binds to cyclin A to form a complex that assists in the progression from the G2 to M phase of the cell cycle.70,71 When APC is phosphorylated by CDC2, it is inhibited from binding to and stabilizing microtubules at their kinetochore sites.70,72 Microtubules are formed from centrosomes during mitosis and help pull chromosomes to the opposite ends of the cell. As stabilization of microtubules would impede dynamic tubulin polymerization during mitosis, the cell would subsequently be arrested in the G2 phase.72 In this way, unphosphorylated APC inhibits G2/M progression, whereas a phosphorylated APC allows mitosis to advance. Consequently, when CDC2 is upregulated in cancer settings, APC is phosphorylated and mitosis can proceed past the G2/M checkpoint.73 APC also regulates the G2/M transition through interactions with topoisomerase IIa (topo IIa).65 Topo IIa introduces double-stranded breaks into the DNA to facilitate strand passage and relegation, both of which are essential for DNA replication and chromosome condensation.65 It has been proposed that APC interacts with topo IIa and regulates its function during chromosome condensation at the beginning of cell cycle during the M phase.65 Consequently, when the APC protein is truncated due to a mutation, it ceases to modulate topo IIa’s effects which allows for chromosome instability during the M phase, a hallmark of several cancers including colorectal cancer.65 From these findings, it has been suggested that topo IIa inhibitors may be an effective chemotherapeutic target in APC-mutated colorectal cancer as this unregulated protein may be an essential component of cell cycle progression.65,74
Role in base excision repair
Lastly, APC may be involved with base excision repair.67 In low levels of DNA damage, the process of base excision repair occurs to replace single mispaired nucleotides.75 However, in high levels of DNA damage, such as significant exposure to alkylating agents, APC gene expression is stimulated.67 APC proteins form complexes with base excision repair modulators such as Fen-1 and Pol-b, which together block the overall base excision repair process. Because of this unrepaired DNA damage, the cell subsequently undergoes apoptosis.67 If APC is inhibited, base excision repair will not be blocked and damaged cells will proceed to survive and replicate. Overall, APC is involved in regulation of several areas of the cell cycle and DNA repair. Therefore, when APC is inhibited or downregulated, unfettered progression of cell replication can occur.
Figure 1.

A mutated APC gene leads to an nonfunctional destruction complex and subsequent accumulation of β-catenin. After translocating to the nucleus, β-catenin complexes with T-cell factor (TCF) to enhance transcription of oncogenic promoting genes, including CCND1, Myc, and EphB.
Path to tumorigenesis
As the APC gene functions as a tumor suppressor, a loss of heterozygosity via somatic mutation is needed for the protein to be rendered functionally inactive.76 This mutation is often a result of a truncation or insertion in the portion of the gene that codes for a protein binding region, which prevents binding to either Large Tumor Kinase 1 (LATS1) or b-catenin.76
Malignant transformation in colorectal cancer is driven by the loss of heterozygosity of APC within the JAK-STAT pathway which regulates IL-6.77Within this pathway, the APC protein binds with LATS1 to form a complex, which inhibits the Yes-Associated Protein (YAP). YAP can function as an inducer to gp130, a protein that is part of the transmembrane tyrosine kinase receptor of IL- 6.78 When IL-6 is bound extracellularly to this receptor, the membrane protein undergoes a conformational change, which allows JAK2 to bind to the intracellular side of the receptor and phosphorylate STAT3.78 This phosphorylation process occurs with the assistance of SRC Family Kinases (SFK’s), which remain activated due to the induction of YAP.79 When STAT3 is phosphorylated, it can act as a transcription factor and regulate expression of numerous genes including NF-kB (46). Subsequently, transcription for cytokines like IL-1 and TNF-a, results in pro-inflammatory effects within the tumor microenvironment.78 Phosphorylated STAT3 can also activate genes that promote proliferation and evasion of apoptosis.78 Therefore, the loss of function of APC renders the inhibitory complex with LATS1 inoperable and causes the constitutional activation of YAP and its downstream effects.
Neoplasms are promoted with DNA hypomethylation. DNA hypomethylation occurs when there is a loss of a maintenance methyltransferase named DNMT1.80 Total loss of DNMT1 in APCMin/+ mouse model with intestinal cancer showed an increase in progression of adenoma initiation due to hypomethylation.81 Two months after the removal of DNMT1, the APCMin/+ also showed an increase in macroadenoma load. However, there was no effect on tumor cell proliferation or apoptosis. Therefore, the loss of DNMT1 that leads to hypomethylation causes genomic instability, which accelerates loss of heterozygosity at the APC gene locus and ultimately causes an increase in tumor initiation.81
DNA hypermethylation in the promoter region of the APC gene leads to the inactivation of the APC gene. Since the APC gene is located on 5q (long arm of chromosome 5), a study investigating 137 sporadic colorectal cancer specimens with and without loss of heterozygosity at the 5q locus was performed.82 The results showed that tumors with a 5q loss of heterozygosity with reduced APC gene expression were more often hypermethylated than tumors with a 5q loss of heterozygosity with normal APC gene expression. Therefore, hypermethylation is associated with loss of APC gene expression.82
Preclinical studies-targeting therapeutics related to APC gene mutations
Role of COX-2 inhibition
The vast majority of studies aimed at understanding the effectiveness of different pharmacological agents for colorectal cancer recognize the role of inflammation in tumorigenesis. Cyclooxygenase-2 (COX-2), an enzyme in the arachidonic acid pathway involved in pro-inflammatory signaling of cytokines such as IL-6, is overexpressed in subgroups of colonic tumors. Celecoxib is a selective COX-2 inhibitor of frequent discussion in the context of inflammation and carcinogenesis. One study explored the effects of celecoxib in combination with piperine, a chemopreventative agent and p-glycoprotein inhibitor thought to increase drug delivery of combined agents.83,84 Evaluated in an HT-29 colon adenocarcinoma cell line, this combination was notably cytotoxic via induction of the caspase cell-death cascade and reactive oxygen species.83
In preclinical studies of APCMin mice, COX-2 has been found to initiate the development of intestinal and colonic polyps. COX- 2 (Ptgs2null mutation) knockouts in APC Δ716 mice demonstrated significant decrease in the number of intestinal polyps.85 Additionally, APC Δ716 mice treated with MF Tricyclic, a selective COX-2 inhibitor, showed marked decrease in tumors compared to treatment with sulindac, a non-selective, non-steroidal anti-inflammatory drug (NSAID); thus, COX-2 inhibition has been implicated as a potential chemo-preventative strategy.85 Furthermore, one study suggested that elevated prostaglandin E2 (PGE2), a pro-inflammatory mediator derived from COX-2 activity, in APCMin mice promotes tumorigenesis.86 The potency of NSAIDs directed at colonic tumors largely relies on the indirect ability to reduce PGE2. In addition, the authors also recognized the involvement of the C-X-C motif chemokine receptor 2 in advancing chronic inflammation and colorectal cancer pathogenesis via recruitment of myeloid-derived suppressor cells (MDSCs). This is significant because it provides another pathway that can be targeted in order to inhibit inflammation and tumor progression.86
A study by Bi et al. (2013), suggested that synergistic effects of sulindac, which is thought to inhibit prostaglandin synthesis via COX-1 and COX-2 inhibition, and selenium, a dietary supplement, can negatively impact tumor growth.87 In APC +/-/p21+/- mouse models given the sulindac/selenium combination for 24 weeks had meaningfully decreased tumorigenesis, with a 52% reduction in tumor incidence and approximately 80% reduction in tumor multiplicities (the number and variety of tumors). Mechanistically, this dual therapy increased expression of tumor suppressors p27 and p53, which are thought to play a role in b-catenin downregulation via E-cadherin/b-catenin adhesion mechanisms and ubiquitin/proteasome degradation systems, respectively.88,89 Additionally, JNK1, a MAPK family protein kinase, is thought to promote functional intestinal development and, like p27 and p53, is thought to negatively regulate b-catenin. It was also found that JNK1 phosphorylation increased in mouse models treated with selenium and sulindac, thus highlighting a promising anti-cancer treatment option.87
Antibiotics: macrolides and aminoglycosides
Aminoglycoside and macrolide family antibiotics have been found to induce read through of nonsense mutations in plasmids and colorectal cancer cell lines.90 Erythromycin, a macrolide antibiotic that inhibits the 50s subunit of bacterial ribosomes, has also been implicated in the treatment of colorectal cancer. Aside from the antimicrobial properties of this drug, erythromycin can also suppress development of intestinal tumors via inhibition of two transcriptional regulators: nuclear factor- kB (NFkB) and activator protein-1 (AP-1). In a sample of APCMin mice who were administered erythromycin for 15 weeks, polyps in the proximal small intestine were reduced approximately 71% compared to untreated control (P<0.01).91 Additionally, mRNA expression of IL-6 and COX-2, downstream targets of AP-1 and NFkB, were reduced in the treatment population.92 Another macrolide antibiotic, tylosin, had similar effects of tumor inhibition in APCMin mice that specifically had a nonsense mutation at codon 850.91
Due to their ability to induce stop codon read through, aminoglycosides, such as gentamicin and negamycin, a dipeptide antibiotic, have been explored to target nonsense mutations of the APC gene in cell lines. Two nonsense mutations, R1114X and L360X, were particularly susceptible to read through mechanisms by gentamicin. 93 Overall, both therapies were able to reintroduce APC activity in cell cultures with APC nonsense mutations.93
Herpesvirus vehicles
A study by Macnab et al. (2011) reintroduced a complementary DNA strand of the APC gene into a virus-based Herpesvirus samiri (HVS) vector with successful translation of full APC protein in two separate colorectal cancer cell lines: SW480 and a metastatic SW620. In both cell lines, further cancer cell growth was inhibited. Specifically in the SW480 line, a monolayer wound assay effectively measured diminished in vitro cell migration for 2 months, thus demonstrating the potential for virally-delivered gene therapies.94
Renin-angiotensin downregulation
Renin-angiontensin system targeting agents, including angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II receptor blockers (ARBs) are widely used in a number of conditions, namely hypertension; however, these agents have recently been implicated in colorectal cancer therapy and may decrease rates of liver metastasis.95-97 One study examined the combination of losartan, an ARB, and/or vitamin D, which is thought to downregulated renin in Apc+/LoxP Cdx2P-Cre mouse models to assess effects on tumor burden.98 Overall, it appeared that addition of vitamin D and losartan individually decreased tumor burden through upregulation of specific metabolites though the combination of vitamin D and losartan was not significant.98
mTOR inhibition
Sirolimus, or rapamycin, is a drug known to bind the binding protein FKBP-12 and inhibit the mechanistic target of rapamycin (mTOR), a serine/threonine protein kinase involved in cell proliferation that has become a topic of discussion for cancer therapies. 99 A mouse model for APC-deficient colonic polyposis, CDX2P-CreERT2 Apcfl/fl, demonstrated that treatment with rapamycin led to better median survival (P=0.003), fewer polyps (P=0.001), and decreased expression of β-catenin and Sox9 compared to the control cohort.100 Similar to rapamycin, everlimous has mTOR blocking effects by specifically downregulating mTORC1.101 In a ApcΔ716 mouse model, everolimus was shown to inhibit adenoma proliferation, tumor angiogenesis, and size and number of polyps.102
Mycophenolate mofetil and tacrolimus
Mycophenolate mofetil (MMF) and tacrolimus are agents that have complex mechanisms in immunosuppression, including inosine monophosphate dehydrogenase inhibition and calcineurin phosphatase inhibition, respectively. Classically, immunosuppressive therapies have been considered to increase risk for certain cancers, including lymphoma, following solid organ transplantation; however, MMF and tacrolimus have not specifically been shown to increase risk of colorectal cancer compared to other agents.103 One study interestingly reported a case of a teenage patient with FAP who following therapy with MMF and tacrolimus for 6 months after a kidney transplant experienced halted proliferation and dysplasia in colonic adenomas and decreased Ki-67 expression following treatment (P<0.001).104,105 In addition to pre- and posttreatment Ki-67 staining of a low grade dysplastic tubular adenoma from the patient, a human colonic adenocarcinoma cell line HT-29 containing a nonsense APC mutation was additionally treated with tacrolimus, MMF, or combination at varying doses. The combination of 10 μg/mL MMF and 0.1 μM tacrolimus demonstrated the greatest inhibition of the proliferating cell line at 24, 48, 72, and 96 hours compared to both solely MMF treatment (P<0.01) and the control (P<0.005). The cell-cycle phases of the HT-29 cells were also analyzed for percentage of cells in S phase, with increasing levels indicating cell arrest. After 48 hours, various treatments and dosing with 10 μg/mL MMF alone, combined 1 μM tacrolimus with 10 μg/mL MMF, combined 1 μM tacrolimus with 1 μg/mL MMF, combined 0.1 μM tacrolimus with 10 μg/mL MMF, and combined 0.1 μM tacrolimus with 1 μg/mL MMF were all significant compared to the control (respectively P<0.04; P<0.001; P<0.03; P<0.001; P<0.001). Overall, combined therapy with MMF and tacrolimus led to a regression in adenoma proliferation in an FAP patient and decreased cell cycle progression in a HT-29 cell line, thus indicating potential for future studies with these immunosuppressing therapies.
Src family kinases
As mentioned previously, APC loss of function mutations rather than b-catenin (CTNNB1) gain of function mutations are specifically linked to colorectal carcinogenesis.78 APC mutations appear to increase IL-6 signal transducers and subsequent activation of phosphorylated Src, YAP, STAT3, and Notch.78 Through antibody staining, 59% of human surgical colorectal cancer tumor samples were found to have overexpression of these signals, which was additionally replicated in Villin-Cre colonic mouse tumors.78 APC-/- mouse small intestinal organoids created via Adeno-Cre virus vectors demonstrated upregulations in the following mediators in mucosal regeneration: STAT3, Src tyrosine (Y) phosphorylation, YAP, mRNA for YAP targets, Jag1 (a Notch ligand), YAP targets cysteine-rich angiogenic inducer 61 (Cyr61) and connective tissue growth factor (Ctgf), leucine-rich repeat-containing Gprotein couple receptor 5 (Lgr5) and polycomb complex protein Bmi1.78 Each of these mediators has a variety of functions. STAT3 and YAP are transcription regulators that promotes cell cycle progression and oncogenesis.106,107 Jag1 is a ligand part of the Notch signaling pathway that through complex mechanisms promote cancer growth and angiogenesis.108 Lgr5 and Bmi1 are act on stem cells and when elevated are thought to be pathologic due to their ability to enhance the Wnt/b-catenin signaling pathway and downregulate E-cadherin, respectively.109,110
The role of the IL-6 signal transducer was also explored, as IL- 6 upregulation in APC-deficient intestinal cells is thought to direct activation of Src, YAP, STAT3, Notch and overall accelerate tumorigenesis. APC-null enteroids also demonstrated increased expression of cytokine markers: Il6st (gp130), IL6r mRNAs, Il11r mRNAs, Lif mRNAs, IL17ra mRNAs, and Il17rc through qRTPCR, with Il6st (gp130) and Il6r mRNAs being the most substantial. Il6st (gp130) has a binding TEA domain transcription factor 4 (Tead4), which forms a complex with YAP in human colorectal cells via chromatin immunoprecipitation.111 Downregulation of YAP and TAZ consequently reduces IL6st mRNA expression, which suggests that YAP is necessary for induction of the gp130 signal transducer.78 Because IL-6, the IL-6 receptor, and IL-11 first activate Src family (SFK) kinases which in turn upregulate YAP, SFK inhibitors are thought to reduce YAP gene expression and may have a potential role to indirectly reduce the expression of these cytokine signal transducers.
PP2, a selective Src family kinase inhibitor, saracatinib (AZD0503), a selective Src and Bcr-Abl tyrosine kinase inhibitor, and dibenzazepine (DBZ), a Notch/γ-secretase inhibitor, were individually administered at 10 μM for 24 hours to both wildtype and APC -/- enteroids to assess targeted treatment efficacy.78 AZD1480, a small JAK2 inhibitor, and ruxolitinib, a JAK1 and JAK2 inhibitor, were also individually administered at 3 μM for 24 hours. Notably, PP2 and saracatinib each inhibited YAP and Src tyrosine phosphorylation in APC-null human enteroids while PP2 additionally inhibited Notch activation.78 No substantial b-catenin downregulation was noted with Src inhibitors and no other agents demonstrated substantial inhibition in their molecular targets.78
As previously described in Taniguchi et al. (2015), gp130 in turn also activates YAP and Notch and leads to epithelial proliferation and colorectal pathology.112 However, intestinal tumor growth in APC+/IECΔ; Vil-gp130Act mice utilizing the Villin promoter for STAT3, SFK, YAP, and Notch signaling was sustained when either YAP or STAT3 was inhibited.112 More importantly, SFK and JAK inhibitors synergistically lead to tumor death. Combination PP2 and Ruxolitinib therapy in Cdx2-Cre x APCF/+mice for 1.5 months reduced tumor load (P<0.05) while PP2 and AZD1480 administration for 3 days lead to tumor death (P<0.05).112
Overall, the work by Taniguchi et al. (2015, 2017) in mouse models highlighted how constitutive expression of IL6st/gp130 in intestinal epithelial cells leads to progression of colorectal tumors whereas SFKs and JAK tyrosine kinase inhibition leads to downregulation of tumorigenesis and shrinkage of previously established colorectal pathologies.78,112 Authors noted that specifically blocking YAP and STAT3 via SFK and JAK inhibitors presents a unique opportunity for targeting therapeutics through interaction with mediators of the aberrant Wnt/b-catenin signaling pathway in APC mutations.
Targeting the Wnt signaling pathway
The APC gene is inactivated through mutation in familial adenomatous polyposis associated and sporadic colorectal cancers.113 However, mutation is not the only mechanism through which loss of expression occurs. Methylation of promoter regions is another common mechanism. Utilizing 22 different colorectal cancer cell lines, one study analyzed the methylation status of the APC promoter. 113 This study found that methylation of the CpG sites in the promoter region of the gene correlated with a loss of the APC gene expression.113 In cell lines lacking APC mutations, 5-Aza-2- Deoxycytidine, a methyltransferase inhibitor, was successful in inducing expression of APC.113
Dysregulation and overactivation of the Wnt pathway have also been found to be associated with cancer. R-spondins, RSPO1-4, are proteins responsible for activation of the Wnt/b-catenin pathway. RSPO fusion proteins were found to be mutually exclusive with APC gene mutations, which shows that these proteins have a role in increasing Wnt signaling and tumorigenesis.114 Chromosomal translocations that combine regulatory sequences with RSPO2 and RSPO3 are found in about 10% of colorectal cancers. 1 Wnt is activated through palmitoylation, a post translational modification which occurs through Porcupine (PORCN). The compound ETC-159 was found to inhibit palmitate incorporation into Wnt3a and inhibit growth of teratocarcinoma in human cell lines (115). Also, ETC-159 was found to inhibit the growth of colon cancers with RSPO translocations, making it a potentially effective treatment.115
CREB binding protein inhibition
Another drug that targets the Wnt pathway is PRI-724. PRI- 724 inhibits the CREB binding protein from interacting with β- catenin. Treatment with PRI-724 has been found to promote stem cell differentiation by increasing p300/β-catenin binding.116 Increased stem cell differentiation inhibits tumor initiating cells and increases sensitivity to targeted drugs.116
Casein kinase 1 inhibitors
The dysregulation of casein kinase 1 (CK1), a type of serine/threonine kinase, is another potential cause of colorectal cancer.117,118 CK1α and CK1δ are isoforms of CK1 and play a regulatory role in the APC/Wnt/β-catenin pathway by a number of complex mechanisms including inducing β-catenin phosphorylation, influencing cell cycle progression, and co-activating mTOR.119,120 When these two isoforms of CK1 are overexpressed, it leads to dysregulation of the APC/Wnt/β-catenin pathway and leads to colorectal cancer.117,118 Comparison of normal and tumor tissue showed that CK1α was overexpressed in colorectal cancer and thus has the potential to serve as prognostic biomarker. The study also determined that higher CK1α expression leads to poorer prognosis and decreased survival, though this finding has not yet been extended to other cancers with CK1α overexpression. The drugs D4476 and lenalidomide have been shown to potentially inhibit and degrade CK1, respectively.117,121,122
Tankyrase inhibitors
Several therapeutic agents that modify axin to increase bcatenin destabilization have been tested. Tankyrase inhibitors small molecule inhibitor, XAV939, promotes destruction of bcatenin via inhibition of b-catenin transcription and Axin stabilization. XAV939 stabilizes Axin, which forms part of the b-catenin destruction complex, by blocking the activity of both tankyrase 1 and tankyrase 2 enzymes that promote axin degradation.123 Another tankyrase inhibitor, G007-LK, has also promoted axin stabilization and decreased tumorigenesis in APC mutant mice, though clinical potential may be limited due to intestinal toxicity. 124 KYA1797K, which binds to axin’s G-protein signaling domain, has noted anti-tumorigenic properties in mutant APC and KRAS mice through stabilization of the proteasome complex that degrades b-catenin.125
TNIK Inhibitors
After translocating to the nucleus, b-catenin accumulates and forms complexes with several different transcription factors that lead to gene expression. The T-cell factor/lymphoid enhancer factor (TCF/LEF) family includes T-cell factor 4 and b-catenin transcriptional complex (TCF4), which is frequently stained in colorectal cancer tissue.126 Its abnormal overactivation is thought to be essential for halting differentiation of intestinal stem cells and promoting malignant proliferation.127 Traf2- and Nck-interacting kinase (TNIK) are known to be vital regulators of Wnt pathway activity by binding b-catenin, phosphorylating, and activating TCF4.128,129 Thus, its inhibition may provide potential clinical relevance for colorectal cancer patients. A small molecule TNIK inhibitor, NCB-0846, decreased tumor proliferation in both in vitro and in vivo APCMin mice and decreased expression of CD44, CD133 and aldehyde dehydrogenase-1 (ALDH1) cell markers, which typically are markers of cancer stem cell functioning and tumorigenesis.130
Frizzled receptor targeting
Another targeted therapeutic approach of the Wnt/b-catenin signaling pathway involves the Frizzled (Fzd) Wnt receptors. Frizzled-7 (Fzd7) receptor expression was recently linked to decreased survival in gastric cancer patients.131 Targeting Fzd7 with vantictumab, an IgG2 monoclonal antibody that binds Fzd receptors, notably mitigated proto-oncogenic Myc expression while additionally reducing chemoresistance.132 In mouse models, Fzd7 deficient cell lines were unable to sustain cancer growth via Wnt signal induction, while Fzd7 deletion or vantictumab therapy were effective in deterring tumor proliferation.132 Vantictumab has promising anti-oncogenic properties, and its effects were noticeable regardless of mutations in the APC gene.132 Ipafricept, a fusion protein containing part of Fzd8 linked to IgG1, has additionally been evaluated in solid tumors and was suggested to have benefit in a patient with a germ cell cancer that overexpressed bcatenin and had stable disease after treatment.133
Dikkopf-1 targeting
Dickkopf-1 (Dkk1) is a glycoprotein involved in antagonizing the Wnt signaling pathway through competitive LRP5/6 inhibition.134 Overexpression of Dkk1 confers a poor prognosis in hepatocellular, esophageal, non-small cell lung, and ovarian cancers and could be an optimal target for therapy.135-140 The humanized monoclonal IgG4 antibody, DKN-01, is emerging as a potential immunomodulatory therapy for patients suffering from cancers with a high Dkk1 burden.141,142 Conversely, Dkk1 has been demonstrated to have decreased expression in colon cancer and melanoma.143,144 In sample of APC-mutated nude mice with D-1 deficiency, reintroduction of a functional DKK-1 gene led to decreased tumorigenesis.145 At present, no targeted therapies specific to low Dkk-1 expression are in preclinical trials; however, the Dkk-3 protein, also in the Dkk family, is being investigated as a potential biomarker in colorectal cancer due to its typical underexpression compared to healthy controls.146
Chemotherapeutic drugs targeting the APC gene
Cisplatin is a common chemotherapeutic drug used to treat different cancers. Cisplatin works by crosslinking purine bases on DNA, which inhibits DNA repair mechanisms and causes cellular apoptosis.147 Cisplatin has been tested in APCMin/+ mice with mouse mammary tumor virus (MMTV) that were crossed to the polyoma middle T antigen (PyMT) transgenic model. Cells were extracted from MMTV-PyMT; ApcMin/+ mice tumors and were treated with cisplatin as well as doxorubicin and paclitaxel, which are also chemotherapeutic drugs. The tumor cells expressed multidrug resistance protein 1 (MDR1) after treatment with the three drugs, meaning that drug resistance occurred. Also, MMTVPyMT; APCMin/+ cells became more resistant to cisplatin and doxorubicin- induced apoptosis.
The same drugs were then tested in human metaplastic breast cancer cell line MDA-MB-157 with an APC knockdown. The results showed that without the APC gene, there was greater resistance to paclitaxel and cisplatin. However, when cisplatin was combined with PP2, a Src inhibitor, or SP600125, a JNK inhibitor, there was less resistance to treatment. Src and JNK regulate increased tumor cell proliferation downstream of the APC mutation, so targeting them would inhibit proliferation. The combination of cisplatin with PP2 and SP6000125 could prove to be clinically beneficial for breast cancer resistance caused by APC gene mutation.148
Targeting APC in the clinical setting
While an APC mutation is an initiating factor in roughly 80% of all colorectal cancers and is increasingly being explored in other cancers, there have not been many therapeutic targets identified. Most of the research conducted has focused on downstream signaling pathways regulated by APC, including WNT/β-catenin. The majority of the clinical trials have been directed at treatment for patients with familial adenomatous polyposis, which as mentioned previously is predominated by APC germline mutations. Table 1 provides a brief outline of the mentioned studies.
Erythromycin
Since the strategy of inducting read through of nonsense mutations in the APC gene was successful in murine models, one study investigated administration of erythromycin to humans with nonsense APC gene mutations. In a clinical trial, ten patients with FAP and APC nonsense mutations were given erythromycin for 4 months and had endoscopies at baseline, 4 months, and 12 months after treatment.149 Results indicated that the number of adenomas and cumulative and maximal polyp size decreased. Tissue samples were also collected for assessment and were then molecularly and genetically analyzed. Treatment not only reduced the number of somatic APC mutations, but also restored APC gene function and decreased cellular proliferation.149
Erlotinib and sulindac
The pro-inflammatory signaling role of COX-2 and the overexpression found in colorectal cancer has previously been described in this review. Aspirin, an irreversible and nonselective inhibitor of COX-1 and COX-2, has been shown to improve the survival for patients with non-metastatic colorectal cancer positive for COX-2 overexpression (HR, 0.39; 95% CI, 0.20-0.76).150
Table 1.
The above are clinical studies that target APC mutations, namely studied in familial adenomatous polyposis.
| Pharmacologic agent | Trial | Phase | Mechanism of action | Therapeutic use | Outcome |
|---|---|---|---|---|---|
| Erythromycin | NCT02175914 | IV | Induction of read through of nonsense mutations in the APC gene | Familial adenomatous polyposis (FAP) | i) Decreased number of` adenomas ii) Decreased polyp size |
| Erlotinib + sulindac | NCT01187901 | II | EGFR inhibition | FAP, attenuated FAP | i) Decreased quantity of polyps |
| Eicosapentaenoic acid | NCT00510692 | II, III | Omega-3 polyunsaturated fatty acid that modulates cyclooxygenase metabolism | FAP | i) Decreased polyp size |
| Celecoxib + difluoromethylornithine | NCT00033371 | II | Blocking upregulation of polyamine synthesis | FAP, colorectal cancer i) No significant decrease in olyp burden compared to placebo |
Prostaglandin E2 (PGE2) is produced by COX-2 and found to transactivate EGFR (epidermal growth factor receptor). EGFR has been increased in APC deficient murine models which indicate EGFR inhibition may be beneficial.151 Patients were given the EGFR inhibitor, erlotinib at 75 mg twice a day and sulindac (nonsteroidal anti-inflammatory) at 150 mg/day for 6 months.152 The goal of this study was to show decreased duodenal polyp amounts after 6 months of treatment. Ninety-two patients with FAP participated in this study and half of them received the drug combination while the other half received a placebo. The total number of polyps found in the colorectum, ileal pouch anal anastomosis, or ileo-rectum were recorded at baseline and reassessed after six months. After six months, it was found that the group that received erlotinib and sulindac had 69% less polyps compared to the placebo group. Four main side effects of these drugs include acne like rash, diarrhea, nausea, and oral mucositis.
Eicosapentaenoic acid
Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid (O3FA) that has been shown to decrease colorectal cancer in preclinical studies through multiple different mechanisms such as modulating cyclooxygenase metabolism, which is elevated in colorectal cancer, as well as reduction of prostaglandins.153 Therefore, the free fatty acid version of EPA (EPA-FFA) was tested in patients who were diagnosed with FAP. These patients were monitored through endoscopies of their retained rectum postcolectomy and were given 2 grams of EPA-FFA for six months. The number and size of their polyps were demarcated with a tattoo so a comparison before and after treatment could be made. At the end of this study, it was found that 22.4% of the polyps decreased after 6 months of treatment with EPA-FFA.154 Also, 29.8% of polyps decreased in the sum of the diameter compared to the placebo group. EPA-FFA was also well tolerated amongst the experimental group.154
Celecoxib and difluoromethylornithine
Another study combined celecoxib with difluoromethylornithine (DFMO) to see if polyp burden decreased in patients with FAP.155 DFMO is an irreversible enzyme-activated inhibitor of ornithine transcarbamylase which assists in polyamine synthesis. Polyamines have been found to potentially contribute to cancer formation in the colon and rectum through upregulation of polyamine biosynthesis enzymes.156 This makes the polyamine pathway a potential target for inhibiting tumor proliferation. Therefore, if DFMO can block the formation of polyamines, it might have an anticancer effect. After analyzing the polyp burden, there was no significant difference in the overall amount between placebo and DFMO treated patients.155
Ongoing clinical trials targeting Wnt signaling
A previous review highlighted emerging therapies for gastrointestinal tumors via different small molecule targets in the Wnt pathway.40 Currently, there are several clinical trials underway that are directed at components that are part of the Wnt signaling cascade. Of note, four of the six trials mentioned in Table 2 are DKK1 inhibitors, which are soluble inhibitors previously mentioned in this review. These trials are mostly related to gastrointestinal tract malignancies, though the PORCN inhibitor ETC-1922159 may be applicable to a number of solid tumors.
The role of azithromycin in inducing readthrough of APC nonsense mutations and inducing functional APC protein has previously been discussed in this review. One proposed phase IV study (NCT04454151) will investigate the effect of azithromycin therapy for 4-6 months on number and size of colonic adenomas, duodenal adenomas, and desmoid tumors in FAP patients. Additionally, samples will be taken to determine APC protein levels following medication administration. This study is not yet recruiting.
A separate study (NCT02521844) is a four-part, open label study evaluating ETC -1922159, a PORCN inhibitor, in a variety of unresectable solid tumors. Overall, the study evaluates the safety and efficacy of this medication for solid malignancies that lack other known treatment options with dose escalation scales. In certain parts of the study, the medication will be combined with denosumab (Part A extension) and pembrolizumab (Phase B). PORCN inhibitors and their connection to the Wnt signaling pathway have been described previously.
Several studies have also begun to investigate the DKN-01, a Wnt signaling pathway effector, in combination with other medications. NCT04166721 is an actively recruiting phase II study evaluating safety, tolerable dosing, and efficacy of DKN-01 and atezolizumab combination therapy in both metastatic esophageal and gastric cancer. NCT0436801 is an additional actively recruiting phase II study evaluating use of IV DKN-01 and tislelizumab with or without CAPOX in patients with inoperable gastric adenocarcinoma or gastroesophageal cancer. In a group of patients with advanced liver cancer, a combination of DKN-01 and sorafenib will be evaluated in an actively recruiting phase 1/II study (NCT03645980). Finally, DKN-01 and nivolumab in being investigated in patients with biliary tract cancer patients who are refractory to other therapies in a recruiting phase II study (NCT04057365).
Table 2.
The listed studies are currently actively recruiting participants for pharmacological trials linked to Wnt pathway inhibition.
| Pharmacologic agent | Trial | Phase | Mechanism of action | Therapeutic use |
|---|---|---|---|---|
| Azithromycin | NCT04454151 | IV | Ribosomal readthrough of APC nonsense mutations | Familial adenomatous polyposis |
| ETC-1922159 | NCT02521844 | I | PORCN inhibitor | Multiple solid tumors |
| DKN-01 plus atezolizumab | NCT04166721 | II | DKK1 inhibitor plus anti-PD-L1 monoclonal antibody | Metastatic esophageal cancer Metastatic gastric cancer |
| DKN-01 plus tislelizumab | NCT04363801 | II | DKK1 inhibitor plus IgG4 anti–PD-1 monoclonal antibody | Gastric cancer Gastric adenocarcinoma Gastroesophageal cancer |
| DKN-01 plus sorafenib | NCT03645980 | I, II | DKK1 inhibitor plus protein kinase inhibitor | Hepatocellular cancer |
| DKN-01 plus nivolumab | NCT04057365 | II | DKK1 inhibitor plus IgG4 monoclonal antibody and PD-L1/L2 inhibitor | Biliary tract cancer |
Conclusions
In the United States alone, it is projected that in 2021 there will be 149,500 new diagnoses and 52,980 deaths from colorectal cancer. 44,45 Recent predictions have estimated that by 2040 the incidence of colorectal cancer will be similar, approximated at 147,000 cases.157 In CRC, the APC gene on chromosome 5q is the most frequent initiating site of sporadic and familial mutations that leads to polyp formation and inevitable carcinoma, if untreated. Of note, APC gene mutations may also be found in a number of malignancies aside from CRC, though CRC is the most hallmark association with these mutations. The progression from adenoma to carcinoma has a typical stepwise progression after the loss APC functionality. In many cases, a subsequent activating KRAS mutation followed by a tumor suppressor genes mutation (TP53, DCC) leads to eventual tumor burden.
Surgery remains a mainstay of treatment to prevent malignant progression of CRC, though at present pharmacological options that can effectively mitigate disease are needed. Several cytotoxic, chemotherapeutic agents, including the FOLFOX regimen, have been implicated in the treatment of colorectal cancer for the past couple decades and are associated with significant side effect profiles; however, CRC remains the third most deadly cancer despite efforts to increase surveillance and scientific advances in treatment.
While several preclinical and clinical studies have addressed targeting upregulated inflammatory factors in CRC, including COX-2 and IL-6, therapies more specific to specific cytokine mediated transcription factors and Wnt pathway signalers are increasingly being explored. Potential molecular targets in mouse model from preclinical studies included IL-6 signal transducers (YAP, Src, STAT3, Notch), tankyrase enzymes, Traf2- and Nckinteracting kinase, and Frizzled 7 Wnt receptors. This review highlights several avenues that may prove efficacious for patients suffering from APC-mutated colorectal cancers and warrant further exploration.
Acknowledgments
the authors would like to acknowledge Brenda Marr for her competent and knowledgeable assistance in the preparation of the manuscript and Christina Egan for facilitating the collaboration among authors.
References
- 1.Furuuchi K, Tada M, Yamada H, et al. Somatic mutations of the APC gene in primary breast cancers. Am J Pathol 2000;156:1997-2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Oreffo VI, Robinson S, You M, et al. Decreased expression of the adenomatous polyposis coli (Apc) and mutated in colorectal cancer (Mcc) genes in mouse lung neoplasia. Mol Carcinogen 1998;21:37-49. [PubMed] [Google Scholar]
- 3.Mori T, Nagase H, Horii A, et al. Germ-line and somatic mutations of the APC gene in patients with Turcot syndrome and analysis of APC mutations in brain tumors. Genes Chromosomes Cancer 1994;9:168-72. [DOI] [PubMed] [Google Scholar]
- 4.Fang D-C, Luo Y-H, Yang S-M, et al. Mutation analysis of APC gene in gastric cancer with microsatellite instability. World J Gastroenterol 2002;8:787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rubinfeld B, Souza B, Albert I, et al. Association of the APC gene product with beta-catenin. Science 1993;262:1731-4. [DOI] [PubMed] [Google Scholar]
- 6.Munemitsu S, Albert I, Souza B, et al. Regulation of intracellular beta-catenin levels by the adenomatous polyposis coli (APC) tumor-suppressor protein. Proc Natl Acad Sci 1995;92:3046-50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Su L-K, Vogelstein B, Kinzler KW. Association of the APC tumor suppressor protein with catenins. Science 1993;262:1734-7. [DOI] [PubMed] [Google Scholar]
- 8.Behrens J, Jerchow B-A, Wurtele M, et al. Functional interaction of an axin homolog, conductin, with β-catenin, APC, and GSK3β. Science 1998;280:596-9. [DOI] [PubMed] [Google Scholar]
- 9.Ikeda S, Kishida S, Yamamoto H, et al. Axin, a negative regulator of the Wnt signaling pathway, forms a complex with GSK-3β and β-catenin and promotes GSK-3β-dependent phosphorylation of β-catenin. The EMBO J 1998;17:1371-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Sakanaka C, Weiss JB, Williams LT. Bridging of β-catenin and glycogen synthase kinase-3β by axin and inhibition of β- catenin-mediated transcription. Proc Natl Acad Sci 1998;95:3020-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sparks AB, Morin PJ, Vogelstein B, Kinzler KW. Mutational analysis of the APC/β-catenin/Tcf pathway in colorectal cancer. Cancer Res 1998;58:1130-4. [PubMed] [Google Scholar]
- 12.Rao CV, Yamada HY. Genomic instability and colon carcinogenesis: from the perspective of genes. Front Oncol 2013;3:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Yamada Y, Jackson-Grusby L, Linhart H, et al. Opposing effects of DNA hypomethylation on intestinal and liver carcinogenesis. Proc Natl Acad Sci 2005;102:13580-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Hiltunen MO, Alhonen L, Koistinaho J, et al. Hypermethylation of the APC (adenomatous polyposis coli) gene promoter region in human colorectal carcinoma. Int J Cancer 1997;70:644-8. [DOI] [PubMed] [Google Scholar]
- 15.Smits R, Ruiz P, Diaz-Cano S, et al. E-cadherin and adenomatous polyposis coli mutations are synergistic in intestinal tumor initiation in mice. Gastroenterology 2000;119:1045-53. [DOI] [PubMed] [Google Scholar]
- 16.Chung DC. The genetic basis of colorectal cancer: insights into critical pathways of tumorigenesis. Gastroenterology 2000;119:854-65. [DOI] [PubMed] [Google Scholar]
- 17.Smith G, Carey FA, Beattie J, et al. Mutations in APC, Kirsten-ras, and p53 - alternative genetic pathways to colorectal cancer. Proc Natl Acad Sci 2002;99:9433-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kameyama H, Nagahashi M, Shimada Y, et al. Genomic characterization of colitis-associated colorectal cancer. World J Surg Oncol 2018;16:121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Beaugerie L, Itzkowitz SH. Cancers complicating inflammatory bowel disease. N Engl J Med 2015;372:1441-52. [DOI] [PubMed] [Google Scholar]
- 20.Beaugerie L, Svrcek M, Seksik P, et al. Risk of colorectal high-grade dysplasia and cancer in a prospective observational cohort of patients with inflammatory bowel disease. Gastroenterology 2013;145:166-75.e8. [DOI] [PubMed] [Google Scholar]
- 21.Herrinton LJ, Liu L, Levin TR, et al. Incidence and mortality of colorectal adenocarcinoma in persons with inflammatory bowel disease from 1998 to 2010. Gastroenterology 2012;143:382-9. [DOI] [PubMed] [Google Scholar]
- 22.Nishikawa M, Oshitani N, Matsumoto T, et al. Accumulation of mitochondrial DNA mutation with colorectal carcinogenesis in ulcerative colitis. Br J Cancer 2005;93:331-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hsu CW, Sowers ML, Hsu W, et al. How does inflammation drive mutagenesis in colorectal cancer? Trends Cancer Res 2017;12:111. [PMC free article] [PubMed] [Google Scholar]
- 24.Liu P-H, Wu K, Ng K, et al. Association of obesity with risk of early-onset colorectal cancer among women. JAMA Oncol 2019;5:37-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bordonaro M, Lazarova D. Hypothesis: obesity is associated with a lower mutation threshold in Colon Cancer. J Cancer 2015;6:825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Stone TW, McPherson M, Darlington LG. Obesity and cancer: existing and new hypotheses for a causal connection. EBioMed 2018;30:14-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jasperson KW, Tuohy TM, Neklason DW, Burt RW. Hereditary and familial colon cancer. Gastroenterology 2010;138:2044-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Laurent-Puig P, Beroud C, Soussi T. APC gene: database of germline and somatic mutations in human tumors and cell lines. Nucleic Acids Res 1998;26:269-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Galiatsatos P, Foulkes WD. Familial adenomatous polyposis. Am J Gastroenterol 2006;101:385-98. [DOI] [PubMed] [Google Scholar]
- 30.Tudyka VN, Clark SK. Surgical treatment in familial adenomatous polyposis. Ann Gastroenterol 2012;25:201. [PMC free article] [PubMed] [Google Scholar]
- 31.Bisgaard ML, Fenger K, Bulow S, et al. Familial adenomatous polyposis (FAP): frequency, penetrance, and mutation rate. Hum Mutat 1994;3:121-5. [DOI] [PubMed] [Google Scholar]
- 32.Mori Y, Nagse H, Ando H, et al. Somatic mutations of the APC gene in colorectal tumors: mutation cluster region in the APC gene. Hum Mol Genet 1992;1:229-33. [DOI] [PubMed] [Google Scholar]
- 33.Lamlum H, Ilyas M, Rowan A, et al. The type of somatic mutation at APC in familial adenomatous polyposis is determined by the site of the germline mutation: a new facet to Knudson’s’ two-hit’hypothesis. Nature Med 1999;5:1071-5. [DOI] [PubMed] [Google Scholar]
- 34.Rowan A, Lamlum H, Ilyas M, et al. APC mutations in sporadic colorectal tumors: A mutational “hotspot” and interdependence of the “two hits”. Proc Natl Acad Sci 2000;97:3352-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Esplin ED, Snyder MP. Genomic era diagnosis and management of hereditary and sporadic colon cancer. World J Clin Oncol 2014;5:1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Spirio LN, Samowitz W, Robertson J, et al. Alleles of APC modulate the frequency and classes of mutations that lead to colon polyps. Nature Genet 1998;20:385-8. [DOI] [PubMed] [Google Scholar]
- 37.Charifa A, Jamil RT, Zhang X. Gardner syndrome. StatPearls [Internet]; 2020. [PubMed] [Google Scholar]
- 38.Khattab A, Monga DK. Turcot syndrome. StatPearls [Internet]; 2020. [PubMed] [Google Scholar]
- 39.Paraf F, Jothy S, Van Meir E. Brain tumor-polyposis syndrome: two genetic diseases? J Clin Oncol 1997;15:2744-58. [DOI] [PubMed] [Google Scholar]
- 40.Kim MJ, Huang Y, Park J-I. Targeting wnt signaling for gastrointestinal cancer therapy: Present and evolving views. Cancers 2020;12:3638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Andre T. Multicenter international study of oxaliplatin/5-fluorouracil/ leukovorin in the adjuvant treatment of colon cancer (MOSAIC) investigators: oxaliplatin, fluorouracil and leukovorin as adjuvant treatment for colon cancer. N Engl J Med 2004;50:2243-51. [Google Scholar]
- 42.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin 2019;69:7-34. [DOI] [PubMed] [Google Scholar]
- 43.Siegel RL, Torre LA, Soerjomataram I, et al. Global patterns and trends in colorectal cancer incidence in young adults. Gut 2019;68:2179-85. [DOI] [PubMed] [Google Scholar]
- 44.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7-30. [DOI] [PubMed] [Google Scholar]
- 45.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin 2021;71:7-33. [DOI] [PubMed] [Google Scholar]
- 46.Aoki K, Taketo MM. Adenomatous polyposis coli (APC): a multi-functional tumor suppressor gene. J Cell Sci 2007;120:3327-35. [DOI] [PubMed] [Google Scholar]
- 47.Midgley CA, White S, Howitt R, et al. APC expression in normal human tissues. J Pathol 1997;181:426-33. [DOI] [PubMed] [Google Scholar]
- 48.Kielman MF, Rindapaa M, Gaspar C, et al. Apc modulates embryonic stem-cell differentiation by controlling the dosage of β-catenin signaling. Nature Genet 2002;32:594-605. [DOI] [PubMed] [Google Scholar]
- 49.Henderson BR, Fagotto F. The ins and outs of APC and β-catenin nuclear transport. EMBO Rep 2002;3:834-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rosin-Arbesfeld R, Cliffe A, Brabletz T, Bienz M. Nuclear export of the APC tumour suppressor controls β-catenin function in transcription. The EMBO J 2003;22:1101-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hamada F, Bienz M. The APC tumor suppressor binds to Cterminal binding protein to divert nuclear β-catenin from TCF. Develop Cell 2004;7:677-85. [DOI] [PubMed] [Google Scholar]
- 52.Fodde R, Edelmann W, Yang K, et al. A targeted chain-termination mutation in the mouse Apc gene results in multiple intestinal tumors. Proc Natl Acad Sci 1994;91:8969-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Batlle E, Henderson JT, Beghtel H, et al. β-Catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell 2002;111:251-63. [DOI] [PubMed] [Google Scholar]
- 54.Tetsu O, McCormick F. β-Catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 1999;398:422-6. [DOI] [PubMed] [Google Scholar]
- 55.He T-C, Sparks AB, Rago C, et al. Identification of c-MYC as a target of the APC pathway. Science 1998;281:1509-12. [DOI] [PubMed] [Google Scholar]
- 56.Rubinfeld B, Albert I, Porfiri E, et al. Binding of GSK3β to the APC-β-catenin complex and regulation of complex assembly. Science 1996;272:1023-6. [DOI] [PubMed] [Google Scholar]
- 57.Kemler R. From cadherins to catenins: cytoplasmic protein interactions and regulation of cell adhesion. Trends Genet 1993;9:317-21. [DOI] [PubMed] [Google Scholar]
- 58.Bienz M, Hamada F. Adenomatous polyposis coli proteins and cell adhesion. Curr Opin Cell Biol 2004;16:528-35. [DOI] [PubMed] [Google Scholar]
- 59.Kawasaki Y, Sato R, Akiyama T. Mutated APC and Asef are involved in the migration of colorectal tumour cells. Nature Cell Biol 2003;5:211-5. [DOI] [PubMed] [Google Scholar]
- 60.Watanabe T, Wang S, Noritake J, et al. Interaction with IQGAP1 links APC to Rac1, Cdc42, and actin filaments during cell polarization and migration. Develop Cell 2004;7:871-83. [DOI] [PubMed] [Google Scholar]
- 61.Wen Y, Eng CH, Schmoranzer J, et al. EB1 and APC bind to mDia to stabilize microtubules downstream of Rho and promote cell migration. Nature Cell Biol 2004;6:820-30. [DOI] [PubMed] [Google Scholar]
- 62.Dikovskaya D, Khoudoli G, Newton IP, et al. The adenomatous polyposis coli protein contributes to normal compaction of mitotic chromatin. PLoS One 2012;7(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fodde R, Kuipers J, Rosenberg C, et al. Mutations in the APC tumour suppressor gene cause chromosomal instability. Nature Cell Biol 2001;3:433-8. [DOI] [PubMed] [Google Scholar]
- 64.Alberici P, de Pater E, Cardoso J, et al. Aneuploidy arises at early stages of Apc-driven intestinal tumorigenesis and pinpoints conserved chromosomal loci of allelic imbalance between mouse and human. Am J Pathol 2007;170:377-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wang Y, Azuma Y, Moore D, et al. Interaction between tumor suppressor adenomatous polyposis coli and topoisomerase IIα: implication for the G2/M transition. Mol Biol Cell 2008;19:4076-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Heinen CD, Goss KH, Cornelius JR, et al. The APC tumor suppressor controls entry into S-phase through its ability to regulate the cyclin D/RB pathway. Gastroenterology 2002;123:751-63. [DOI] [PubMed] [Google Scholar]
- 67.Jaiswal AS, Narayan S. A novel function of adenomatous polyposis coli (APC) in regulating DNA repair. Cancer Lett 2008;271:272-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Dimova DK, Dyson NJ. The E2F transcriptional network: old acquaintances with new faces. Oncogene 2005;24:2810-26. [DOI] [PubMed] [Google Scholar]
- 69.Ishidate T, Matsumine A, Toyoshima K, Akiyama T. The APC-hDLG complex negatively regulates cell cycle progression from the G0/G1 to S phase. Oncogene 2000;19:365-72. [DOI] [PubMed] [Google Scholar]
- 70.Bhattacharjee RN, Hamada F, Toyoshima K, Akiyama T. The tumor suppressor gene product APC is hyperphosphorylated during the M phase. Biochem Biophys Res Commun 1996;220:192-5. [DOI] [PubMed] [Google Scholar]
- 71.Marcote MJ, Pagano M, Draetta G. cdc2 protein kinase: structure -function relationships. Regul Eukar Cell Cycle 1992:30-41. [DOI] [PubMed] [Google Scholar]
- 72.Kaplan KB, Burds AA, Swedlow JR, et al. A role for the adenomatous polyposis coli protein in chromosome segregation. Nature Cell Biol 2001;3:429-32. [DOI] [PubMed] [Google Scholar]
- 73.Yamamoto H, Monden T, Miyoshi H, et al. Cdk2/cdc2 expression in colon carcinogenesis and effects of cdk2/cdc2 inhibitor in colon cancer cells. Int J Oncol 1998;13:233-42. [DOI] [PubMed] [Google Scholar]
- 74.Murphy KJ, Nielson KR, Albertine KH. Defining a molecularly normal colon. J Histochem Cytochem 2001;49:667-8. [DOI] [PubMed] [Google Scholar]
- 75.Liu Y, Prasad R, Beard WA, et al. Coordination of steps in single- nucleotide base excision repair mediated by apurinic/apyrimidinic endonuclease 1 and DNA polymerase β. J Biol Chem 2007;282:13532-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Oshima M, Oshima H, Kitagawa K, et al. Loss of Apc heterozygosity and abnormal tissue building in nascent intestinal polyps in mice carrying a truncated Apc gene. Proc Natl Acad Sci 1995;92:4482-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bromberg J, Wang TC. Inflammation and cancer: IL-6 and STAT3 complete the link. Cancer Cell 2009;15:79-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Taniguchi K, Moroishi T, De Jong PR, et al. YAP–IL-6ST autoregulatory loop activated on APC loss controls colonic tumorigenesis. Proc Natl Acad Sci 2017;114:1643-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Parsons SJ, Parsons JT. Src family kinases, key regulators of signal transduction. Oncogene 2004;23:7906-9. [DOI] [PubMed] [Google Scholar]
- 80.Smith ZD, Meissner A. DNA methylation: roles in mammalian development. Nat Rev Genet 2013;14:204-20. [DOI] [PubMed] [Google Scholar]
- 81.Sheaffer KL, Elliott EN, Kaestner KH. DNA hypomethylation contributes to genomic instability and intestinal cancer initiation. Cancer Prev Res (Phila) 2016;9:534-46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Arnold CN, Goel A, Niedzwiecki D, et al. APC promoter hypermethylation contributes to the loss of APC expression in colorectal cancers with allelic loss on 5q. Cancer Biol Ther 2004;3:960-4. [DOI] [PubMed] [Google Scholar]
- 83.Srivastava S, Dewangan J, Mishra S, et al. Piperine and Celecoxib synergistically inhibit colon cancer cell proliferation via modulating Wnt/β-catenin signaling pathway. Phytomedicine 2021;84:153484. [DOI] [PubMed] [Google Scholar]
- 84.Rather RA, Bhagat M. Cancer chemoprevention and piperine: molecular mechanisms and therapeutic opportunities. Front Cell Develop Biol 2018;6:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Oshima M, Dinchuk JE, Kargman SL, et al. Suppression of intestinal polyposis in ApcΔ716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell 1996;87:803-9. [DOI] [PubMed] [Google Scholar]
- 86.Dubois RN. Role of inflammation and inflammatory mediators in colorectal cancer. Trans Am Clin Climatol Assoc 2014;125:358. [PMC free article] [PubMed] [Google Scholar]
- 87.Bi X, Pohl N, Dong H, Yang W. Selenium and sulindac are synergistic to inhibit intestinal tumorigenesis in Apc/p21 mice. J Hematol Oncol 2013;6:8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Theard D, Raspe MA, Kalicharan D, et al. Formation of Ecadherin/ β-catenin-based Adherens junctions in hepatocytes requires serine-10 in p27 (Kip1). Mol Biol Cell 2008;19:1605-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Sadot E, Geiger B, Oren M, Ben-Ze’ev A. Down-regulation of β-catenin by activated p53. Mol Cell Biol 2001;21:6768-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Zilberberg A, Lahav L, Rosin-Arbesfeld R. Restoration of APC gene function in colorectal cancer cells by aminoglycoside- and macrolide-induced read-through of premature termination codons. Gut 2010;59:496-507. [DOI] [PubMed] [Google Scholar]
- 91.Zilberberg A, Lahav L, Rosin-Arbesfeld R. Restoration of APC gene function in colorectal cancer cells by aminoglycoside- and macrolide-induced read-through of premature termination codons. Gut 2010;59:496-507. [DOI] [PubMed] [Google Scholar]
- 92.Hamoya T, Miyamoto S, Tomono S, et al. Chemopreventive effects of a low-side-effect antibiotic drug, erythromycin, on mouse intestinal tumors. J Clin Biochem Nutr 2017;60:199-207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Floquet C, Rousset J-P, Bidou L. Readthrough of premature termination codons in the adenomatous polyposis coli gene restores its biological activity in human cancer cells. PLoS One 2011;6(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Macnab SA, Turrell SJ, Carr IM, et al. Herpesvirus saimirimediated delivery of the adenomatous polyposis coli tumour suppressor gene reduces proliferation of colorectal cancer cells. Int J Oncol 2011;39:1173-81. [DOI] [PubMed] [Google Scholar]
- 95.Bangalore S, Kumar S, Kjeldsen SE, et al. Antihypertensive drugs and risk of cancer: network meta-analyses and trial sequential analyses of 324 168 participants from randomised trials. Lancet Oncol 2011;12:65-82. [DOI] [PubMed] [Google Scholar]
- 96.Luo Y, Ohmori H, Shimomoto T, et al. Anti-angiotensin and hypoglycemic treatments suppress liver metastasis of colon cancer cells. Pathobiology 2011;78:285-90. [DOI] [PubMed] [Google Scholar]
- 97.Kuniyasu H. Multiple roles of angiotensin in colorectal cancer. World J Clin Oncol 2012;3:150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Dougherty U, Mustafi R, Haider HI, et al. Losartan and vitamin D inhibit colonic tumor development in a conditional Apc-deleted mouse model of sporadic colon cancer. Cancer Prevent Res 2019;12:433-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Ballou LM, Lin RZ. Rapamycin and mTOR kinase inhibitors. J Chem Biol 2008;1:27-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hardiman KM, Liu J, Feng Y, et al. Rapamycin inhibition of polyposis and progression to dysplasia in a mouse model. PLoS One 2014;9:e96023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Houghton PJ. Everolimus. Clin Cancer Res 2010;16:1368-72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Fujishita T, Aoki K, Lane HA, et al. Inhibition of the mTORC1 pathway suppresses intestinal polyp formation and reduces mortality in ApcΔ716 mice. Proc Natl Acad Sci 2008;105:13544-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Safaeian M, Robbins H, Berndt S, et al. Risk of colorectal cancer after solid organ transplantation in the United States. Am J Transplant 2016;16:960-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ling G, Lamprecht S, Shubinsky G, et al. Mycophenolate mofetil alone and in combination with tacrolimus inhibits the proliferation of HT-29 human colonic adenocarcinoma cell line and might interfere with colonic tumorigenesis. Anticancer Res 2018;38:3333-9. [DOI] [PubMed] [Google Scholar]
- 105.Pinsk V, Pinsk I, Ling G, et al. Complete reversion of familial adenomatous polyposis phenotype associated with tacrolimus and mycophenolate mofetil treatment following kidney transplantation. Anticancer Res 2017;37:3105-9. [DOI] [PubMed] [Google Scholar]
- 106.Corvinus FM, Orth C, Moriggl R, et al. Persistent STAT3 activation in colon cancer is associated with enhanced cell proliferation and tumor growth. Neoplasia (New York, NY) 2005;7:545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ou C, Sun Z, Li S, et al. Dual roles of yes-associated protein (YAP) in colorectal cancer. Oncotarget 2017;8:75727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Xiu M-X, Liu Y-M, Kuang B-H. The oncogenic role of Jagged1/Notch signaling in cancer. Biomed Pharmacother 2020;129:110416. [DOI] [PubMed] [Google Scholar]
- 109.Xu L, Lin W, Wen L, Li G. Lgr5 in cancer biology: functional identification of Lgr5 in cancer progression and potential opportunities for novel therapy. Stem Cell Res Ther 2019;10:219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Zhang Z, Bu X, Chen H, et al. Bmi-1 promotes the invasion and migration of colon cancer stem cells through the downregulation of E-cadherin. Int J Mol Med 2016;38:1199-207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Chen L, Chan SW, Zhang X, et al. Structural basis of YAP recognition by TEAD4 in the hippo pathway. Genes Develop 2010;24:290-300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Taniguchi K, Wu L-W, Grivennikov SI, et al. A gp130–Src– YAP module links inflammation to epithelial regeneration. Nature 2015;519:57-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Deng G, Song G-A, Pong E, et al. Promoter methylation inhibits APC gene expression by causing changes in chromatin conformation and interfering with the binding of transcription factor CCAAT-binding factor. Cancer Res 2004;64:2692-8. [DOI] [PubMed] [Google Scholar]
- 114.Seshagiri S, Stawiski EW, Durinck S, et al. Recurrent Rspondin fusions in colon cancer. Nature 2012;488:660-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Madan B, Ke Z, Harmston N, et al. Wnt addiction of genetically defined cancers reversed by PORCN inhibition. Oncogene 2016;35:2197-207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.El-Khoueiry AB, Ning Y, Yang D, et al. A phase I first-inhuman study of PRI-724 in patients (pts) with advanced solid tumors. American Society of Clinical Oncology; 2013. [Google Scholar]
- 117.Jiang S, Zhang M, Sun J, Yang X. Casein kinase 1α: biological mechanisms and theranostic potential. Cell Commun Signal 2018;16:1-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Richter J, Kretz A-L, Lemke J, et al. CK1α overexpression correlates with poor survival in colorectal cancer. BMC Cancer 2018;18:1-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Schittek B, Sinnberg T. Biological functions of casein kinase 1 isoforms and putative roles in tumorigenesis. Mol Cancer 2014;13:231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Duan S, Skaar JR, Kuchay S, et al. mTOR generates an autoamplification loop by triggering the βTrCP-and CK1α-dependent degradation of DEPTOR. Mol Cell 2011;44:317-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kronke J, Fink EC, Hollenbach PW, et al, et al. Lenalidomide induces ubiquitination and degradation of CK1α in del (5q) MDS. Nature 2015;523:183-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Honaker Y, Piwnica-Worms H. Casein kinase 1 functions as both penultimate and ultimate kinase in regulating Cdc25A destruction. Oncogene 2010;29:3324-34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Huang S-MA, Mishina YM, Liu S, et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 2009;461:614-20. [DOI] [PubMed] [Google Scholar]
- 124.Lau T, Chan E, Callow M, et al. A novel tankyrase small-molecule inhibitor suppresses APC mutation–driven colorectal tumor growth. Cancer Res 2013;73:3132-44. [DOI] [PubMed] [Google Scholar]
- 125.Jeong W-J, Ro EJ, Choi K-Y. Interaction between Wnt/β- catenin and RAS-ERK pathways and an anti-cancer strategy via degradations of β-catenin and RAS by targeting the Wnt/β-catenin pathway. NPJ Precision Oncol 2018;2:1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Van der Flier LG, Sabates–Bellver J, Oving I, et al. The intestinal Wnt/TCF signature. Gastroenterology 2007;132:628-32. [DOI] [PubMed] [Google Scholar]
- 127.Korinek V, Barker N, Moerer P, et al. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nature Genetics 1998;19:379-83. [DOI] [PubMed] [Google Scholar]
- 128.Mahmoudi T, Li VS, Ng SS, et al. The kinase TNIK is an essential activator of Wnt target genes. The EMBO J 2009;28:3329-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Shitashige M, Satow R, Jigami T, et al. Traf2-and Nck-interacting kinase is essential for Wnt signaling and colorectal cancer growth. Cancer Res 2010;70:5024-33. [DOI] [PubMed] [Google Scholar]
- 130.Masuda M, Uno Y, Ohbayashi N, et al. TNIK inhibition abrogates colorectal cancer stemness. Nature Commun 2016;7:12586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Li G, Su Q, Liu H, et al. Frizzled7 promotes epithelial-to-mesenchymal transition and stemness via activating canonical Wnt/β-catenin pathway in gastric cancer. Int J Biol Sci 2018;14:280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Flanagan DJ, Barker N, Di Costanzo NS, et al. Frizzled-7 is required for Wnt signaling in gastric tumors with and without Apc mutations. Cancer Res 2019;79:970-81. [DOI] [PubMed] [Google Scholar]
- 133.Jimeno A, Gordon M, Chugh R, et al. A first-in-human phase I study of the anticancer stem cell agent ipafricept (OMP- 54F28), a decoy receptor for Wnt ligands, in patients with advanced solid tumors. Clinical Cancer Res 2017;23:7490-7. [DOI] [PubMed] [Google Scholar]
- 134.Pinzone JJ, Hall BM, Thudi NK, et al. The role of Dickkopf- 1 in bone development, homeostasis, and disease. Blood 2009;113:517-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Shen Q, Yang X-R, Tan Y, et al. High level of serum protein DKK1 predicts poor prognosis for patients with hepatocellular carcinoma after hepatectomy. Hepat Oncol 2015;2:231-44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Yu B, Yang X, Xu Y, et al. Elevated expression of DKK1 is associated with cytoplasmic/nuclear β-catenin accumulation and poor prognosis in hepatocellular carcinomas. J Hepatol 2009;50:948-57. [DOI] [PubMed] [Google Scholar]
- 137.Lyros O, Lamprecht A-K, Nie L, et al. Dickkopf-1 (DKK1) promotes tumor growth via Akt-phosphorylation and independently of Wnt-axis in Barrett’s associated esophageal adenocarcinoma. Am J Cancer Res 2019;9:330. [PMC free article] [PubMed] [Google Scholar]
- 138.Liu Y, Tang W, Xie L, et al. Prognostic significance of dickkopf- 1 overexpression in solid tumors: a meta-analysis. Tumor Biol 2014;35:3145-54. [DOI] [PubMed] [Google Scholar]
- 139.Sui Q, Zheng J, Liu D, et al. Dickkopf-related protein 1, a new biomarker for local immune status and poor prognosis among patients with colorectal liver oligometastases: a retrospective study. BMC Cancer 2019;19:1-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Dong L-L, Qu L-Y, Chu L-Y, et al. Serum level of DKK-1 and its prognostic potential in non–small cell lung cancer. Diagnost Pathol 2014;9:52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Wall JA, Klempner SJ, Arend RC. The anti-DKK1 antibody DKN-01 as an immunomodulatory combination partner for the treatment of cancer. Exp Opin Investig Drugs 2020. [In press]. [DOI] [PubMed] [Google Scholar]
- 142.Betella I, Turbitt WJ, Szul T, et al. Wnt signaling modulator DKK1 as an immunotherapeutic target in ovarian cancer. Gynecol Oncol 2020. [In press]. [DOI] [PubMed] [Google Scholar]
- 143.Gonzalez-Sancho JM, Aguilera O, Garcia JM, et al. The Wnt antagonist DICKKOPF-1 gene is a downstream target of β- catenin/TCF and is downregulated in human colon cancer. Oncogene 2005;24:1098-103. [DOI] [PubMed] [Google Scholar]
- 144.Kuphal S, Lodermeyer S, Bataille F, et al. Expression of Dickkopf genes is strongly reduced in malignant melanoma. Oncogene 2006;25:5027-36. [DOI] [PubMed] [Google Scholar]
- 145.Aguilera O, Fraga MF, Ballestar E, et al. Epigenetic inactivation of the Wnt antagonist DICKKOPF-1 (DKK-1) gene in human colorectal cancer. Oncogene 2006;25:4116-21. [DOI] [PubMed] [Google Scholar]
- 146.Safari E, Mosayebi G, Khorram S. Dkk-3 as a potential biomarker for diagnosis and prognosis of colorectal cancer. Med J Islam Rep Iran 2018;32:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Dasari S, Tchounwou PB. Cisplatin in cancer therapy: molecular mechanisms of action. Eur J Pharmacol 2014;740:364-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Van Klompenberg MK, Bedalov CO, Soto KF, Prosperi JR. APC selectively mediates response to chemotherapeutic agents in breast cancer. BMC Cancer 2015;15:457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Kariv R, Caspi M, Fliss-Isakov N, et al. Resorting the function of the colorectal cancer gatekeeper adenomatous polyposis coli. Int J Cancer 2020;146:1064-74. [DOI] [PubMed] [Google Scholar]
- 150.Chan AT, Ogino S, Fuchs CS. Aspirin use and survival after diagnosis of colorectal cancer. JAMA 2009;302:649-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Moran AE, Hunt DH, Javid SH, et al. Apc deficiency is associated with increased Egfr activity in the intestinal enterocytes and adenomas of C57BL/6J-Min/+ mice. J Biol Chem 2004;279:43261-72. [DOI] [PubMed] [Google Scholar]
- 152.Samadder NJ, Kuwada SK, Boucher KM, et al. Association of sulindac and erlotinib vs placebo with colorectal neoplasia in familial adenomatous polyposis: secondary analysis of a randomized clinical trial. JAMA Oncol 2018;4:671-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Hawcroft G, Loadman PM, Belluzzi A, Hull MA. Effect of eicosapentaenoic acid on E-type prostaglandin synthesis and EP4 receptor signaling in human colorectal cancer cells. Neoplasia 2010;12:618-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.West NJ, Clark SK, Phillips RK, et al. Eicosapentaenoic acid reduces rectal polyp number and size in familial adenomatous polyposis. Gut 2010;59:918-25. [DOI] [PubMed] [Google Scholar]
- 155.Lynch PM, Burke CA, Phillips R, et al. An international randomised trial of celecoxib versus celecoxib plus difluoromethylornithine in patients with familial adenomatous polyposis. Gut 2016;65:286-95. [DOI] [PubMed] [Google Scholar]
- 156.Nowotarski SL, Woster PM, Casero RA, Polyamines and cancer: implications for chemotherapy and chemoprevention. Expert Rev Mol Med 2013;15:e3-e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Rahib L, Wehner MR, Matrisian LM, Nead KT. Estimated projection of US cancer incidence and death to 2040. JAMA Netw Open 2021;4:e214708-e. [DOI] [PMC free article] [PubMed] [Google Scholar]
