Next Article in Journal
Role of mTOR Complexes in Neurogenesis
Next Article in Special Issue
The Effect of GPRC5a on the Proliferation, Migration Ability, Chemotherapy Resistance, and Phosphorylation of GSK-3β in Pancreatic Cancer
Previous Article in Journal
Protein Tyrosine Phosphatase 1B Inhibition and Glucose Uptake Potentials of Mulberrofuran G, Albanol B, and Kuwanon G from Root Bark of Morus alba L. in Insulin-Resistant HepG2 Cells: An In Vitro and In Silico Study
Previous Article in Special Issue
EP4 as a Therapeutic Target for Aggressive Human Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Frizzled Receptors as Potential Therapeutic Targets in Human Cancers

1
Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Research Centre, Shenzhen University School of Medicine, Shenzhen 518060, China
2
Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310006, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2018, 19(5), 1543; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19051543
Submission received: 30 March 2018 / Revised: 12 May 2018 / Accepted: 19 May 2018 / Published: 22 May 2018
(This article belongs to the Special Issue Cancer-Driver G Protein-Coupled Receptors as Therapeutic Targets)

Abstract

:
Frizzled receptors (FZDs) are a family of seven-span transmembrane receptors with hallmarks of G protein-coupled receptors (GPCRs) that serve as receptors for secreted Wingless-type (WNT) ligands in the WNT signaling pathway. Functionally, FZDs play crucial roles in regulating cell polarity, embryonic development, cell proliferation, formation of neural synapses, and many other processes in developing and adult organisms. In this review, we will introduce the basic structural features and review the biological function and mechanism of FZDs in the progression of human cancers, followed by an analysis of clinical relevance and therapeutic potential of FZDs. We will focus on the development of antibody-based and small molecule inhibitor-based therapeutic strategies by targeting FZDs for human cancers.

Graphical Abstract

1. Introduction

The frizzled receptors (FZDs) are comprised of ten members (FZD1–FZD10). The common characteristics of these members include an N-terminal signal sequence followed by highly conserved cysteine-rich domain (CRD) in extracellular region, a seven-pass transmembrane domain and an intracellular C-terminal domain [1]. The WNT proteins, a kind of secreted glycoproteins and ligands of FZDs, are comprised of nineteen members which can bind to the cell surface complex consisted of two elements: a member of the FZD family and the co-receptor Low density lipoprotein receptor related protein 5/6 (LRP5/6). Other heterodimeric receptor complexes comprised of an FZD and the cell surface receptors encoded by the receptor tyrosine kinase like orphan receptor 2 (ROR2) and receptor-like tyrosine kinase (RYK) genes have also been described [2]. Subsequently, the intracellular C-terminal domain of FZDs interacts with the Dishevelled (DVL) protein to transduce the WNT signals to the downstream of the pathway [3]. Each member of FZDs can interact with several different WNT proteins to activate either the canonical WNT/β-catenin or the non-canonical WNT/PCP (planar cell polarity) and WNT/Ca2+ signaling pathways [4], thereby activating multiple downstream transcription factors that are important for stem cell regulation, embryonic development, cell polarity, proliferation and differentiation, and especially tumorigenesis [5,6]. To date, the selectivity between individual WNTs and FZDs remains poorly understood. Taking a panoramic view of the WNT signaling pathway, it is easy for us to draw a conclusion that FZDs serve as a core component in both canonical and non-canonical pathways. It is worth mentioning that the co-receptors also play a key role in forming a complex with FZDs for binding WNT ligands and are involved in tumorigenesis. Additionally, hetero-dimerization of FZDs and co-receptors is well-known. Nile et al. recently reported that the CRD of FZDs recognized WNT cis-unsaturated fatty acyl groups which bridged two CRD monomers and mediated FZD receptor dimerization [7]. Earlier, Carron et al. implied that FZD receptor dimerization contributed to transduction of WNT/β-catenin signaling [8].
In the past years, accumulating studies have demonstrated that activation of WNT signaling is crucial for both initiation and progression of human cancers. FZDs function as cell surface receptors of WNT signaling pathway. If FZDs are blocked by antibodies or small molecule inhibitors, the WNT signaling pathway will not be activated so that those downstream target genes (e.g., CCND1, MYC) will not be activated as well. Therefore, targeting FZDs might be a potential approach for cancer therapy.
In this review, we will shortly introduce the structural features of FZDs and present the current knowledge of biological function, mechanism and clinical relevance of FZDs. We aim to focus on the development of antibody-based and small molecule inhibitor-based therapeutic strategies by targeting FZDs for human cancers.

2. Structural Features of Frizzled Receptors

FZDs could be further divided into four subgroups according to their identity. FZD1, FZD2, and FZD7 share about 75% identity with each other, FZD4, FZD9, and FZD10 share 65% identity, FZD5 and FZD8 share 70% identity, FZD3 and FZD6 share 50% amino acid identity. The range of identities shared by receptors from different subgroups is from 20 to 40% [9]. The length of FZDs ranges from 500 to 700 amino acids. The extracellular CRD comprises of 120 to 125 amino residues with ten conserved cysteines and is followed by a hydrophilic linker region which contains 40 to 100 amino residues. The proteins also contain seven hydrophobic domains that are predicted to form transmembrane α-helices. The length of the intracellular C-terminal domain is variable and is not very conserved compared with each family member [10].
In addition to the basic structural features of FZDs, the common characteristics between FZDs and other GPCRs include the following: (1) the conserved cysteines in the extracellular loops 1 and 2 that are implicated in the formation of disulfide bonds and (2) a series of charged residues at the N- and C-terminal of intracellular loop 3 that are indispensable for the coupling of receptor and G protein. However, FZDs lack some domains common to other GPCRs, such as the DRY motif at the C-terminal end of the third transmembrane domain [11].

3. Biological Functions and Mechanism

The biological functions of FZDs have been elucidated in various cancers and in normal development. A lot of functions that promote cancer are mediated by FZDs including cancer cell proliferation, migration, invasion, angiogenesis, stemness, and chemoresistance after cancer recurrence. Notably, FZDs do not work through a single, well-known WNT signaling pathway but are intertwined with other signaling cascades.

3.1. FZD1

It has been reported that FZD1 silencing induced parallel strong decrease in the expression of MDR1 (multidrug resistance) gene with a significant restoration of drug sensitivity in neuroblastoma (NB) cancer cells, which confirmed the FZD1-associated chemoresistance [12]. Another study also demonstrated that interference of FZD1 reversed multidrug resistance in breast cancer cells through the WNT/β-catenin pathway with suppression of MDR1 [13]. Rosiglitazone, a member of the synthetic PPARγ (Peroxisome proliferator-activated receptor γ) ligands, suppressed the multidrug resistance in human ovarian cancer cells by downregulating the expression of FZD1 and MDR1 in a concentration-dependent manner [14]. A recent study has shown that downregulation of FZD1 by miR-135b reversed chemoresistance of non-small cell lung cancer (NSCLC) cells, further confirmed the association between FZD1 and chemoresistance [15]. Collectively, the biological function of FZD1 could be involved in modulating the sensitivity of various human cancers to chemotherapeutic drugs. During normal development, Lapointe et al. indicated that FZD1 is required for normal female fertility and may partly regulate cumulus cell function and oocyte maturation in mice [16].

3.2. FZD2

Gujral et al. revealed that FZD2 drives epithelial-mesenchymal transition (EMT) and metastasis through a previously unrecognized, non-canonical pathway that included FYN proto-oncogene (FYN), a Src family tyrosine kinase, and signal transducer and activator of transcription 3 (STAT3) transcriptional regulator in diverse solid tumors (Figure 1) [17]. Other studies also suggested that FZD2 contributed to the migration and invasion of human oral squamous cell carcinoma (OSCC) cells, at least partly through regulation of the STAT3 pathway [18]. FZD2 overexpression promoted migration by canonical WNT pathway and facilitated EMT phenotype in endometrial cancer (EC) cells [19]. One study has shown that FZD2 stimulated cell proliferation and promoted cell migration in high-risk NB by regulating β-catenin-dependent and β-catenin-independent signaling pathways. FZD2 blockade by siRNA inhibited NB cell proliferation and xenograft growth, reduced cell motility and induced a less vascularized phenotype [20]. However, one study showed that FZD2 inhibited the cell growth and migration in salivary adenoid cystic carcinomas (SACC), indicating that FZD2 may exert different functions in various cancers [21]. During normal development, Kadzik et al. showed that FZD2 is necessary for domain branch formation during the initial establishment of the respiratory tree and facilitates changes in epithelial cell length and shape in mice. Loss of FZD2 in the lung epithelium leads to the formation of distal cysts in the lung and disrupts the molecular branching program of the lung [22].

3.3. FZD3

FZD3 inhibited cell migration and activated cAMP/PKA/Gα(s) signaling pathway by interacting with WNT5a in breast cancer [23]. FZD3 mediated neurite outgrowth in the presence of WNT3a in Ewing tumor cells [24]. During normal development, FZD3 controls axonal polarity and intermediate target entry during striatal pathway development [25] and axonal development in distinct populations of cranial and spinal motor neurons [26]. FZD3-knockout mice show defects in striatal pathway development [25]. FZD3 is a receptor poorly studied among FZD family in human cancers. Previous studies focused on the role of FZD3 in the development of organisms. There is currently no explicit method or antibody that could be taken to target FZD3 for cancer therapy.

3.4. FZD4

Interaction of WNT3a with FZD4 activated WNT signaling pathway in acute myeloid leukemia. Functionally, FZD4 expression modulated apoptosis and enhanced WNT3a-induced β-catenin stability in myeloid progenitor cells [27]. Activation of WNT signaling by FZD4 was found in ERG [Erythroblast transformation-specific (ETS)-related gene]-positive prostate cancers. ERG, a member of ETS family of transcription factors, is an oncogene involved in chromosomal translocations and gene fusions. ERG often co-overexpressed with FZD4 in prostate cancer, which led to cancer-promoting phenotypic effects, such as EMT and loss of cell adhesion [28]. FZD4 not only mediated the invasive potential of glioma cells, but also promoted stemness of glioma cells by activating canonical WNT signaling [29]. Downregulation of FZD4 by miR-493 inhibited cell migration in human bladder cancer cells [30]. In cervical carcinoma, miR-505 acted as tumor inhibitor with inverse association with FZD4 [31]. During normal development, Xu et al. revealed that FZD4 is indispensable for vascular development in the retina, and the retinal vasculature of FZD4-knockout mice is defective and abnormal [32].

3.5. FZD5

FZD5 increased adhesion to extracellular matrix components including collagen IV, fibronectin and vitronectin in ovarian cancer cells [33]. Inhibition of FZD5/PKC signaling by miR-124 overcame P-glycoprotein-mediated multidrug resistance in renal cell carcinoma [34]. MiR-224 inhibited cell proliferation and migration by targeting FZD5 and inhibiting WNT/β-catenin signaling in breast cancer [35]. Knockout of FZD5 robustly inhibited cell growth in human Ring finger protein 43 (RNF43)-mutant pancreatic ductal adenocarcinoma cells (PDAC) in clonogenic growth assays, and editing of FZD5 significantly inhibited the expression of WNT target genes AXIN2 and NKD1(Naked cuticle homolog 1) [36]. As for the functions of FZD5 in normal development, Burns et al. uncovered that FZD5-mutant mice exhibit abnormal eye development, increased cell death and decreased retinal proliferation by the optic cup stage [37].

3.6. FZD6

FZD6-positive NB cells were resistant to doxorubicin and overexpressed mesenchymal markers such as Twist1 and Notch1 [38]. FZD6 also exerted a mediating role in β-carotene-induced uncontrolled proliferation of lung cells [39]. FZD6, targeted by miR-199a-5p and miR-21, played a significant role in the progression of colorectal cancer (CRC) [40]. However, FZD6 suppressed cell proliferation and migration in gastric cancer via the non-canonical WNT pathway [41]. FZD6 regulated fibronectin deposition and assembly of the actin cytoskeleton in MDA-MB-231 cells and metastatic process in vivo, but not proliferation of breast cancer cell lines [42]. One study argued that FZD6 activated CaMKII–TAK1–NLK signaling and inhibited WNT pathway activity through TAK1 while promoting STAT3 and NF-κB signaling (Figure 1) that are important regulators of the mesenchymal-associated phenotype in glioblastoma. Knockdown of endogenous FZD6 significantly decreased sphere formation and cell proliferation of mesenchymal spheres in vitro [43]. As for the functions of FZD6 in normal development, Cui et al. reported that keratin is significantly affected in FZD6-knockout mice. FZD6 is required for the differentiation process of nail development [44].

3.7. FZD7

FZD7 plays an important role in stem cell biology and cancer development and progression. Flanagan et al. reported that intestinal epithelium regeneration is impaired in FZD7-knockout mice. FZD7 is indispensable for the maintenance of intestinal epithelial Lgr5+ stem cells and the regeneration of Intestinal Organoids [5], which has been documented recently by Nile et al. by using a small peptide inhibitor dFz7-21 [45]. ΔNp63, an isoform of p63, regulates both normal mammary stem cell activity and tumor initiating activity of breast cancer by increasing FZD7 expression by binding in a conserved active enhancer region of FZD7. And FZD7 knockdown significantly reduced tumor sphere formation in patient derived xenograft tumor [46]. SOX8 and SOX9 induced the FZD7-mediated activation of the WNT/β-catenin pathway to regulate chemoresistance, stem-like properties and EMT in tongue squamous cell carcinoma (TSCC) [47] and hepatocellular carcinoma (HCC) [48]. FZD7 enhanced cell viability and survival, as well as inhibited apoptosis in lung cancer cell lines NCI-H446 and A549 [49]. MiR-142-3p suppressed proliferation and invasion of cervical cells by directly regulating FZD7 expression [50]. FZD7 promoted cell growth, mobility, chemoresistance, metastasis and EMT in esophageal squamous cell carcinoma (ESCC) [51,52]. FZD7 promoted glioma cell proliferation by upregulation of Tafazzin (TAZ) via the β-catenin/TCF-mediated transcription in glioma cells [53]. FZD7 increased ovarian cancer cell proliferation and aggressiveness in vitro via regulation of non-canonical WNT/PCP pathway [54]. Knockdown of FZD7 inhibited metastasis and EMT in cervical cancer cells and gastric cancer cells [55,56], and suppressed tumor transformation in triple negative breast cancer (TNBC) cell lines MDA-MB-231 and BT-20 [57]. FZD7 could also enhance invasion and metastatic capabilities of colon cancer cells through both non-canonical and canonical WNT signaling pathways [58]. Tiwary et al. found that FZD7 related to tumor initiation and metastasis in melanoma cells [59]. It is reported that Helicobacter pylori infection promotes FZD7 expression in gastric cancer cells [60]. To this date, FZD7 is the most studied member among FZD family in cancer research.

3.8. FZD8

Overexpression of FZD8 promoted, while silencing of FZD8 suppressed prostate cancer cell migration, invasion, stem cell-like phenotypes in vitro and bone metastasis in vivo by the activation of WNT/β-catenin signaling. In addition, wild-type p53 directly interacts with FZD8 promoter transcriptionally repressing FZD8 [61]. It has been reported that FZD8, targeted by long noncoding RNA AK126698, activated by WNT2 ligand and downregulated by miR-100 or miR-520b, could promote cell proliferation, migration, and invasion via activation of WNT/β-catenin pathway in non-small cell lung cancer, spinal osteosarcoma and breast cancer [62,63,64,65]. In addition, FZD8 was found to mediate the interaction of c-Met and WNT/β-catenin signaling, rescue the effects of c-Met inhibition and increase the tumor-initiating ability in cancer stem-like cells of head and neck squamous carcinoma [66]. FZD8 expression was reported to be upregulated after Cisplatin plus TRAIL [Tumor necrosis factor (TNF)-related apoptosis-inducing ligand] treatment in TNBC cells, and inhibition of FZD8 inhibition reduced β-catenin and survivin levels that led to increased apoptosis, indicating that FZD8 plays an important role in drug resistance in TNBC [67]. However, more recent study showed that downregulation of FZD8 expression by K-Ras resulted in a sustained suppression of non-canonical WNT/Ca2+ signaling, which led to increased tumorigenicity [68]. In lung cancer, knockdown of FZD8 significantly downregulated the expression of both cyclin D1 and survivin, inhibited cell proliferation and sensitized cell to taxotere treatment in vitro [69].

3.9. FZD9

FZD9 expression was reported to be upregulated in astrocytoma [70] and osteosarcoma [71]. FZD9 may relate to angiogenesis in human astrocytoma [70]. FZD9 knockdown inhibited cell proliferation, motility and cyclin D1 expression in HCC and hepatoblastoma cell lines [72].
However, FZD9 was downregulated in acute myeloid leukemia due to the promoter methylation, suggesting it may also function as a tumor suppressor [73]. It has been reported that the direct interaction of WNT7a ligand and its receptor FZD9 repressed cell growth and promoted cell differentiation in NSCLC, indicating an antitumor effect of WNT7a and FZD9 in human cancers [74,75]. Therefore, FZD9 may not be the best target for cancer therapy due to its dual character. During normal development, knockout of FZD9 results in hippocampal and visuospatial learning defects and abnormal B cell development in mice [76,77].

3.10. FZD10

FZD10 was involved in the progression of synovial sarcoma by regulating actin reorganization and anchorage-independent cell growth [78]. It has been reported that FZD10 is a direct target of SS18-SSX2 which is an oncogenic fusion protein in synovial sarcoma [79]. Nagayama et al. found a strong inverse correlation between FZD10 expression and nuclear β-catenin accumulation in synchronous colorectal tumors, indicating that FZD10 may exert functions via non-canonical WNT signaling pathway [80]. FZD10 overexpression in breast cancer cells due to reduced breast cancer metastasis suppressor 1 like (BRMS1L) level led to aberrant activation of canonical WNT signaling and thus induced EMT and promoted metastasis [81]. Hypoxia-inducible protein-2 (HIG2) was reported to bind to the extracellular domain of FZD10 and activated oncogenic WNT signaling in renal cell carcinoma (RCC) [82].

4. Clinical Relevance

There is some evidence that link the overexpression of FZD receptors to poor prognosis in human cancers. Previous studies have demonstrated that FZD receptors were frequently overexpressed in tumor tissues relative to normal tissues. Here, we present the clinical relevance of each FZD member in various cancers (Table 1).

5. Therapeutic Potential of FZD Receptors

Disruption of the WNT signaling pathway by blocking FZD receptors provides possibility for rational cancer therapy. Antibody-based and small molecule inhibitor-based therapeutic strategies against the overexpressed FZD receptors could be a translatable approach to overcome cancer. Fukukawa et al. previously reported that FZD10 was significantly transactivated in synovial sarcoma, but not expressed in normal human organs except placenta [85]. Therefore, they generated a murine monoclonal antibody (mAb) named mAb 92-13 that could specifically bind to FZD10 in vitro and vivo, and was internalized into and accumulated in cells expressing FZD10. Moreover, a single intravenous (i.v.) injection of radioisotope-labeled mAb 92-13 strongly suppressed growth of synovial sarcoma in mice without any obvious toxicity [86]. Another study displayed that an anti-FZD7 antibody was able to induce cell death and deplete stem cell properties in FZD7-positve Wilms’ tumor, and reduce in vivo cell proliferation and survival in mouse xenograft model. Moreover, the exogenous expression of two kinds of WNT antagonists, sFRP1 (secreted Frizzled-related protein 1) and DKK1 (Dickkopf WNT signaling pathway inhibitor 1), could promote the sensitivity of Wilms’ tumor cells to the antibody treatment, indicating that a combination of anti-FZDs antibodies and WNT inhibitors may greatly benefit cancer therapy [87]. In addition, an anti-FZD7 antibody with nanoshell was reported to display higher binding affinity for FZD7, increased WNT inhibitory effects and decreased cell viability compared with freely delivered antibody in TNBC cells due to multivalency, which indicated that higher binding affinity is related to higher signaling inhibition and higher therapeutic effects [88]. Nickho and colleagues have isolated single chain variable fragments (scFvs) against FZD7. These scFvs antibodies contain variable domains of heavy and light chains that were linked by an artificial flexible linker, and bind to specific peptide of extracellular domain of FZD7. Functionally, the scFvs inhibited cell growth by inducing cell apoptosis in a TNBC cell line, MDA-MB-231. The reasons why scFvs antibodies are potential therapeutic agents include small size, high affinity, human origin, and specificity [89,90].
OMP-18R5, generated by Gurney et al., was a monoclonal antibody that could interact with FZD1, 2, 5, 7, and 8 by binding to the conserved epitope of extracellular domain of FZD receptors and thereby prevented WNT ligands from binding to FZD receptors, which directly inactivated the canonical WNT pathway. Functionally, OMP-18R5 treatment triggered a reduced growth of human tumor xenografts in mice in several types of human cancers, including breast, pancreatic, colon, and lung tumors [91]. And it was found that combined treatment of OMP-18R5 and several standard-of-care chemotherapeutic agents including Taxol, Irinotecan and Gemcitabine showed striking synergy and significantly reduced cell proliferation and tumorigenicity in both pancreatic and breast tumors [91]. In general, this antibody-based therapeutic strategy by targeting FZDs may be beneficial to the treatment of a broad range of human cancers, and OMP-18R5 currently is in phase I trial [92]. However, one thing to note is that the side effects of OMP-18R5 need to be further evaluated due to its universality and non-specificity.
In a recent study, a small molecule compound SRI37892 was screened to target the transmembrane domain of FZD7 thereby blocking the WNT signal transmission. SRI37892 treatment inhibited LRP6 phosphorylation and downregulated the level of cytosolic free β-catenin, and functionally repressed breast cancer cell viability and colony formation. Therefore, targeting the transmembrane domain of FZD receptors could be a potential therapeutic strategy [93].
Nambotin et al. previously reported that FZD7 was overexpressed in HCC and mediated cancer phenotype. Hence, they have designed small interfering peptides named Rhodamine-tagged heptapeptide protein transduction domain for binding DVL (RHPDs) that were able to disrupt the binding of FZD7 and the PDZ domain of DVL. In vitro, the viability of human HCC cell lines was decreased through apoptosis after RHPDs treatment. In vivo, RHPD-P1, a type of RHPDs, was injected intratumorously in HCC mouse model. It was revealed that RHPD-P1 inhibited tumor growth, which displayed the anti-tumor effect of this small interfering peptide [94]. FJ9, a small molecule inhibitor, was also found to exert the same function as RHPDs to disrupt the interaction between the C-terminal tail of FZD7 and the PDZ domain of DVL. FJ9 induced apoptosis in melanoma cell line and non-small cell lung cancer cell line, triggered a reduction of tumor growth in mouse xenograft model [95]. Therefore, small molecule inhibitors that antagonize protein-protein interactions in oncogenic signaling pathways show promise in treating cancer.
dFz7-21, a selective peptide of the FZD7 receptor subclass including FZD1, FZD2, and FZD7, inhibited WNT signaling by binding to the FZD7 CRD subclass at a new site proximal to the lipid-binding groove and altering the dimer interface in turn disrupting the formation of the WNT–FZD–LRP ternary complex. The drastic conformational changes of dFz7-21-FZD7-CRD complex may make the FZD receptor incompetent for ternary complex formation and proper signaling. Although the anti-cancer effect of dFz7-21 has not been demonstrated in this paper, dFz7-21 was able to block Lgr5+ stem cell function, which indicated its potential ability to block FZD7 subclass in cancers [45].
Zachary et al. developed anti-FZD5 antibodies IgG-2919 and IgG-2921. A dose-dependent anti-proliferative effect was observed when treating RNF43-mutant PDAC cell lines HPAF-II, PaTu8988S and AsPC-1 with the FZD5 IgGs, but no obvious effect observed in non-RNF43-mutant PDAC cells. Furthermore, FZD5 antibodies-induced inhibition of RNF43-mutant PDAC cells in vitro was through G0/G1 cell-cycle arrest but not through apoptosis. Experiments in vivo in immunodeficient mice were also performed, 74% and 60% growth inhibition were observed respectively in HPAF-II and AsPC-1 orthotopic tumors treated with 2 mg/kg IgG-2919 compared with control tumors over a 32-days experiment. More importantly, there were no weight loss or visual signs of toxicity. Besides, FZD5 antibodies also inhibited the growth of RNF43-mutant CRC-patient-derived organoids suggesting that FZD5 may be associated with RNF43 mutation across multiple cancer types, expanding the use of FZD5 antibody in cancer treatment [36].
OMP-54F28, a recombinant protein formed by the fusion of the immunoglobulin Fc to the CRD of FZD8, was undergoing clinical trials. OMP-54F28 serves as a decoy receptor competing with FZD8 for WNT ligands. This recombinant protein has been shown to inhibit tumor growth, synergize with chemotherapeutic drugs and decrease frequency of cancer stem cell by blocking the WNT signaling pathway in preclinical models. Recently, a first-in-human phase 1 study of OMP-54F28 was performed in patients with advanced solid tumors. The results showed that the toxicities of OMP-54F28 were mild, and included fatigue, taste alterations, decreased appetite and muscle cramps. Although no notable anti-cancer responses with single OMP-54F28 were observed, the joint treatment of OMP-54F28 and chemotherapeutic drugs may facilitate the anti-cancer activity [96,97,98]. Similarly, soluble FZD7 (sFZD7) created by utilization of the extracellular peptide of FZD7 by Wei et al. also serves as a decoy receptor. sFZD7 was deemed to bind to extracellular WNT ligands, thereby reducing the combination of ligands and FZD7 and inhibiting WNT/β-catenin signaling in HCC. After sFZD7 treatment, the viability of hepatoma cells decreased but normal hepatocytes were uninfluenced, which is the crucial for clinical trials. Moreover, sFZD7 increased the sensitivity of HCC cells to doxorubicin in vitro [99]. And previously, several secreted WNT pathway antagonists have been identified: WNT inhibitor factor (WIF) proteins can interact with WNT ligands; DKK has been reported to bind to LRP5/6; SFRPs, a kind of proteins that share sequence with the CRD of FZDs, compete with FZDs for WNT ligands [100]. From what we have reviewed above, the method of blocking the upstream WNT signal by preventing the binding of a ligand to a receptor looks as effective as directly targeting FZDs.
In addition to FZDs, targeting other components of WNT signaling pathway is not seemingly easy, because they have extra functions. For instance, Adenomatous Polyposis Coli (APC) is not only a part of the destruction complex that limits the level of β-catenin, but also was reported to have a role in directed cell migration [101]. A recent study indicated that APC also contributes to the inhibition of WNT receptor activation independently of WNT ligand [102]. E-cadherin, a protein related to cell adhesion, also can bind to β-catenin to form a complex anchored in cell membrane which ensure the stability of cell-cell junction [103,104]. Thus, if these components are targeted, it’s not just WNT pathway to be influenced. Collectively, FZDs located in cell membrane seem to be more appropriate to be considered as therapeutic targets (Table 2).

6. Conclusions and Future Perspectives

WNT signaling pathways have been found to be involved in many important biological processes, especially in tumor development and progression from studies in the past years. As a crucial component of WNT signaling pathway, the FZD family anchored in cell membrane functions in binding to extracellular WNT ligands and transmitting WNT signals to the intracellular molecules, which results in the increased transcription of target genes, many of which encode known oncogenes. In addition, overexpression of FZD family was frequently discovered in various cancers, which mediated cell proliferation, motility, metastasis, stem cell properties, and resistance of tumor cells to chemotherapy. The clinical samples also show the significant relevance between increased expression of FZD family and poor prognosis in a large panel of patients. Therefore, targeting FZD receptors will be a potential and attractive approach for human cancer therapy. Other therapeutic agents specifically targeting the WNT-secretion (Porcupine inhibitors) [105] or β-catenin destruction complex (Tankyrase inhibitors) [106,107] have only recently entered clinical trials and none has yet been approved. Porcupine is a member of O-acyltransferases which specifically palmitoylates WNTs, thereby enabling all WNTs secretion [108]. Tankyrase 1 and tankyrase 2 (TNKS1/2) are part of the larger family of poly (ADP-ribose) polymerase (PARP) enzymes and are the regulators of β-catenin destruction complex. However, the anti-tumor potential of both porcupine inhibitors and tankyrase inhibitors need to be carefully evaluated owing to on-target toxicity (i.e., WNT inhibition and effects on intestinal stem cells, bone turnover, and haematopoiesis) as well as off-target toxicity (i.e., inhibition of other family members of O-acyltransferases or PARP enzymes). In terms of the specificity and selectivity, targeting the signaling cascade at the level of the FZD receptors that are aberrantly overexpressed in tumors may potentiate the therapeutic advantages of WNT-driven cancers. Furthermore, FZD7 expression was reported to be downregulated in all differentiated cell populations relative to undifferentiated human embryonic stem cells, which meant that FZD7 is largely restricted to embryonic tissues with little expression in differentiated tissues [109]. This point makes FZD7 a more attractive target. Currently, several measures have been adopted to target FZD receptors, including the utilization of antibodies, small interfering peptides and small molecule inhibitors (Figure 2). All of these molecules display notable anti-tumor effects in vivo or in vitro. However, it is well-known that some FZDs are still moderately expressed in normal tissues. Therefore, the safety of drugs or antibodies against FZD family need to be further assessed before they are used to treat human cancers. Here, we are looking forward to seeing low or non-toxic and potent anti-FZDs antibody or small molecule inhibitor to be invented in the foreseeable future.

Author Contributions

C.-M.Z.: drafting of the manuscript, figure design; Z.C.: revision of the manuscript; L.F.: concept and design, critical revision of the manuscript and obtained funding.

Acknowledgments

This work was supported by the National Key R&D program of China (2017YFA0503900 and 2016YFC0904600), the National Natural Science Foundation of China (81772957), the Science and Technology Foundation of Shenzhen (KQTD20140630100658078), and the Shenzhen Peacock Program Startup Fund.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

APCAdenomatous Polyposis Coli
BRMS1LBreast cancer metastasis suppressor 1 like
CaMKIICa2+/calmodulin-dependent protein kinase II
CRCColorectal cancer
CRDCysteine-rich domain
DKKDickkopf WNT signaling pathway inhibitor
DVLDishevelled
ECEndometrial cancer
EMTEpithelial-mesenchymal transition
ERGETS-related gene
ESCCEsophageal squamous cell carcinoma
FZDFrizzled receptors
GPCRsG protein-coupled receptor proteins
HCCHepatocellular carcinoma
HIG2Hypoxia-inducible protein-2
LRP5/6Low density lipoprotein receptor related protein 5/6
NBNeuroblastoma
NF-kBNuclear factor kB
NLKNemo-like kinase
NSCLCNon-small cell lung cancer
PCPPlanar cell polarity
PDACPancreatic ductal adenocarcinoma cells
PPARγPeroxisome proliferator-activated receptor γ
ROR2Receptor tyrosine kinase like orphan receptor 2
RYKReceptor-like tyrosine kinase
SACCSalivary adenoid cystic carcinomas
scFvsSingle chain variable fragments
SFRPSecreted frizzled-related protein
sFZD7Soluble FZD7
STAT3Signal transducer and activator of transcription 3
TAK1TGFβ-activated kinase 1
TAZTafazzin
TNBCTriple negative breast cancer
TSCCTongue squamous cell carcinoma
WIFWNT inhibitor factor

References

  1. Vinson, C.R.; Conover, S.; Adler, P.N. A drosophila tissue polarity locus encodes a protein containing seven potential transmembrane domains. Nature 1989, 338, 263–264. [Google Scholar] [CrossRef] [PubMed]
  2. Pez, F.; Lopez, A.; Kim, M.; Wands, J.R.; Caron de Fromentel, C.; Merle, P. Wnt signaling and hepatocarcinogenesis: Molecular targets for the development of innovative anticancer drugs. J. Hepatol. 2013, 59, 1107–1117. [Google Scholar] [CrossRef] [PubMed]
  3. Bhanot, P.; Brink, M.; Samos, C.H.; Hsieh, J.C.; Wang, Y.; Macke, J.P.; Andrew, D.; Nathans, J.; Nusse, R. A new member of the frizzled family from drosophila functions as a wingless receptor. Nature 1996, 382, 225–230. [Google Scholar] [CrossRef] [PubMed]
  4. Schulte, G. International union of basic and clinical pharmacology. Lxxx. The class frizzled receptors. Pharmacol. Rev. 2010, 62, 632–667. [Google Scholar] [CrossRef] [PubMed]
  5. Flanagan, D.J.; Phesse, T.J.; Barker, N.; Schwab, R.H.; Amin, N.; Malaterre, J.; Stange, D.E.; Nowell, C.J.; Currie, S.A.; Saw, J.T.; et al. Frizzled7 functions as a wnt receptor in intestinal epithelial lgr5(+) stem cells. Stem Cell Rep. 2015, 4, 759–767. [Google Scholar] [CrossRef] [PubMed]
  6. King, T.D.; Zhang, W.; Suto, M.J.; Li, Y. Frizzled7 as an emerging target for cancer therapy. Cell. Signal. 2012, 24, 846–851. [Google Scholar] [CrossRef] [PubMed]
  7. Nile, A.H.; Mukund, S.; Stanger, K.; Wang, W.; Hannoush, R.N. Unsaturated fatty acyl recognition by frizzled receptors mediates dimerization upon wnt ligand binding. Proc. Natl. Acad. Sci. USA 2017, 114, 4147–4152. [Google Scholar] [CrossRef] [PubMed]
  8. Carron, C.; Pascal, A.; Djiane, A.; Boucaut, J.C.; Shi, D.L.; Umbhauer, M. Frizzled receptor dimerization is sufficient to activate the wnt/β-catenin pathway. J. Cell Sci. 2003, 116, 2541–2550. [Google Scholar] [CrossRef] [PubMed]
  9. Fredriksson, R.; Lagerstrom, M.C.; Lundin, L.G.; Schioth, H.B. The g-protein-coupled receptors in the human genome form five main families. Phylogenetic analysis, paralogon groups, and fingerprints. Mol. Pharmacol. 2003, 63, 1256–1272. [Google Scholar] [CrossRef] [PubMed]
  10. Huang, H.C.; Klein, P.S. The frizzled family: Receptors for multiple signal transduction pathways. Genome Biol. 2004, 5, 234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Dijksterhuis, J.P.; Petersen, J.; Schulte, G. Wnt/frizzled signalling: Receptor-ligand selectivity with focus on FZD-G protein signalling and its physiological relevance: Iuphar review 3. Br. J. Pharmacol. 2014, 171, 1195–1209. [Google Scholar] [CrossRef] [PubMed]
  12. Flahaut, M.; Meier, R.; Coulon, A.; Nardou, K.A.; Niggli, F.K.; Martinet, D.; Beckmann, J.S.; Joseph, J.M.; Muhlethaler-Mottet, A.; Gross, N. The wnt receptor FZD1 mediates chemoresistance in neuroblastoma through activation of the wnt/β-catenin pathway. Oncogene 2009, 28, 2245–2256. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, H.; Zhang, X.; Wu, X.; Li, W.; Su, P.; Cheng, H.; Xiang, L.; Gao, P.; Zhou, G. Interference of frizzled 1 (FZD1) reverses multidrug resistance in breast cancer cells through the wnt/β-catenin pathway. Cancer Lett. 2012, 323, 106–113. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, H.; Jing, X.; Wu, X.; Hu, J.; Zhang, X.; Wang, X.; Su, P.; Li, W.; Zhou, G. Suppression of multidrug resistance by rosiglitazone treatment in human ovarian cancer cells through downregulation of FZD1 and MDR1 genes. Anticancer Drugs 2015, 26, 706–715. [Google Scholar] [CrossRef] [PubMed]
  15. Su, W.; Mo, Y.; Wu, F.; Guo, K.; Li, J.; Luo, Y.; Ye, H.; Guo, H.; Li, D.; Yang, Z. miR-135b reverses chemoresistance of non-small cell lung cancer cells by downregulation of FZD1. Biomed. Pharmacother. 2016, 84, 123–129. [Google Scholar] [CrossRef] [PubMed]
  16. Lapointe, E.; Boyer, A.; Rico, C.; Paquet, M.; Franco, H.L.; Gossen, J.; DeMayo, F.J.; Richards, J.S.; Boerboom, D. FZD1 regulates cumulus expansion genes and is required for normal female fertility in mice. Biol. Reprod. 2012, 87, 104. [Google Scholar] [CrossRef] [PubMed]
  17. Gujral, T.S.; Chan, M.; Peshkin, L.; Sorger, P.K.; Kirschner, M.W.; MacBeath, G. A noncanonical frizzled2 pathway regulates epithelial-mesenchymal transition and metastasis. Cell 2014, 159, 844–856. [Google Scholar] [CrossRef] [PubMed]
  18. Zhang, E.; Li, Z.; Xu, Z.; Duan, W.; Sun, C.; Lu, L. Frizzled2 mediates the migration and invasion of human oral squamous cell carcinoma cells through the regulation of the signal transducer and activator of transcription-3 signaling pathway. Oncol. Rep. 2015, 34, 3061–3067. [Google Scholar] [CrossRef] [PubMed]
  19. Bian, Y.; Chang, X.; Liao, Y.; Wang, J.; Li, Y.; Wang, K.; Wan, X. Promotion of epithelial-mesenchymal transition by frizzled2 is involved in the metastasis of endometrial cancer. Oncol. Rep. 2016, 36, 803–810. [Google Scholar] [CrossRef] [PubMed]
  20. Zins, K.; Schafer, R.; Paulus, P.; Dobler, S.; Fakhari, N.; Sioud, M.; Aharinejad, S.; Abraham, D. Frizzled2 signaling regulates growth of high-risk neuroblastomas by interfering with β-catenin-dependent and β-catenin-independent signaling pathways. Oncotarget 2016, 7, 46187–46202. [Google Scholar] [CrossRef] [PubMed]
  21. Ding, L.C.; Huang, X.Y.; Zheng, F.F.; Xie, J.; She, L.; Feng, Y.; Su, B.H.; Zheng, D.L.; Lu, Y.G. FZD2 inhibits the cell growth and migration of salivary adenoid cystic carcinomas. Oncol. Rep. 2016, 35, 1006–1012. [Google Scholar] [CrossRef] [PubMed]
  22. Kadzik, R.S.; Cohen, E.D.; Morley, M.P.; Stewart, K.M.; Lu, M.M.; Morrisey, E.E. Wnt ligand/frizzled 2 receptor signaling regulates tube shape and branch-point formation in the lung through control of epithelial cell shape. Proc. Natl. Acad. Sci. USA 2014, 111, 12444–12449. [Google Scholar] [CrossRef] [PubMed]
  23. Hansen, C.; Howlin, J.; Tengholm, A.; Dyachok, O.; Vogel, W.F.; Nairn, A.C.; Greengard, P.; Andersson, T. Wnt-5a-induced phosphorylation of DARPP-32 inhibits breast cancer cell migration in a CREB-dependent manner. J. Biol. Chem. 2009, 284, 27533–27543. [Google Scholar] [CrossRef] [PubMed]
  24. Endo, Y.; Beauchamp, E.; Woods, D.; Taylor, W.G.; Toretsky, J.A.; Uren, A.; Rubin, J.S. Wnt-3a and Dickkopf-1 stimulate neurite outgrowth in ewing tumor cells via a frizzled3- and c-jun N-terminal kinase-dependent mechanism. Mol. Cell. Biol. 2008, 28, 2368–2379. [Google Scholar] [CrossRef] [PubMed]
  25. Morello, F.; Prasad, A.A.; Rehberg, K.; Vieira de Sa, R.; Anton-Bolanos, N.; Leyva-Diaz, E.; Adolfs, Y.; Tissir, F.; Lopez-Bendito, G.; Pasterkamp, R.J. Frizzled3 controls axonal polarity and intermediate target entry during striatal pathway development. J. Neurosci. 2015, 35, 14205–14219. [Google Scholar] [CrossRef] [PubMed]
  26. Hua, Z.L.; Smallwood, P.M.; Nathans, J. Frizzled3 controls axonal development in distinct populations of cranial and spinal motor neurons. Elife 2013, 2, e01482. [Google Scholar] [CrossRef] [PubMed]
  27. Tickenbrock, L.; Hehn, S.; Sargin, B.; Choudhary, C.; Baumer, N.; Buerger, H.; Schulte, B.; Muller, O.; Berdel, W.E.; Muller-Tidow, C.; et al. Activation of wnt signalling in acute myeloid leukemia by induction of frizzled-4. Int. J. Oncol. 2008, 33, 1215–1221. [Google Scholar] [CrossRef] [PubMed]
  28. Gupta, S.; Iljin, K.; Sara, H.; Mpindi, J.P.; Mirtti, T.; Vainio, P.; Rantala, J.; Alanen, K.; Nees, M.; Kallioniemi, O. FZD4 as a mediator of erg oncogene-induced wnt signaling and epithelial-to-mesenchymal transition in human prostate cancer cells. Cancer Res. 2010, 70, 6735–6745. [Google Scholar] [CrossRef] [PubMed]
  29. Jin, X.; Jeon, H.Y.; Joo, K.M.; Kim, J.K.; Jin, J.; Kim, S.H.; Kang, B.G.; Beck, S.; Lee, S.J.; Kim, J.K.; et al. Frizzled 4 regulates stemness and invasiveness of migrating glioma cells established by serial intracranial transplantation. Cancer Res. 2011, 71, 3066–3075. [Google Scholar] [CrossRef] [PubMed]
  30. Ueno, K.; Hirata, H.; Majid, S.; Yamamura, S.; Shahryari, V.; Tabatabai, Z.L.; Hinoda, Y.; Dahiya, R. Tumor suppressor microrna-493 decreases cell motility and migration ability in human bladder cancer cells by downregulating rhoc and fzd4. Mol. Cancer Ther. 2012, 11, 244–253. [Google Scholar] [CrossRef] [PubMed]
  31. Ma, C.; Xu, B.; Husaiyin, S.; Wang, L.; Wusainahong, K.; Ma, J.; Zhu, K.; Niyazi, M. Microrna-505 predicts prognosis and acts as tumor inhibitor in cervical carcinoma with inverse association with fzd4. Biomed. Pharmacother. 2017, 92, 586–594. [Google Scholar] [CrossRef] [PubMed]
  32. Xu, Q.; Wang, Y.; Dabdoub, A.; Smallwood, P.M.; Williams, J.; Woods, C.; Kelley, M.W.; Jiang, L.; Tasman, W.; Zhang, K.; et al. Vascular development in the retina and inner ear: Control by Norrin and Frizzled-4, a high-affinity ligand-receptor pair. Cell 2004, 116, 883–895. [Google Scholar] [CrossRef]
  33. Bobbs, A.; Gellerman, K.; Hallas, W.M.; Joseph, S.; Yang, C.; Kurkewich, J.; Cowden Dahl, K.D. ARID3B directly regulates ovarian cancer promoting genes. PLoS ONE 2015, 10, e0131961. [Google Scholar] [CrossRef] [PubMed]
  34. Long, Q.Z.; Du, Y.F.; Liu, X.G.; Li, X.; He, D.L. miR-124 represses FZD5 to attenuate P-glycoprotein-mediated chemo-resistance in renal cell carcinoma. Tumor Biol. 2015, 36, 7017–7026. [Google Scholar] [CrossRef] [PubMed]
  35. Liu, F.; Liu, Y.; Shen, J.; Zhang, G.; Han, J. Microrna-224 inhibits proliferation and migration of breast cancer cells by down-regulating fizzled 5 expression. Oncotarget 2016, 7, 49130–49142. [Google Scholar] [PubMed]
  36. Steinhart, Z.; Pavlovic, Z.; Chandrashekhar, M.; Hart, T.; Wang, X.; Zhang, X.; Robitaille, M.; Brown, K.R.; Jaksani, S.; Overmeer, R.; et al. Genome-wide CRISPR screens reveal a wnt-FZD5 signaling circuit as a druggable vulnerability of RNF43-mutant pancreatic tumors. Nat. Med. 2017, 23, 60–68. [Google Scholar] [CrossRef] [PubMed]
  37. Burns, C.J.; Zhang, J.; Brown, E.C.; Van Bibber, A.M.; Van Es, J.; Clevers, H.; Ishikawa, T.O.; Taketo, M.M.; Vetter, M.L.; Fuhrmann, S. Investigation of frizzled-5 during embryonic neural development in mouse. Dev. Dyn. 2008, 237, 1614–1626. [Google Scholar] [CrossRef] [PubMed]
  38. Cantilena, S.; Pastorino, F.; Pezzolo, A.; Chayka, O.; Pistoia, V.; Ponzoni, M.; Sala, A. Frizzled receptor 6 marks rare, highly tumourigenic stem-like cells in mouse and human neuroblastomas. Oncotarget 2011, 2, 976–983. [Google Scholar] [CrossRef] [PubMed]
  39. Piga, R.; van Dartel, D.; Bunschoten, A.; van der Stelt, I.; Keijer, J. Role of frizzled6 in the molecular mechanism of β-carotene action in the lung. Toxicology 2014, 320, 67–73. [Google Scholar] [CrossRef] [PubMed]
  40. Kim, B.K.; Yoo, H.I.; Kim, I.; Park, J.; Kim Yoon, S. FZD6 expression is negatively regulated by miR-199a-5p in human colorectal cancer. BMB Rep. 2015, 48, 360–366. [Google Scholar] [CrossRef] [PubMed]
  41. Yan, J.; Liu, T.; Zhou, X.; Dang, Y.; Yin, C.; Zhang, G. FZD6, targeted by miR-21, represses gastric cancer cell proliferation and migration via activating non-canonical wnt pathway. Am. J. Transl. Res. 2016, 8, 2354–2364. [Google Scholar] [PubMed]
  42. Corda, G.; Sala, G.; Lattanzio, R.; Iezzi, M.; Sallese, M.; Fragassi, G.; Lamolinara, A.; Mirza, H.; Barcaroli, D.; Ermler, S.; et al. Functional and prognostic significance of the genomic amplification of frizzled 6 (FZD6) in breast cancer. J. Pathol. 2017, 241, 350–361. [Google Scholar] [CrossRef] [PubMed]
  43. Huang, T.; Alvarez, A.A.; Pangeni, R.P.; Horbinski, C.M.; Lu, S.; Kim, S.H.; James, C.D.; Raizer, J.J.; Kessler, J.A.; Brenann, C.W.; et al. A regulatory circuit of miR-125b/miR-20b and wnt signalling controls glioblastoma phenotypes through fzd6-modulated pathways. Nat. Commun. 2016, 7, 12885. [Google Scholar] [CrossRef] [PubMed]
  44. Cui, C.Y.; Klar, J.; Georgii-Heming, P.; Frojmark, A.S.; Baig, S.M.; Schlessinger, D.; Dahl, N. Frizzled6 deficiency disrupts the differentiation process of nail development. J. Investig. Dermatol. 2013, 133, 1990–1997. [Google Scholar] [CrossRef] [PubMed]
  45. Nile, A.H.; de Sousa, E.M.F.; Mukund, S.; Piskol, R.; Hansen, S.; Zhou, L.; Zhang, Y.; Fu, Y.; Gogol, E.B.; Komuves, L.G.; et al. A selective peptide inhibitor of Frizzled 7 receptors disrupts intestinal stem cells. Nat. Chem. Biol. 2018, 14, 582–590. [Google Scholar] [CrossRef] [PubMed]
  46. Chakrabarti, R.; Wei, Y.; Hwang, J.; Hang, X.; Andres Blanco, M.; Choudhury, A.; Tiede, B.; Romano, R.A.; DeCoste, C.; Mercatali, L.; et al. Deltanp63 promotes stem cell activity in mammary gland development and basal-like breast cancer by enhancing FZD7 expression and wnt signalling. Nat. Cell Biol. 2014, 16, 1004–1015. [Google Scholar] [CrossRef] [PubMed]
  47. Xie, S.L.; Fan, S.; Zhang, S.Y.; Chen, W.X.; Li, Q.X.; Pan, G.K.; Zhang, H.Q.; Wang, W.W.; Weng, B.; Zhang, Z.; et al. SOX8 regulates cancer stem-like properties and cisplatin-induced EMT in tongue squamous cell carcinoma by acting on the wnt/β-catenin pathway. Int. J. Cancer 2018, 142, 1252–1265. [Google Scholar] [CrossRef] [PubMed]
  48. Leung, C.O.; Mak, W.N.; Kai, A.K.; Chan, K.S.; Lee, T.K.; Ng, I.O.; Lo, R.C. SOX9 confers stemness properties in hepatocellular carcinoma through Frizzled-7 mediated wnt/β-catenin signaling. Oncotarget 2016, 7, 29371–29386. [Google Scholar] [CrossRef] [PubMed]
  49. Sun, Y.; Xu, T.; Cao, Y.W.; Ding, X.Q. Antitumor effect of miR-27b-3p on lung cancer cells via targeting FZD7. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 4113–4123. [Google Scholar] [PubMed]
  50. Deng, B.; Zhang, Y.; Zhang, S.; Wen, F.; Miao, Y.; Guo, K. Microrna-142-3p inhibits cell proliferation and invasion of cervical cancer cells by targeting FZD7. Tumor Biol. 2015, 36, 8065–8073. [Google Scholar] [CrossRef] [PubMed]
  51. Cao, T.T.; Xiang, D.; Liu, B.L.; Huang, T.X.; Tan, B.B.; Zeng, C.M.; Wang, Z.Y.; Ming, X.Y.; Zhang, L.Y.; Jin, G.; et al. FZD7 is a novel prognostic marker and promotes tumor metastasis via wnt and EMT signaling pathways in esophageal squamous cell carcinoma. Oncotarget 2017, 8, 65957–65968. [Google Scholar] [CrossRef] [PubMed]
  52. Liu, X.; Yan, Y.; Ma, W.; Wu, S. Knockdown of frizzled-7 inhibits cell growth and metastasis and promotes chemosensitivity of esophageal squamous cell carcinoma cells by inhibiting wnt signaling. Biochem. Biophys. Res. Commun. 2017, 490, 1112–1118. [Google Scholar] [CrossRef] [PubMed]
  53. Qiu, X.; Jiao, J.; Li, Y.; Tian, T. Overexpression of FZD7 promotes glioma cell proliferation by upregulating taz. Oncotarget 2016, 7, 85987–85999. [Google Scholar] [CrossRef] [PubMed]
  54. Asad, M.; Wong, M.K.; Tan, T.Z.; Choolani, M.; Low, J.; Mori, S.; Virshup, D.; Thiery, J.P.; Huang, R.Y. FZD7 drives in vitro aggressiveness in stem-a subtype of ovarian cancer via regulation of non-canonical wnt/pcp pathway. Cell Death Dis. 2014, 5, e1346. [Google Scholar] [CrossRef] [PubMed]
  55. Deng, B.; Zhang, S.; Miao, Y.; Zhang, Y.; Wen, F.; Guo, K. Down-regulation of frizzled-7 expression inhibits migration, invasion, and epithelial-mesenchymal transition of cervical cancer cell lines. Med. Oncol. 2015, 32, 102. [Google Scholar] [CrossRef] [PubMed]
  56. Li, G.; Su, Q.; Liu, H.; Wang, D.; Zhang, W.; Lu, Z.; Chen, Y.; Huang, X.; Li, W.; Zhang, C.; 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, 280–293. [Google Scholar] [CrossRef] [PubMed]
  57. Yang, L.; Wu, X.; Wang, Y.; Zhang, K.; Wu, J.; Yuan, Y.C.; Deng, X.; Chen, L.; Kim, C.C.; Lau, S.; et al. FZD7 has a critical role in cell proliferation in triple negative breast cancer. Oncogene 2011, 30, 4437–4446. [Google Scholar] [CrossRef] [PubMed]
  58. Ueno, K.; Hazama, S.; Mitomori, S.; Nishioka, M.; Suehiro, Y.; Hirata, H.; Oka, M.; Imai, K.; Dahiya, R.; Hinoda, Y. Down-regulation of frizzled-7 expression decreases survival, invasion and metastatic capabilities of colon cancer cells. Br. J. Cancer 2009, 101, 1374–1381. [Google Scholar] [CrossRef] [PubMed]
  59. Tiwary, S.; Xu, L. Correction: Frizzled7 is required for tumor inititation and metastatic growth of melanoma cells. PLoS ONE 2017, 12, e0187388. [Google Scholar] [CrossRef] [PubMed]
  60. Geng, Y.; Lu, X.; Wu, X.; Xue, L.; Wang, X.; Xu, J. Microrna-27b suppresses helicobacter pylori-induced gastric tumorigenesis through negatively regulating frizzled7. Oncol. Rep. 2016, 35, 2441–2450. [Google Scholar] [CrossRef] [PubMed]
  61. Li, Q.; Ye, L.; Zhang, X.; Wang, M.; Lin, C.; Huang, S.; Guo, W.; Lai, Y.; Du, H.; Li, J.; et al. FZD8, a target of p53, promotes bone metastasis in prostate cancer by activating canonical wnt/β-catenin signaling. Cancer Lett. 2017, 402, 166–176. [Google Scholar] [CrossRef] [PubMed]
  62. Bravo, D.T.; Yang, Y.L.; Kuchenbecker, K.; Hung, M.S.; Xu, Z.; Jablons, D.M.; You, L. Frizzled-8 receptor is activated by the wnt-2 ligand in non-small cell lung cancer. BMC Cancer 2013, 13, 316. [Google Scholar] [CrossRef] [PubMed]
  63. Fu, X.; Li, H.; Liu, C.; Hu, B.; Li, T.; Wang, Y. Long noncoding RNA AK126698 inhibits proliferation and migration of non-small cell lung cancer cells by targeting frizzled-8 and suppressing wnt/β-catenin signaling pathway. OncoTargets Ther. 2016, 9, 3815–3827. [Google Scholar] [CrossRef] [PubMed]
  64. Jiang, Q.; He, M.; Guan, S.; Ma, M.; Wu, H.; Yu, Z.; Jiang, L.; Wang, Y.; Zong, X.; Jin, F.; et al. Microrna-100 suppresses the migration and invasion of breast cancer cells by targeting fzd-8 and inhibiting wnt/β-catenin signaling pathway. Tumor Biol. 2016, 37, 5001–5011. [Google Scholar] [CrossRef] [PubMed]
  65. Wang, J.; Pang, W.; Zuo, Z.; Zhang, W.; He, W. Microrna-520b suppresses proliferation, migration, and invasion of spinal osteosarcoma cells via downregulation of frizzled-8. Oncol. Res. 2017, 25, 1297–1304. [Google Scholar] [CrossRef] [PubMed]
  66. Sun, S.; Liu, S.; Duan, S.Z.; Zhang, L.; Zhou, H.; Hu, Y.; Zhou, X.; Shi, C.; Zhou, R.; Zhang, Z. Targeting the c-met/fzd8 signaling axis eliminates patient-derived cancer stem-like cells in head and neck squamous carcinomas. Cancer Res. 2014, 74, 7546–7559. [Google Scholar] [CrossRef] [PubMed]
  67. Yin, S.; Xu, L.; Bonfil, R.D.; Banerjee, S.; Sarkar, F.H.; Sethi, S.; Reddy, K.B. Tumor-initiating cells and fzd8 play a major role in drug resistance in triple-negative breast cancer. Mol. Cancer Ther. 2013, 12, 491–498. [Google Scholar] [CrossRef] [PubMed]
  68. Wang, M.T.; Holderfield, M.; Galeas, J.; Delrosario, R.; To, M.D.; Balmain, A.; McCormick, F. K-Ras promotes tumorigenicity through suppression of non-canonical wnt signaling. Cell 2015, 163, 1237–1251. [Google Scholar] [CrossRef] [PubMed]
  69. Wang, H.Q.; Xu, M.L.; Ma, J.; Zhang, Y.; Xie, C.H. Frizzled-8 as a putative therapeutic target in human lung cancer. Biochem. Biophys. Res. Commun. 2012, 417, 62–66. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, Z.; Schittenhelm, J.; Guo, K.; Buhring, H.J.; Trautmann, K.; Meyermann, R.; Schluesener, H.J. Upregulation of frizzled 9 in astrocytomas. Neuropathol. Appl. Neurobiol. 2006, 32, 615–624. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, Q.; Liu, H.; Wang, Q.; Zhou, F.; Liu, Y.; Zhang, Y.; Ding, H.; Yuan, M.; Li, F.; Chen, Y. Involvement of c-Fos in cell proliferation, migration, and invasion in osteosarcoma cells accompanied by altered expression of Wnt2 and Fzd9. PLoS ONE 2017, 12, e0180558. [Google Scholar] [CrossRef] [PubMed]
  72. Fujimoto, T.; Tomizawa, M.; Yokosuka, O. Sirna of frizzled-9 suppresses proliferation and motility of hepatoma cells. Int. J. Oncol. 2009, 35, 861–866. [Google Scholar] [PubMed]
  73. Zhang, Y.; Jiang, Q.; Kong, X.; Yang, L.; Hu, W.; Lv, C.; Li, Y. Methylation status of the promoter region of the human frizzled 9 gene in acute myeloid leukemia. Mol. Med. Rep. 2016, 14, 1339–1344. [Google Scholar] [CrossRef] [PubMed]
  74. Winn, R.A.; Marek, L.; Han, S.Y.; Rodriguez, K.; Rodriguez, N.; Hammond, M.; Van Scoyk, M.; Acosta, H.; Mirus, J.; Barry, N.; et al. Restoration of wnt-7a expression reverses non-small cell lung cancer cellular transformation through frizzled-9-mediated growth inhibition and promotion of cell differentiation. J. Biol. Chem. 2005, 280, 19625–19634. [Google Scholar] [CrossRef] [PubMed]
  75. Winn, R.A.; Van Scoyk, M.; Hammond, M.; Rodriguez, K.; Crossno, J.T., Jr.; Heasley, L.E.; Nemenoff, R.A. Antitumorigenic effect of wnt 7a and fzd9 in non-small cell lung cancer cells is mediated through erk-5-dependent activation of peroxisome proliferator-activated receptor gamma. J. Biol. Chem. 2006, 281, 26943–26950. [Google Scholar] [CrossRef] [PubMed]
  76. Zhao, C.; Aviles, C.; Abel, R.A.; Almli, C.R.; McQuillen, P.; Pleasure, S.J. Hippocampal and visuospatial learning defects in mice with a deletion of frizzled 9, a gene in the williams syndrome deletion interval. Development 2005, 132, 2917–2927. [Google Scholar] [CrossRef] [PubMed]
  77. Ranheim, E.A.; Kwan, H.C.; Reya, T.; Wang, Y.K.; Weissman, I.L.; Francke, U. Frizzled 9 knock-out mice have abnormal b-cell development. Blood 2005, 105, 2487–2494. [Google Scholar] [CrossRef] [PubMed]
  78. Fukukawa, C.; Nagayama, S.; Tsunoda, T.; Toguchida, J.; Nakamura, Y.; Katagiri, T. Activation of the non-canonical Dvl-Rac1-JNK pathway by frizzled homologue 10 in human synovial sarcoma. Oncogene 2009, 28, 1110–1120. [Google Scholar] [CrossRef] [PubMed]
  79. Tamaki, S.; Fukuta, M.; Sekiguchi, K.; Jin, Y.; Nagata, S.; Hayakawa, K.; Hineno, S.; Okamoto, T.; Watanabe, M.; Woltjen, K.; et al. SS18-SSX, the oncogenic fusion protein in synovial sarcoma, is a cellular context-dependent epigenetic modifier. PLoS ONE 2015, 10, e0142991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Nagayama, S.; Yamada, E.; Kohno, Y.; Aoyama, T.; Fukukawa, C.; Kubo, H.; Watanabe, G.; Katagiri, T.; Nakamura, Y.; Sakai, Y.; et al. Inverse correlation of the up-regulation of FZD10 expression and the activation of β-catenin in synchronous colorectal tumors. Cancer Sci. 2009, 100, 405–412. [Google Scholar] [CrossRef] [PubMed]
  81. Gong, C.; Qu, S.; Lv, X.B.; Liu, B.; Tan, W.; Nie, Y.; Su, F.; Liu, Q.; Yao, H.; Song, E. Brms1l suppresses breast cancer metastasis by inducing epigenetic silence of FZD10. Nat. Commun. 2014, 5, 5406. [Google Scholar] [CrossRef] [PubMed]
  82. Togashi, A.; Katagiri, T.; Ashida, S.; Fujioka, T.; Maruyama, O.; Wakumoto, Y.; Sakamoto, Y.; Fujime, M.; Kawachi, Y.; Shuin, T.; et al. Hypoxia-inducible protein 2 (hig2), a novel diagnostic marker for renal cell carcinoma and potential target for molecular therapy. Cancer Res. 2005, 65, 4817–4826. [Google Scholar] [CrossRef] [PubMed]
  83. Wong, S.C.; He, C.W.; Chan, C.M.; Chan, A.K.; Wong, H.T.; Cheung, M.T.; Luk, L.L.; Au, T.C.; Chiu, M.K.; Ma, B.B.; et al. Clinical significance of frizzled homolog 3 protein in colorectal cancer patients. PLoS ONE 2013, 8, e79481. [Google Scholar] [CrossRef] [PubMed]
  84. Thiele, S.; Gobel, A.; Rachner, T.D.; Fuessel, S.; Froehner, M.; Muders, M.H.; Baretton, G.B.; Bernhardt, R.; Jakob, F.; Gluer, C.C.; et al. WNT5A has anti-prostate cancer effects in vitro and reduces tumor growth in the skeleton in vivo. J. Bone Miner. Res. 2015, 30, 471–480. [Google Scholar] [CrossRef] [PubMed]
  85. Nagayama, S.; Fukukawa, C.; Katagiri, T.; Okamoto, T.; Aoyama, T.; Oyaizu, N.; Imamura, M.; Toguchida, J.; Nakamura, Y. Therapeutic potential of antibodies against FZD10, a cell-surface protein, for synovial sarcomas. Oncogene 2005, 24, 6201–6212. [Google Scholar] [CrossRef] [PubMed]
  86. Fukukawa, C.; Hanaoka, H.; Nagayama, S.; Tsunoda, T.; Toguchida, J.; Endo, K.; Nakamura, Y.; Katagiri, T. Radioimmunotherapy of human synovial sarcoma using a monoclonal antibody against FZD10. Cancer Sci. 2008, 99, 432–440. [Google Scholar] [CrossRef] [PubMed]
  87. Pode-Shakked, N.; Harari-Steinberg, O.; Haberman-Ziv, Y.; Rom-Gross, E.; Bahar, S.; Omer, D.; Metsuyanim, S.; Buzhor, E.; Jacob-Hirsch, J.; Goldstein, R.S.; et al. Resistance or sensitivity of wilms’ tumor to anti-FZD7 antibody highlights the wnt pathway as a possible therapeutic target. Oncogene 2011, 30, 1664–1680. [Google Scholar] [CrossRef] [PubMed]
  88. Riley, R.S.; Day, E.S. Frizzled7 antibody-functionalized nanoshells enable multivalent binding for wnt signaling inhibition in triple negative breast cancer cells. Small 2017, 13. [Google Scholar] [CrossRef] [PubMed]
  89. Nickho, H.; Younesi, V.; Aghebati-Maleki, L.; Motallebnezhad, M.; Majidi Zolbanin, J.; Movassagh Pour, A.; Yousefi, M. Developing and characterization of single chain variable fragment (scfv) antibody against frizzled 7 (FZD7) receptor. Bioengineered 2017, 8, 501–510. [Google Scholar] [CrossRef] [PubMed]
  90. Zarei, N.; Fazeli, M.; Mohammadi, M.; Nejatollahi, F. Cell growth inhibition and apoptosis in breast cancer cells induced by anti-FZD7 scfvs: Involvement of bioinformatics-based design of novel epitopes. Breast Cancer Res. Treat. 2018, 169, 427–436. [Google Scholar] [CrossRef] [PubMed]
  91. Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt pathway inhibition via the targeting of frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [Google Scholar] [CrossRef] [PubMed]
  92. Katoh, M.; Katoh, M. Molecular genetics and targeted therapy of wnt-related human diseases (review). Int. J. Mol. Med. 2017, 40, 587–606. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, W.; Lu, W.; Ananthan, S.; Suto, M.J.; Li, Y. Discovery of novel frizzled-7 inhibitors by targeting the receptor’s transmembrane domain. Oncotarget 2017, 8, 91459–91470. [Google Scholar] [PubMed]
  94. Nambotin, S.B.; Lefrancois, L.; Sainsily, X.; Berthillon, P.; Kim, M.; Wands, J.R.; Chevallier, M.; Jalinot, P.; Scoazec, J.Y.; Trepo, C.; et al. Pharmacological inhibition of frizzled-7 displays anti-tumor properties in hepatocellular carcinoma. J. Hepatol. 2011, 54, 288–299. [Google Scholar] [CrossRef] [PubMed]
  95. Fujii, N.; You, L.; Xu, Z.; Uematsu, K.; Shan, J.; He, B.; Mikami, I.; Edmondson, L.R.; Neale, G.; Zheng, J.; et al. An antagonist of dishevelled protein-protein interaction suppresses β-catenin-dependent tumor cell growth. Cancer Res. 2007, 67, 573–579. [Google Scholar] [CrossRef] [PubMed]
  96. Le, P.N.; McDermott, J.D.; Jimeno, A. Targeting the wnt pathway in human cancers: Therapeutic targeting with a focus on omp-54f28. Pharmacol. Ther. 2015, 146, 1–11. [Google Scholar] [CrossRef] [PubMed]
  97. Bahrami, A.; Amerizadeh, F.; ShahidSales, S.; Khazaei, M.; Ghayour-Mobarhan, M.; Sadeghnia, H.R.; Maftouh, M.; Hassanian, S.M.; Avan, A. Therapeutic potential of targeting wnt/β-catenin pathway in treatment of colorectal cancer: Rational and progress. J. Cell. Biochem. 2017, 118, 1979–1983. [Google Scholar] [CrossRef] [PubMed]
  98. Jimeno, A.; Gordon, M.; Chugh, R.; Messersmith, W.; Mendelson, D.; Dupont, J.; Stagg, R.; Kapoun, A.M.; Xu, L.; Uttamsingh, S.; 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. Clin. Cancer Res. 2017, 23, 7490–7497. [Google Scholar] [CrossRef] [PubMed]
  99. Wei, W.; Chua, M.S.; Grepper, S.; So, S.K. Soluble frizzled-7 receptor inhibits wnt signaling and sensitizes hepatocellular carcinoma cells towards doxorubicin. Mol. Cancer 2011, 10, 16. [Google Scholar] [CrossRef] [PubMed]
  100. Kawano, Y.; Kypta, R. Secreted antagonists of the wnt signalling pathway. J. Cell Sci. 2003, 116, 2627–2634. [Google Scholar] [CrossRef] [PubMed]
  101. Nathke, I.S.; Adams, C.L.; Polakis, P.; Sellin, J.H.; Nelson, W.J. The adenomatous polyposis coli tumor suppressor protein localizes to plasma membrane sites involved in active cell migration. J. Cell Biol. 1996, 134, 165–179. [Google Scholar] [CrossRef] [PubMed]
  102. Saito-Diaz, K.; Benchabane, H.; Tiwari, A.; Tian, A.; Li, B.; Thompson, J.J.; Hyde, A.S.; Sawyer, L.M.; Jodoin, J.N.; Santos, E.; et al. APC inhibits ligand-independent wnt signaling by the clathrin endocytic pathway. Dev. Cell 2018, 44, 566–581.e568. [Google Scholar] [CrossRef] [PubMed]
  103. Hulsken, J.; Birchmeier, W.; Behrens, J. E-cadherin and APC compete for the interaction with β-catenin and the cytoskeleton. J. Cell Biol. 1994, 127, 2061–2069. [Google Scholar] [CrossRef] [PubMed]
  104. Hirano, S.; Kimoto, N.; Shimoyama, Y.; Hirohashi, S.; Takeichi, M. Identification of a neural α-catenin as a key regulator of cadherin function and multicellular organization. Cell 1992, 70, 293–301. [Google Scholar] [CrossRef]
  105. Chen, B.; Dodge, M.E.; Tang, W.; Lu, J.; Ma, Z.; Fan, C.W.; Wei, S.; Hao, W.; Kilgore, J.; Williams, N.S.; et al. Small molecule-mediated disruption of wnt-dependent signaling in tissue regeneration and cancer. Nat. Chem. Biol. 2009, 5, 100–107. [Google Scholar] [CrossRef] [PubMed]
  106. Huang, S.M.; Mishina, Y.M.; Liu, S.; Cheung, A.; Stegmeier, F.; Michaud, G.A.; Charlat, O.; Wiellette, E.; Zhang, Y.; Wiessner, S.; et al. Tankyrase inhibition stabilizes axin and antagonizes wnt signalling. Nature 2009, 461, 614–620. [Google Scholar] [CrossRef] [PubMed]
  107. Lehtio, L.; Chi, N.W.; Krauss, S. Tankyrases as drug targets. FEBS J. 2013, 280, 3576–3593. [Google Scholar] [CrossRef] [PubMed]
  108. Takada, R.; Satomi, Y.; Kurata, T.; Ueno, N.; Norioka, S.; Kondoh, H.; Takao, T.; Takada, S. Monounsaturated fatty acid modification of wnt protein: Its role in wnt secretion. Dev. Cell 2006, 11, 791–801. [Google Scholar] [CrossRef] [PubMed]
  109. Fernandez, A.; Huggins, I.J.; Perna, L.; Brafman, D.; Lu, D.; Yao, S.; Gaasterland, T.; Carson, D.A.; Willert, K. The wnt receptor FZD7 is required for maintenance of the pluripotent state in human embryonic stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, 1409–1414. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Diagram of a FYN (FYN proto-oncogene)/STAT3 (Signal transducer and activator of transcription 3) signaling for FZD2 in diverse solid tumors and a CaMKII (Ca2+/calmodulin-dependent protein kinase II)–TAK1 (TGFβ-activated kinase 1)–NLK (Nemo-like kinase) signaling for FZD6 in mesenchymal-associated phenotype glioblastoma.
Figure 1. Diagram of a FYN (FYN proto-oncogene)/STAT3 (Signal transducer and activator of transcription 3) signaling for FZD2 in diverse solid tumors and a CaMKII (Ca2+/calmodulin-dependent protein kinase II)–TAK1 (TGFβ-activated kinase 1)–NLK (Nemo-like kinase) signaling for FZD6 in mesenchymal-associated phenotype glioblastoma.
Ijms 19 01543 g001
Figure 2. Diagram of WNT (Wingless-type) signaling pathway. OMP-54F28 and sFZD7 act as decoy receptors to reduce the binding of ligands and receptors. The anti-FZD antibody binds to the conserved epitope of extracellular domain of FZDs to form a ligand-receptor complex, preventing the WNT ligands from interacting with FZDs. The small molecule compound SRI37892 targets the transmembrane of FZD7 and block WNT signal transmission. Small interfering peptides RHPDs (Rhodamine-tagged heptapeptide protein transduction domain for binding DVL) and Small molecule inhibitor FJ9 disrupt the joint of FZD7 and DVL. The DVL is not activated so that the WNT signal is unable to be transmitted into the cell. Abbreviations: FZDAb: FZD antibodies; CK1: casein kinase 1; APC: adenomatous polyposis coli; GSK3β: glycogen synthase kinase-3β; DVL: Dishevelled.
Figure 2. Diagram of WNT (Wingless-type) signaling pathway. OMP-54F28 and sFZD7 act as decoy receptors to reduce the binding of ligands and receptors. The anti-FZD antibody binds to the conserved epitope of extracellular domain of FZDs to form a ligand-receptor complex, preventing the WNT ligands from interacting with FZDs. The small molecule compound SRI37892 targets the transmembrane of FZD7 and block WNT signal transmission. Small interfering peptides RHPDs (Rhodamine-tagged heptapeptide protein transduction domain for binding DVL) and Small molecule inhibitor FJ9 disrupt the joint of FZD7 and DVL. The DVL is not activated so that the WNT signal is unable to be transmitted into the cell. Abbreviations: FZDAb: FZD antibodies; CK1: casein kinase 1; APC: adenomatous polyposis coli; GSK3β: glycogen synthase kinase-3β; DVL: Dishevelled.
Ijms 19 01543 g002
Table 1. The clinical relevance of FZDs in different cancers.
Table 1. The clinical relevance of FZDs in different cancers.
GeneCancerClinical RelevanceRef.
FZD1NeuroblastomaPatients with neuroblastoma who relapsed after chemotherapy showed a significant increase of FZD1 expression, but no significant increase was observed in the non-relapsed group of patients.[12]
FZD2Liver or lung FZD2 mRNA expression was found to be significantly increased in late stages of primary liver and lung cancers compared with normal tissue and early stage cancer.[17]
Endometrial FZD2 was overexpressed in endometrial cancer tissues compared with the level in normal tissues. Moreover, the expression of FZD2 was positively correlated with markers of mesenchymal cells, such as vimentin and N-cadherin.[19]
Salivary adenoid cystic carcinomaFZD2 expression was downregulated in the samples with metastasis and recurrence when compared to the samples without metastasis.[21]
FZD3ColorectalFZD3 protein expression was highly correlated with colorectal carcinogenesis and progression, indicating that it may potentially serve as a prognostic marker.[83]
FZD4Acute myeloid leukemiaFZD4 protein was expressed in about 80% samples from acute myeloid leukemia patients but rarely expressed in normal bone marrow.[27]
ProstateFZD4 was highly correlated with ERG[Erythroblast transformation-specific (ETS)-related gene] in clinical prostate samples. Higher FZD4 protein expression was often detected in ERG-positive tumors compared with ERG-negative samples.[28]
Cervical FZD4 mRNA expression level in cervical carcinomas was much higher than that in normal cervical tissues in human cervical carcinoma samples.[31]
FZD5ProstateFZD5 mRNA expression was significantly higher in the prostate cancer tissues compared with healthy controls.[84]
FZD6ColorectalBoth FZD6 protein expression and FZD6 mRNA expression were found to be significantly increased compared with adjacent non-tumor samples, which indicated that upregulation of FZD6 correlated with the development of colorectal cancer.[40]
BreastFZD6 expression was significantly associated with reduced distant relapse-free survival in the triple negative breast cancer patient subgroup, but no significant association with other subtypes.[42]
FZD7Esophageal FZD7 protein expression was significantly higher in ESCC than that in the adjacent non-tumor tissues. FZD7 overexpression was significantly associated with shorter survival time of patients with ESCC. [51]
GliomaFZD7 protein expression was significantly increased in glioma compared with the adjacent normal tissues.[53]
GastricFZD7 mRNA expression was found to be significantly upregulated in gastric cancers compared with normal gastric tissues. IHC staining also confirmed the result above and FZD7 overexpression associated with advanced tumor stages and poor survival.[56]
BreastFZD7 expression was significantly higher in the TNBC samples compared with non-TNBC samples.[57]
ColorectalFZD7 mRNA expression examined by real-time PCR was significantly increased in CRC compared with non-tumor tissues. FZD7 overexpression was significantly associated with higher tumor stage.[58]
FZD8ProstateBoth FZD8 mRNA and protein expression were found to be more notably increased in bone metastatic prostate cancer than in primary prostate cancer and normal prostate tissues.[61]
BreastFZD8 expression was significantly higher in human breast cancer tissues compared with the adjacent normal tissues, and high expression of FZD8 was closely correlated with lymph node metastasis.[64]
LungUpregulation of FZD8 expression was observed in 85% tumor samples from lung cancer patients when compared to their matched normal lung tissues.[69]
FZD9AstrocytomaFZD9 protein was more frequently expressed in malignant astrocytoma than in low-grade astrocytoma.[70]
OsteosarcomaFZD9 expression was higher in osteosarcoma tissues than in the adjacent non-cancerous tissues. The high level expression of FZD9 occurred more frequently in late stage than in the early stage of osteosarcoma.[71]
FZD10ColorectalFZD10 mRNA expression was confined to CRC tissues and almost absent from normal mucosa. No IHC staining was observed for FZD10 in normal mucosa, and only tumor cells in polyps and CRC tissues showed spotted staining.[80]
Synovial sarcomaFZD10 mRNA was expressed in synovial sarcoma, but not expressed in normal human organs except placenta. FZD10 protein expression appeared to be absent or low in normal vital organs compared to its highly increased expression in synovial sarcoma tissues.[85]
Table 2. FZD antibodies/inhibitors for various cancers.
Table 2. FZD antibodies/inhibitors for various cancers.
Antibodies/InhibitorsTargetsCancer Types
mAb 92-13FZD10Synovial sarcoma
anti-FZD7 antibodyFZD7Wilms’ tumor
scFvsFZD7Triple negative breast cancer
OMP-18R5FZD1,2,5,7, and 8Breast, Pancreatic, Colon and Lung Cancers
SRI37892FZD7Breast cancer
RHPDsFZD7Hepatocellular carcinoma
FJ9FZD7Melanoma, Non-small cell lung cancer
dFz7-21FZD1, FZD2, FZD7Unknown
IgG-2919 and IgG-2921FZD5RNF43-mutant pancreatic ductal adenocarcinoma
OMP-54F28Compete with FZD8Advanced solid tumors
sFZD7Compete with FZD7Hepatocellular carcinoma

Share and Cite

MDPI and ACS Style

Zeng, C.-M.; Chen, Z.; Fu, L. Frizzled Receptors as Potential Therapeutic Targets in Human Cancers. Int. J. Mol. Sci. 2018, 19, 1543. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19051543

AMA Style

Zeng C-M, Chen Z, Fu L. Frizzled Receptors as Potential Therapeutic Targets in Human Cancers. International Journal of Molecular Sciences. 2018; 19(5):1543. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19051543

Chicago/Turabian Style

Zeng, Chui-Mian, Zhe Chen, and Li Fu. 2018. "Frizzled Receptors as Potential Therapeutic Targets in Human Cancers" International Journal of Molecular Sciences 19, no. 5: 1543. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms19051543

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop