Targeting the Wnt signalling pathway in cancer: prospects and perils

DOI: https://doi.org/10.4414/smw.2019.20129

Holly V. Shawab*, Alexey Kovala*, Vladimir L. Katanaevabcd

aDepartment of Cell Physiology and Metabolism, Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Switzerland

bDepartment of Pharmacology and Toxicology, Faculty of Biology and Medicine, University of Lausanne, Switzerland

cSchool of Biomedicine, Far Eastern Federal University, Vladivostok, Russia

dInstitute of Oceangraphy, Minjiang University, Fuzhou, China

Summary

The Wnt pathway, involved in cancer development and progression, has for a long time been said to be undruggable, owing to its complexity and involvement in stem cell biology. This mindset has shifted in the last few years as new research and insights into the pathway mechanisms specific to tumour cells become apparent, leading to the development of multiple compounds targeting the pathway. In this review, we introduce the Wnt pathway and its connections to cancer biology and therapy resistance. We further dive into the details of drugs that have entered clinical trials, examining their successes and side effects. We show that these drugs all have one thing in common: in order to be successful, the drugs must target tumour specific activated sub-branches of the pathway, either at the receptor level or at the nuclear transcription level.

The Wnt pathway – a double-edged sword

One of the most fascinating features of multicellular organisms is the precise and tightly controlled communication among cells, necessary for the development, coordination and functioning of the individual organs and the body as a whole. To communicate, the cells employ chemical signals, which, when received by the recipient cells, trigger defined intracellular signalling pathways in order to relay the information and provide the adequate response to the external stimuli. This enables the body to coordinate patterning and organ development during embryogenesis, to keep the organism in homeostasis and to respond to external stresses and inputs, and to regenerate after injury. On the cellular level, a signalling cascade is initiated by secreted ligands (e.g., hormones, cytokines, neurotransmitters, growth factors) produced by one cell, which then bind to a receptor on another cell. The receptors in most cases are located on the cell surface, and the signal is then relayed through intracellular components of the pathway, called transducers and second messengers, resulting in the corresponding cellular effect, for example target gene transcription or changes in an enzymatic activity [1].

The Wnt pathway is one of the most important signalling cascades in the early events of embryonic development, where it controls cell proliferation and differentiation [2, 3]. To date, the Wnt signalling is still not fully understood. This is mainly because it is composed of a complicated network of a total of ten GPCR homologue Frizzled (FZD) receptors [4], three transmembrane tyrosine kinases Ryk, ROR and PTK7, muscle skeletal tyrosine kinase (MuSK) [5], the co-receptors LRP5/6 [6], and 19 glycolipoprotein Wnt ligands [7]. There is a high degree of promiscuity in the ligand-receptor interactions, although certain Wnts have higher affinities to certain FZD receptors and co-receptors [4, 8]. To further complicate matters, there are additionally secreted antagonists such as Secreted FZD-related proteins (Sfrp1, 2, 4 and 5), Wnt inhibitory factor (Wif) and Dickkopf 1 (Dkk1) reducing signalling activity, and the agonists R-spondin 1 to 4 potentiating the Wnt signalling through their receptors Lgr4, 5 and 6 [9, 10].

Wnt signalling is generally divided into three distinct branches: the canonical β-catenin/TCF pathway, the planar cell polarity (PCP) pathway and the Ca2+ pathway. Whereas some ligands are attributed to one distinct branch, others are competent to initiate signalling in several branches, depending on the receptor-ligand combination. It has also been demonstrated that under certain circumstances the β-catenin branch and the PCP branch antagonise each other [11].

The by far most studied branch is the canonical β-catenin/TCF pathway. It is characterised by accumulation of cytoplasmic protein β-catenin upon pathway initiation. It further translocates to the nucleus to bind the TCF family of transcription factors, leading to specific gene expression. The Wnt-dependent transcriptional programme in the nucleus – in a manner reminiscent of the complexity at the cell surface – is again controlled in a diversified manner. It has been shown that, depending on which co-activators β-catenin recruits, it will either upregulate genes responsible for self-renewal and proliferation (through, e.g., β-catenin binding to CBP) or will lead to upregulation of genes involved in differentiation (binding to p300) [12]. In the absence of Wnt ligands, a specific complex containing Axin, APC, CK1 and GSK3β phosphorylates β-catenin, targeting it for degradation. In the adult tissues, Wnt signalling is mainly silent with the exception of stem cells, where the pathway regulates replenishment and regenerative processes, for example in the intestinal crypt [13], haematopoietic stem cells [14] and bone [15]. This vital role of the Wnt pathway in maintaining tissues healthy, as one edge of the sword, stands in a sharp opposition to the other edge, which is the key role of the pathway in disease. If not held in check, aberrant Wnt signalling can lead to uncontrolled cell proliferation and cancer [2, 16].

Unlike the canonical signalling, β-catenin is not part of the PCP and Ca2+ pathways. The PCP pathway, involving small GTPases and JUN-N-terminal kinase, controls cell polarity, cytoskeletal remodelling, directional cell migration and c-Jun-dependent transcription. The Ca2+ signalling branch leads to activation of phospholipase C (PKC) followed by opening of intracellular stores of Ca2+, in turn leading to activation of downstream effectors such as NFAT and CREB transcription factors, also controlling cell migration and cell survival [17]. As these two branches involve cytoskeletal changes and cell migration, it is not surprising that they have been associated with cell invasion and metastasis in cancer [18–20].

The Wnt signalling-dependent cancers can be divided into those that harbour mutations in components of the pathway, and those cancers that have a dysregulation of Wnt signalling due to epigenetically driven up- or downregulation in expression levels of the pathway components. The most famous example of pathway mutations is that of the Wnt pathway suppressor APC. It was first associated with patients with familial adenomatous polyposis (FAP) and occurs in >80% of colorectal carcinomas [21, 22].

Immunohistological analyses of tissues from colorectal patients show that β-catenin relocalisation is a typical sign of the canonical pathway activation. Indeed, the loss of membrane β-catenin (where it plays Wnt-independent functions) is significantly associated with poor prognosis when using overall survival as the endpoint, as shown in a study of 720 colorectal patient samples [23]. Additional reports have demonstrated that loss of membranous β-catenin is especially prominent in the invasive front in colorectal cancer and that membranous localisation in general and in the invasive front in particular are both prognostic markers for longer disease-free survival [24], whereas high nuclear accumulation in colorectal cancer has been associated with worse disease-free and overall survival and higher probability of developing lymph node metastasis [25, 26].

In additional to mutational activation of the Wnt signalling, the pathway can be aberrantly activated by overexpression of the pathway components, such as Wnts or their FZD receptors [27]. Analysis of tumour tissues of 201 patients with colorectal cancer showed high Wnt1 and low expression of the non-canonical Wnt5a correlating with cytoplasmic and nuclear β-catenin; all three characteristics are indicative of shortened disease-free survival. High Wnt1 and nuclear β-catenin also correlated with lower overall survival [25]. In non-small-cell lung cancer, cytoplasmic Wnt1 is also significantly upregulated and correlates with β-catenin, c-myc and cyclin D1 overexpression. Although there was no link between high Wnt1/β-catenin expression and the disease stage, high expression correlated significantly with a lower 5-year survival rate [28].

FZD expression has also been analysed in various studies (reviewed by [29]). As expected, tumour tissues show an upregulation of FZD receptor expression compared with healthy tissues. The expression is even stronger towards later stages of cancer development. For example, in gastric cancer, high FZD7 expression significantly correlates with tumour invasion, metastasis and late stage cancer. In an analysis of 5-year survival, patients with high FZD7 expression had a 30.3% survival rate (median survival 23.5 months) versus 65.4% in patients with low or no FZD7 expression (median survival 77 months) [30].

Analysis of individual Wnt pathway markers, such as select ligands and receptors, is a useful tool for clinicians to predict prognosis and for researchers to determine the molecular mechanisms behind the tumour. However, such analysis often fails to uncover the whole picture. A broader look at the cancer transcriptome of the whole pathway and its numerous target genes is more suitable in this regard, as we have recently done for breast cancer. Exhaustive analysis of the TCGA and GTex databases has revealed that it is not single gene upregulation that is responsible for the aberrant signalling across the patients, but rather epigenetic dysregulation of the whole Wnt system. Such generalised dysregulation is behind the consistent pathway overactivation leading to uncontrolled cancer cell proliferation in breast cancer patients [27]. Network correlation analysis further permitted us to highlight signalling nodes within the Wnt pathway, which emerge as new promising drug targets and biomarkers in clinical studies and personalised medicine treatments [27].

Stemness and therapy resistance in Wnt dependent cancers

Apart from its involvement in tumourigenesis and cell proliferation, the Wnt pathway contributes to chemoresistance and cancer stem cell (CSC) propagation, the two factors ultimately responsible for tumour recurrence after therapy, metastasis and poor patient survival [31]. CSCs are a subpopulation of cancer cells; similarly to normal stem cells, they can self-renew or differentiate [32]. Being activated in CSCs, the Wnt pathway upregulates transcription of genes necessary for proliferation (such as c-myc) [33], cell cycling (such as cyclin-D) [34], anti-apoptosis (e.g., survivin) [31, 35], metabolic switching to aerobic glycolysis (PDK1, MCT-1) [36, 37], and invasion and metastasis (SLUG, MMP) [38, 39]. The role of active Wnt signalling in chemo- and radio-resistance is linked to the survival of CSCs: being relatively dormant, they can better withstand the therapy to repopulate the shrunken tumour, which results in tumour recurrence. There is also a separate mechanism of involvement of the Wnt pathway in cancer chemoresistence, mediated by the ill-famed multidrug resistance protein 1 (MDR1, also known as ABCB1 or P-glycoprotein) [40]. It was first demonstrated in early colorectal cancer that MDR1 is a target gene of the Wnt/β-catenin/TCF4 pathway, thus activation of the pathway led to increased levels of MDR1, increased drug efflux and drug resistance [41]. Similarly, increased MDR1 expression was found to be mediated by FZD1 in neuroblastoma, and a significant correlation in expression levels of FZD1 and MDR1 was found in patients relapsed after chemotherapy [42]. Other drug pumps involved in chemoresistance, ABCG2 (BCRP) and MRP2, were also shown to be induced by the Wnt pathway [43–45]. Finally, another contribution of the Wnt signalling to drug resistance is mediated by the DNA repair gene O6-methylguanin-DNA-methyltransferase (MGMT) in CSCs [46–48]. MGMT specifically repairs alkylated DNA and therefore upregulation of the protein leads to inefficiency of DNA alkylating agents and PARP inhibitors [49].

Wnt signalling plays several roles in tumour radioresistance. Firstly, radiotherapy induces upregulation of a panel of growth factors including Wnts, both in the tumour and in the surrounding stroma leading to enrichment of the CSC population [50]. Secondly, the Wnt pathway can directly protect against irradiation-induced DNA damage driving expression of DNA ligase 4 (LIG4) in colorectal cancer cells [51]. Further, histone modifier high-mobility group box 1 protein (HMGB1) involved in chromatin remodelling and DNA repair can be induced by Wnt/TCF4 signalling; blocking HMGB1 in oesophageal squamous cell carcinoma cells was found to suppress the Wnt1-dependent radioresistance [50].

Targeting the Wnt pathway is therefore beneficial at multiple levels: inhibition of tumour growth and survival with minimal effects on somatic cells, inhibition of CSC maintenance (and thus, of tumour relapse), and prevention of the development of tumour resistance to chemo- and radio-therapy. Inhibitors of the Wnt pathway are therefore in high therapeutic demand, and platforms dedicated to the search and development of such inhibitors are needed [52]. Although no Wnt-targeting drugs have yet reached the market, some are in preclinical and early clinical stages of development.

Wnt inhibitors in clinical development

The pathological and physiological roles of Wnt signalling, as well as the complexity of this pathway with its numerous sub-branches utilised in different cell types, underlie the practical difficulties in finding therapeutically relevant Wnt-targeting agents. The total number of Wnt pathway inhibitors active in vitro is around fifty [53, 54], many of them having reached different stages of preclinical development, but so far only a few have attained early phase clinical trials.

Figure 1 shows the anti-Wnt agents that have reached clinical trials. It can be appreciated that these agents target the pathway at the levels where the pathway diverges into several sub-pathways (perhaps with the exception of the Porcupine-targeting drugs, but see below). Such diversification into sub-pathways can be found at the level of the plasma membrane, as well as in the nucleus; in contrast, the events in the cytoplasm are poorly diversified and are rather common for all the Wnt signalling subtypes [55, 56]. For a drug candidate being selected for clinical studies, we consider it critical for it to act on Wnt signalling subtypes active specifically in pathological tissues, instead of affecting all Wnt signalling subtypes. Indeed, pan-Wnt inhibitors fail to show acceptable safety profiles at preclinical levels and cannot advance further, as exemplified by attempts to develop tankyrase inhibitors [57, 58] or Dickkopf-1 as a biologic to block the Wnt pathway [59].

Figure 1 Drug compounds of past or current clinical trials target the Wnt pathway at the ligand/receptor level (rosmantuzumab, ipafricept, vantictumab and Foxy-5) or at the transcriptional level (CWP232291 and PRI-724). Porcupine inhibitors (WNT974 and ETC-159) target Wnt secretion.

Vantictumab, ipafricept and rosmantuzumab

The only agent directly targeting FZDs having entered clinical development is a humanised antibody vantictumab (OMP-18R5). Initially developed against the Wnt-binding CRD-domain of FZD7, it was found to act on FZD1, 2, 5, 7 and 8 – five FZDs out of the ten encoded by the human genome [60]. The preclinical activity profile against a panel of tumour cells [60] prompted it’s entry into phase I clinical trials (NCT01345201, NCT01973309, NCT02005315 and NCT01957007). All the trials have been completed by now and reports are available for the first three. The first phase Ia study measured the dose escalation effects following the intravenous administration of doses ranging between 0.5mg/kg weekly and 2.5mg/kg once in 3 weeks. The main finding was bone toxicity manifested as a bone fracture on day 110 in one patient. Other adverse events were fatigue, vomiting, abdominal pain, constipation, diarrhoea and nausea of grades 1 and 2, with grade 3 diarrhoea and vomiting reported in one patient. To tackle bone toxicity, the study monitored β-C-terminal telopeptide (β-CTX), a marker for bone degradation, and was able to manage its levels by administering zolendronic acid [61]. The other two studies, phase Ib on pancreatic and breast cancer using vantictumab in combination with paclitaxel (90 g/m2) or nab-paclitaxel (125 g/m2), adopted the same strategy for tackling the bone fragility and reported similar adverse effects of grade 2 observed in phase Ia and few additional grade 3 events (neutropenia, leukopenia, pelvic pain, fatigue and nausea). Both studies used increased vantictumab dosages (between 3.5 and 14 mg/kg) and reported further bone fragility events, which required improvements in the zolendronic acid administration regimen and resulted in a temporary halt of the trials in 2014 [62–64].

Similar results were obtained for ipafricept (OMP-54F28) – another anti-Wnt biologic from OncoMed, representing the FZD8 CRD-domain fused to an IgG1 constant fragment. By mechanism, this compound can be expected to possess a certain selectivity towards a subset of Wnt proteins, although its specificity among the 19 Wnts encoded by the human genome is unclear because of the lack of comprehensive data on mutual affinities of Wnt and FZD proteins. Four clinical trials were launched for ipafricept (NCT01608867, NCT02092363, NCT02069145 and NCT02050178) with two of them reporting the results. As with vantictumab, the trials used the scheme with zolendronic acid to counteract the bone-related adverse effects, apparently with more success since only one fracture was recorded at 20 mg/kg; the on-target dose was estimated to be at >10mg/kg. Non-bone-related adverse events with the compound included grade 1 and 2 dysgeusia, decreased appetite, fatigue, muscle spasms, alopecia and vomiting and grade 3 events such as anaemia hypophosphataemia, neutropenia and weight loss [65, 66].

Finally, Oncomed had one more anti-Wnt compound in its portfolio – the R-spondin 3-targeting antibody rosmantuzumab (OMP-131R10). R-spondins are soluble ligands that enhance Wnt signal transduction, especially the canonical branch, through different mechanisms [10, 67]. The phase Ia/b clinical trial (NCT02482441) of the agent demonstrated a set of adverse effects similar to the other two agents of the company: doses from 2.5 to 15 mg/kg every 2 weeks resulted in nausea, decreased appetite, diarrhoea, vomiting and weight decrease of unspecified grades. Additionally, the treatment resulted in changes in bone turnover markers – which is somewhat unexpected since R-spondin 3 (unlike related R-spondin 1 and 2) is not known to be involved in bone formation and maintenance [68, 69]. This might hint towards insufficient specificity of rosmantuzumab, which is difficult to assess since no preclinical report was published for the agent.

Overall, one may conclude that vantictumab, ipafricept and rosmantuzumab, biologics interfering with Wnt signalling at the level of the ligands, receptors and extracellular enhancers, which have been designed to achieve selectivity in targeting different Wnt signalling subtypes, have ultimately revealed poorer specificity than intended. The similar adverse effects of the three drug candidates in safety clinical trials suggest a too-general wiping out of the Wnt pathway instead of the selective inhibition of the pathway subtype active in the tumour. These adverse effects were likely behind the strategic decisions made regarding the drugs: in 2017, Bayer opted out of licensing vantictumab or ipafricept from Oncomed for “strategic reasons”; rosmantuzumab was stated to have “failed to provide compelling evidence of clinical benefit” [70]. These decisions resulted in discontinuation of the clinical development of the three candidates. Since the molecules did not advance beyond the safety trials, conclusions on compound efficacy in human subjects could not be drawn.

Porcupine inhibitors WNT974 (LKG974) and ETC-159 (ETC-1922159)

Another clinically relevant attempt to inhibit Wnt signalling at the upstream levels is currently spearheaded by two competing inhibitors of Porcupine, the acyltransferase responsible for posttranslational modification of all Wnt proteins. By conception, molecules of this type were supposed to be pan-Wnt inhibitors, preventing both autocrine and paracrine signalling since the acylation is considered an absolute prerequisite for the Wnt protein secretion and activity [71]. However, as described below, both molecules show quite acceptable preclinical and clinical safety profiles, which might be explained by novel insights into the signalling by non-acylated Wnts, meaning that inhibition of acylation might affect the pathway only partially [72]. Both competitors from Novartis (WNT974) and Singapore State D3 consortium (ETC-159) have successfully passed preclinical investigations, with significant reduction of the tumour burden and no toxicity – either overt or at the level of tissue morphology following the analysis of several Wnt-dependent tissues. In phase I clinical trials (NCT01351103 for WNT974 and NCT02521844 for ETC-159), both were tested at similar doses – 5 to 30 mg/day for WNT974 and 1 to 30 mg for ETC-159. The more representative study of WNT974, enrolling 94 patients by 2017, showed that it induces grade 1 and 2 dysgeusia, decreased appetite, nausea, fatigue, diarrhoea, vomiting, hypercalcaemia, alopecia, asthenia and hypomagnesaemia. Additionally, in a small number (3–4%) of patients, grade 3 and 4 adverse events included asthenia, fatigue, decreased appetite and enteritis [73, 74]. Analysis of tumour specimens for various markers has shown a profound Wnt inhibitory effect; moreover, in some patients the compound was used in combination with spartalizumab (an anti-PD-1 antibody), giving a positive outlook on potential combination of the anti-Wnt and immune-therapy. Surprisingly, authors do not report any events or even any attempts to follow bone-related effects, which is in contrast to the ETC-159 trial. The latter enrolled 16 patients and the study reported vomiting, anorexia, fatigue, dysgeusia and constipation as the adverse events of unspecified grade identified in >20% of patients. Beta-CTX levels were analysed and found to be expectedly elevated in two patients with concomitant loss in bone density, which was counteracted by vitamin D and calcium supplements [75].

Although that WNT974 was shown to affect bone structure in animals [76, 77], the effect does not seem to manifest clinically. Perhaps this is a reason why WNT974 is currently the most advanced anti-Wnt agent and the only one to have moved to a phase II trial (NCT02649530), and therefore might become the first anti-Wnt agent with a comprehensive assessment of clinical pharmacodynamics.

Wnt5a-mimetic Foxy-5

An interesting approach to Wnt inhibition is employed by the WntResearch start-up company from Sweden, which has identified a Wnt5a-mimicking peptide called Foxy-5 as an efficient anti-metastatic agent [78]. Specific to the compound is its intervention into the non-canonical Wnt pathway, suppressing migration and adhesion of breast cancer cells. Therefore, this approach does not target tumour bulk, but is rather oriented to metastasis prevention and is used in combination with surgery, irradiation and other drugs. The compound has passed a phase I clinical trial [79, 80]. According to the sparse information provided, Foxy-5 was reported to be “non-toxic” at any dose, and showing good pharmacokinetics and stabilisation of the levels of circulating tumour cell in patients with metastatic breast, colon, or prostate cancer [81]. Currently the company has reported recruitment of the first patient for the phase II study. This study will compare patients undergoing colon cancer surgery followed by a 6 month regimen with FOLFOX with patients receiving a treatment of Foxy-5 before and after surgery until starting the FOLFOX regimen (NCT03883802).

Downstream pathway component inhibitors PRI-724 and CWP232291

These two drug candidates make use of the “downstream target window” to achieve the necessary specificity (fig. 1). Both compounds affect the pathway at the transcriptional level, but through entirely different mechanisms: the small molecule PRI-724 affects interaction of β-catenin with transcription co-activator CBP, whereas the peptidomimetic CWP232291 (sometimes called CWP-291) binds to Sam68, an RNA-binding protein that regulates alternative splicing of the TCF-1 transcription factor in a complex with CBP. This selectivity of the compounds towards the Wnt pathway components employed by cancer cells allowed both PRI-724 (as its early analogue ICG-001) [82–84] and CWP232291 [85] to succeed in the preclinical setting and enter phase I clinical trials.

PRI-724 was tested in three phase I trials: in patients with advanced solid tumours (NCT01302405), acute and chronic myeloid leukaemia (NCT01606579), and pancreatic cancer (NCT01764477). In the phase I trials including 18 patients, the compound showed a promising safety profile with only dose-limiting grade 3 hyperbilirubinaemia registered in one patient (out of 7 presenting grade 3 events) at the highest dose of the compound (1280 mg/m2/day). Grade 2 adverse events included diarrhoea, bilirubin elevation, hypophosphataemia, nausea, fatigue, anorexia, thrombocytopenia and alkaline phosphatase elevation. The compound also showed a decrease in survivin expression in circulating tumour cells as an efficacy readout in colon cancer subjects [86]. At the same dosage, no grade 3 adverse events were recorded for refractory leukaemia patients, with only four cases of grade 1 nausea, vomiting and diarrhoea attributed to the drug. Analysis of patient samples demonstrated a 44% median blast decrease [87]. However, in the third study, when combined with gemcitabine against pancreatic adenocarcinoma, the compound induced seven grade 3 and 4 adverse events in 20 patients, inducing abdominal pain, neutropenia, anaemia, fatigue and alkaline phosphatase rise. Stable disease was observed in 40% of patients. Despite this somewhat worse performance, none of the adverse events met the dose-limiting definition, thus the combination was considered overall safe with “modest clinical activity” [88]. Interestingly, since CBP/β-catenin interaction was found to be important during the onset of liver fibrosis, PRI-724 is in clinical trials against this disease as well, reporting similar adverse reactions against the background of a clinical benefit [89]. This later indication is being continued, as currently a phase I/II study is announced for PRI-724 in fibrosis, whereas for its anti-cancer application no follow-up is in sight.

Peptidomimetic CWP232291 was used in a single phase I trial in patients with relapsed and refractory acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS) comprising 56 patients. Grade 3 and 4 events made up 9% of the all recorded adverse events and included fever, nausea and anaphylactic reaction, with the first two being dose limiting. Grade 1 and 2 adverse events included nausea, infusion-related reaction, vomiting, diarrhoea and anorexia. There were also some indications of efficacy, since remission was observed in one patient and reduction in β-catenin and survivin as markers consistently observed in other subjects.

Conclusions and perspectives

The agents described above result from different approaches to inhibiting the Wnt pathway in cancer and are clearly unified by one motif: in order to target the pathway, one needs to identify disease-specific vulnerabilities in it to avoid systemic toxicity. Such specific vulnerabilities in the Wnt pathway are best to be found among its most divergent levels of the pathway – the one at the plasma membrane and the one inside the nucleus [55]. It should be noted that in the current review we have focused on the de novo and dedicated Wnt-targeting compounds; however, a wealth of Wnt inhibitors have been found among already approved drugs, prompting attempts to reposition them against Wnt-dependent cancers, as reviewed by us and elsewhere [90, 91]. Our own recent preclinical study shows that clofazimine, known as an anti-leprosy agent with a well-established safety profile, can efficiently inhibit Wnt signalling at the doses comparable to those used against leprosy and is safe to administer in combination with chemotherapy [92, 93]. Other well-known small molecule compounds, such as niclosamide, sulindac, pimozide show promise in various preclinical studies and will hopefully soon appear in clinical studies. Other Wnt inhibitors, including some natural products, may turn out to be promising agents against select Wnt-dependent cancers [94, 95]. Future developments will show whether the new wave of effort to target the “undruggable” Wnt pathway will bear fruit [96]. The main message of our review is that, in order to be successful, such efforts should aim not at the Wnt pathway as a whole, but at a particular variant of it, selectively active in a disease state.

Notes

Disclosure statement

No financial support and no other potential conflict of interest relevant to this article was reported.

* Contributed equally to the manuscript

References

1 Uings IJ , Farrow SN . Cell receptors and cell signalling. Mol Pathol. 2000;53(6):295–9. doi:.https://doi.org/10.1136/mp.53.6.295

2 Nusse R . Wnt signaling in disease and in development. Cell Res. 2005;15(1):28–32. doi:.https://doi.org/10.1038/sj.cr.7290260

3 Komiya Y , Habas R . Wnt signal transduction pathways. Organogenesis. 2008;4(2):68–75. doi:.https://doi.org/10.4161/org.4.2.5851

4 Schulte G . Frizzleds and WNT/β-catenin signaling--The black box of ligand-receptor selectivity, complex stoichiometry and activation kinetics. Eur J Pharmacol. 2015;763(Pt B):191–5. doi:.https://doi.org/10.1016/j.ejphar.2015.05.031

5 Roy L , Cowden Dahl KD . Can Stemness and Chemoresistance Be Therapeutically Targeted via Signaling Pathways in Ovarian Cancer? Cancers (Basel). 2018;10(8):241. doi:.https://doi.org/10.3390/cancers10080241

6 MacDonald BT , He X . Frizzled and LRP5/6 receptors for Wnt/β-catenin signaling. Cold Spring Harb Perspect Biol. 2012;4(12):a007880. doi:.https://doi.org/10.1101/cshperspect.a007880

7 Miller JR . The Wnts. Genome Biol. 2002;3(1):S3001.  

8 Dijksterhuis JP , Baljinnyam B , Stanger K , Sercan HO , Ji Y , Andres O , et al. Systematic mapping of WNT-FZD protein interactions reveals functional selectivity by distinct WNT-FZD pairs. J Biol Chem. 2015;290(11):6789–98. doi:.https://doi.org/10.1074/jbc.M114.612648

9 Cruciat C-M , Niehrs C . Secreted and transmembrane wnt inhibitors and activators. Cold Spring Harb Perspect Biol. 2013;5(3):a015081. doi:.https://doi.org/10.1101/cshperspect.a015081

10 de Lau WB , Snel B , Clevers HC . The R-spondin protein family. Genome Biol. 2012;13(3):242. doi:.https://doi.org/10.1186/gb-2012-13-3-242

11 Topol L , Jiang X , Choi H , Garrett-Beal L , Carolan PJ , Yang Y . Wnt-5a inhibits the canonical Wnt pathway by promoting GSK-3-independent β-catenin degradation. J Cell Biol. 2003;162(5):899–908. doi:.https://doi.org/10.1083/jcb.200303158

12 Kahn M . Wnt Signaling in Stem Cells and Cancer Stem Cells: A Tale of Two Coactivators. Prog Mol Biol Transl Sci. 2018;153:209–44. doi:.https://doi.org/10.1016/bs.pmbts.2017.11.007

13 Krausova M , Korinek V . Wnt signaling in adult intestinal stem cells and cancer. Cell Signal. 2014;26(3):570–9. doi:.https://doi.org/10.1016/j.cellsig.2013.11.032

14 Malhotra S , Kincade PW . Wnt-related molecules and signaling pathway equilibrium in hematopoiesis. Cell Stem Cell. 2009;4(1):27–36. doi:.https://doi.org/10.1016/j.stem.2008.12.004

15 Houschyar KS , Tapking C , Borrelli MR , Popp D , Duscher D , Maan ZN , et al. Wnt Pathway in Bone Repair and Regeneration - What Do We Know So Far. Front Cell Dev Biol. 2019;6:170. doi:.https://doi.org/10.3389/fcell.2018.00170

16 Reya T , Clevers H . Wnt signalling in stem cells and cancer. Nature. 2005;434(7035):843–50. doi:.https://doi.org/10.1038/nature03319

17 De A . Wnt/Ca2+ signaling pathway: a brief overview. Acta Biochim Biophys Sin (Shanghai). 2011;43(10):745–56. doi:.https://doi.org/10.1093/abbs/gmr079

18 Kurayoshi M , Oue N , Yamamoto H , Kishida M , Inoue A , Asahara T , et al. Expression of Wnt-5a is correlated with aggressiveness of gastric cancer by stimulating cell migration and invasion. Cancer Res. 2006;66(21):10439–48. doi:.https://doi.org/10.1158/0008-5472.CAN-06-2359

19 Corda G , Sala G , Lattanzio R , Iezzi M , Sallese M , Fragassi G , et al. Functional and prognostic significance of the genomic amplification of frizzled 6 (FZD6) in breast cancer. J Pathol. 2017;241(3):350–61. doi:.https://doi.org/10.1002/path.4841

20 Weeraratna AT , Jiang Y , Hostetter G , Rosenblatt K , Duray P , Bittner M , et al. Wnt5a signaling directly affects cell motility and invasion of metastatic melanoma. Cancer Cell. 2002;1(3):279–88. doi:.https://doi.org/10.1016/S1535-6108(02)00045-4

21 Rowan AJ , Lamlum H , Ilyas M , Wheeler J , Straub J , Papadopoulou A , et al. APC mutations in sporadic colorectal tumors: A mutational “hotspot” and interdependence of the “two hits”. Proc Natl Acad Sci USA. 2000;97(7):3352–7. doi:.https://doi.org/10.1073/pnas.97.7.3352

22 Schneikert J , Behrens J . The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut. 2007;56(3):417–25. doi:.https://doi.org/10.1136/gut.2006.093310

23 Bruun J , Kolberg M , Nesland JM , Svindland A , Nesbakken A , Lothe RA . Prognostic Significance of β-Catenin, E-Cadherin, and SOX9 in Colorectal Cancer: Results from a Large Population-Representative Series. Front Oncol. 2014;4:118. doi:.https://doi.org/10.3389/fonc.2014.00118

24 Kamposioras K , Konstantara A , Kotoula V , Lakis S , Kouvatseas G , Akriviadis E , et al. The prognostic significance of WNT pathway in surgically-treated colorectal cancer: β-catenin expression predicts for disease-free survival. Anticancer Res. 2013;33(10):4573–84.  

25 Yoshida N , Kinugasa T , Ohshima K , Yuge K , Ohchi T , Fujino S , et al. Analysis of Wnt and β-catenin Expression in Advanced Colorectal Cancer. Anticancer Res. 2015;35(8):4403–10.  

26 Veloudis G , Pappas A , Gourgiotis S , Falidas E , Dimitriou N , Karavokiros I , et al. Assessing the clinical utility of Wnt pathway markers in colorectal cancer. J BUON. 2017;22(2):431–6.  

27 Koval A , Katanaev VL . Dramatic dysbalancing of the Wnt pathway in breast cancers. Sci Rep. 2018;8(1):7329. doi:.https://doi.org/10.1038/s41598-018-25672-6

28 Xu X , Sun P-L , Li J-Z , Jheon S , Lee C-T , Chung J-H . Aberrant Wnt1/β-catenin expression is an independent poor prognostic marker of non-small cell lung cancer after surgery. J Thorac Oncol. 2011;6(4):716–24. doi:.https://doi.org/10.1097/JTO.0b013e31820c5189

29 Zeng C-M , Chen Z , Fu L . Frizzled Receptors as Potential Therapeutic Targets in Human Cancers. Int J Mol Sci. 2018;19(5):1543. doi:.https://doi.org/10.3390/ijms19051543

30 Li G , Su Q , Liu H , Wang D , Zhang W , Lu Z , 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(3):280–93. doi:.https://doi.org/10.7150/ijbs.23756

31 Reya T , Morrison SJ , Clarke MF , Weissman IL . Stem cells, cancer, and cancer stem cells. Nature. 2001;414(6859):105–11. doi:.https://doi.org/10.1038/35102167

32 Batlle E , Clevers H . Cancer stem cells revisited. Nat Med. 2017;23(10):1124–34. doi:.https://doi.org/10.1038/nm.4409

33 Wang H , Mannava S , Grachtchouk V , Zhuang D , Soengas MS , Gudkov AV , et al. c-Myc depletion inhibits proliferation of human tumor cells at various stages of the cell cycle. Oncogene. 2008;27(13):1905–15. doi:.https://doi.org/10.1038/sj.onc.1210823

34 Klein EA , Assoian RK . Transcriptional regulation of the cyclin D1 gene at a glance. J Cell Sci. 2008;121(23):3853–7. doi:.https://doi.org/10.1242/jcs.039131

35 Jaiswal PK , Goel A , Mittal RD . Survivin: A molecular biomarker in cancer. Indian J Med Res. 2015;141(4):389–97. doi:.https://doi.org/10.4103/0971-5916.159250

36 Pate KT , Stringari C , Sprowl-Tanio S , Wang K , TeSlaa T , Hoverter NP , et al. Wnt signaling directs a metabolic program of glycolysis and angiogenesis in colon cancer. EMBO J. 2014;33(13):1454–73. doi:.https://doi.org/10.15252/embj.201488598

37 Sprowl-Tanio S , Habowski AN , Pate KT , McQuade MM , Wang K , Edwards RA , et al. Lactate/pyruvate transporter MCT-1 is a direct Wnt target that confers sensitivity to 3-bromopyruvate in colon cancer. Cancer Metab. 2016;4(1):20. doi:.https://doi.org/10.1186/s40170-016-0159-3

38 Lowy AM , Clements WM , Bishop J , Kong L , Bonney T , Sisco K , et al. β-Catenin/Wnt signaling regulates expression of the membrane type 3 matrix metalloproteinase in gastric cancer. Cancer Res. 2006;66(9):4734–41. doi:.https://doi.org/10.1158/0008-5472.CAN-05-4268

39 Wu Z-Q , Li X-Y , Hu CY , Ford M , Kleer CG , Weiss SJ . Canonical Wnt signaling regulates Slug activity and links epithelial-mesenchymal transition with epigenetic Breast Cancer 1, Early Onset (BRCA1) repression. Proc Natl Acad Sci USA. 2012;109(41):16654–9. doi:.https://doi.org/10.1073/pnas.1205822109

40 Lage H . Gene Therapeutic Approaches to Overcome ABCB1-Mediated Drug Resistance. Recent Results Cancer Res. 2016;209:87–94. doi:.https://doi.org/10.1007/978-3-319-42934-2_6

41 Yamada T , Takaoka AS , Naishiro Y , Hayashi R , Maruyama K , Maesawa C , et al. Transactivation of the multidrug resistance 1 gene by T-cell factor 4/β-catenin complex in early colorectal carcinogenesis. Cancer Res. 2000;60(17):4761–6.  

42 Flahaut M , Meier R , Coulon A , Nardou KA , Niggli FK , Martinet D , et al. The Wnt receptor FZD1 mediates chemoresistance in neuroblastoma through activation of the Wnt/β-catenin pathway. Oncogene. 2009;28(23):2245–56. doi:.https://doi.org/10.1038/onc.2009.80

43 Zhang Z-M , Wu J-F , Luo Q-C , Liu Q-F , Wu Q-W , Ye G-D , et al. Pygo2 activates MDR1 expression and mediates chemoresistance in breast cancer via the Wnt/β-catenin pathway. Oncogene. 2016;35(36):4787–97. doi:.https://doi.org/10.1038/onc.2016.10

44 Chikazawa N , Tanaka H , Tasaka T , Nakamura M , Tanaka M , Onishi H , et al. Inhibition of Wnt signaling pathway decreases chemotherapy-resistant side-population colon cancer cells. Anticancer Res. 2010;30(6):2041–8.  

45 Chau WK , Ip CK , Mak ASC , Lai H-C , Wong AST . c-Kit mediates chemoresistance and tumor-initiating capacity of ovarian cancer cells through activation of Wnt/β-catenin-ATP-binding cassette G2 signaling. Oncogene. 2013;32(22):2767–81. doi:.https://doi.org/10.1038/onc.2012.290

46 Wickström M , Dyberg C , Milosevic J , Einvik C , Calero R , Sveinbjörnsson B , et al. Wnt/β-catenin pathway regulates MGMT gene expression in cancer and inhibition of Wnt signalling prevents chemoresistance. Nat Commun. 2015;6(1):8904. doi:.https://doi.org/10.1038/ncomms9904

47 Johnsen JI , Wickström M , Baryawno N . Wingless/β-catenin signaling as a modulator of chemoresistance in cancer. Mol Cell Oncol. 2016;3(2):e1131356. doi:.https://doi.org/10.1080/23723556.2015.1131356

48 Li Z-Y , Huang G-D , Chen L , Zhang C , Chen B-D , Li Q-Z , et al. Tanshinone IIA induces apoptosis via inhibition of Wnt/β‑catenin/MGMT signaling in AtT‑20 cells. Mol Med Rep. 2017;16(5):5908–14. doi:.https://doi.org/10.3892/mmr.2017.7325

49 Yamamoto TM , McMellen A , Watson ZL , Aguilera J , Sikora MJ , Ferguson R , et al. Targeting Wnt Signaling To Overcome PARP Inhibitor Resistance. bioRxiv. 2018;378463. Preprint. doi: 

50 Zhao Y , Tao L , Yi J , Song H , Chen L . The Role of Canonical Wnt Signaling in Regulating Radioresistance. Cell Physiol Biochem. 2018;48(2):419–32. doi:.https://doi.org/10.1159/000491774

51 Jun S , Jung Y-S , Suh HN , Wang W , Kim MJ , Oh YS , et al. LIG4 mediates Wnt signalling-induced radioresistance. Nat Commun. 2016;7(1):10994. doi:.https://doi.org/10.1038/ncomms10994

52 Shaw HV , Koval A , Katanaev VL . A high-throughput assay pipeline for specific targeting of frizzled GPCRs in cancer. Methods Cell Biol. 2019;149:57–75. doi:.https://doi.org/10.1016/bs.mcb.2018.08.006

53Lee HJ, Zhang X, Zheng JJ. Inhibiting the Wnt Signaling Pathway with Small Molecules. In Goss KH, Kahn, M (eds): Targeting the Wnt Pathway in Cancer. Berlin: springer; 2011. pp 183–209. 

54Wnt Homepage. Small molecules in Wnt signaling | The Wnt Homepage [Internet]. 2019 [cited 2019 May 16]. Available from: https://web.stanford.edu/group/nusselab/cgi-bin/wnt/smallmolecules

55 Blagodatski A , Poteryaev D , Katanaev VL . Targeting the Wnt pathways for therapies. Mol Cell Ther. 2014;2(1):28. doi:.https://doi.org/10.1186/2052-8426-2-28

56 Koval A , Katanaev VL . Platforms for high-throughput screening of Wnt/Frizzled antagonists. Drug Discov Today. 2012;17(23-24):1316–22. doi:.https://doi.org/10.1016/j.drudis.2012.07.007

57 Zhong Y , Katavolos P , Nguyen T , Lau T , Boggs J , Sambrone A , et al. Tankyrase Inhibition Causes Reversible Intestinal Toxicity in Mice with a Therapeutic Index < 1. Toxicol Pathol. 2016;44(2):267–78. doi:.https://doi.org/10.1177/0192623315621192

58 Mariotti L , Pollock K , Guettler S . Regulation of Wnt/β-catenin signalling by tankyrase-dependent poly(ADP-ribosyl)ation and scaffolding. Br J Pharmacol. 2017;174(24):4611–36. doi:.https://doi.org/10.1111/bph.14038

59 Aguilera O , Peña C , García JM , Larriba MJ , Ordóñez-Morán P , Navarro D , et al. The Wnt antagonist DICKKOPF-1 gene is induced by 1α,25-dihydroxyvitamin D3 associated to the differentiation of human colon cancer cells. Carcinogenesis. 2007;28(9):1877–84. doi:.https://doi.org/10.1093/carcin/bgm094

60 Gurney A , Axelrod F , Bond CJ , Cain J , Chartier C , Donigan L , 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(29):11717–22. doi:.https://doi.org/10.1073/pnas.1120068109

61 Smith DC , Rosen LS , Chugh R , Goldman JW , Xu L , Kapoun A , et al. First-in-human evaluation of the human monoclonal antibody vantictumab (OMP-18R5; anti-Frizzled) targeting the WNT pathway in a phase I study for patients with advanced solid tumors. J Clin Oncol. 2013;31(15_suppl):2540. 

62 Mita MM , Becerra C , Richards DA , Mita AC , Shagisultanova E , Osborne CRC , et al. Phase 1b study of WNT inhibitor vantictumab (VAN, human monoclonal antibody) with paclitaxel (P) in patients (pts) with 1st- to 3rd-line metastatic HER2-negative breast cancer (BC). J Clin Oncol. 2016;34(15_suppl):2516. doi:.https://doi.org/10.1200/JCO.2016.34.15_suppl.2516

63 Messersmith W , Cohen S , Shahda S , Lenz H-J , Weekes C , Dotan E , et al. Phase 1b study of WNT inhibitor vantictumab (VAN, human monoclonal antibody) with nab-paclitaxel (Nab-P) and gemcitabine (G) in patients (pts) with previously untreated stage IV pancreatic cancer (PC). Ann Oncol. 2016;27(6_suppl, suppl_6). doi:.https://doi.org/10.1093/annonc/mdw371.69

64 Davis SL , Cardin DB , Shahda S , Lenz H-J , Dotan E , O’Neil BH , et al. A phase 1b dose escalation study of Wnt pathway inhibitor vantictumab in combination with nab-paclitaxel and gemcitabine in patients with previously untreated metastatic pancreatic cancer. Invest New Drugs. 2019. doi:.https://doi.org/10.1007/s10637-019-00824-1

65 Jimeno A , Gordon M , Chugh R , Messersmith W , Mendelson D , Dupont J , 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(24):7490–7. doi:.https://doi.org/10.1158/1078-0432.CCR-17-2157

66 Tai D , Wells K , Arcaroli J , Vanderbilt C , Aisner DL , Messersmith WA , et al. Targeting the WNT Signaling Pathway in Cancer Therapeutics. Oncologist. 2015;20(10):1189–98. doi:.https://doi.org/10.1634/theoncologist.2015-0057

67 Kim K-A , Zhao J , Andarmani S , Kakitani M , Oshima T , Binnerts ME , et al. R-Spondin proteins: a novel link to β-catenin activation. Cell Cycle. 2006;5(1):23–6. doi:.https://doi.org/10.4161/cc.5.1.2305

68 Knight MN , Karuppaiah K , Lowe M , Mohanty S , Zondervan RL , Bell S , et al. R-spondin-2 is a Wnt agonist that regulates osteoblast activity and bone mass. Bone Res. 2018;6(1):24. doi:.https://doi.org/10.1038/s41413-018-0026-7

69 Wang H , Brennan TA , Russell E , Kim J-H , Egan KP , Chen Q , et al. R-Spondin 1 promotes vibration-induced bone formation in mouse models of osteoporosis. J Mol Med (Berl). 2013;91(12):1421–9. doi:.https://doi.org/10.1007/s00109-013-1068-3

70Oncomed Annual Report [Internet]. 2017. Available from: http://www.oncomed.com/SEC-Documents/0001564590-18-005014.pdf

71 Janda CY , Waghray D , Levin AM , Thomas C , Garcia KC . Structural basis of Wnt recognition by Frizzled. Science. 2012;337(6090):59–64. doi:.https://doi.org/10.1126/science.1222879

72 Speer KF , Sommer A , Tajer B , Mullins MC , Klein PS , Lemmon MA . Non-acylated Wnts can promote signaling. Cell Rep. 2019;26(4):875–883.e5. doi:.https://doi.org/10.1016/j.celrep.2018.12.104

73 Janku F , Connolly R , LoRusso P , de Jonge M , Vaishampayan U , Rodon J , et al. Abstract C45: Phase I study of WNT974, a first-in-class Porcupine inhibitor, in advanced solid tumors. Mol Cancer Ther. 2015;14(12, Supplement 2):C45. 

74 Rodon J , Argilés G , Connolly RM , Vaishampayan U , de Jonge M , Garralda E , et al. Abstract CT175: Biomarker analyses from a phase I study of WNT974, a first-in-class Porcupine inhibitor, in patients (pts) with advanced solid tumors. Cancer Res. 2018;78(13, Supplement):CT175. 

75 Ng M , Tan DS , Subbiah V , Weekes CD , Teneggi V , Diermayr V , et al. First-in-human phase 1 study of ETC-159 an oral PORCN inhbitor in patients with advanced solid tumours. J Clin Oncol. 2017;35(15_suppl):2584. doi:.https://doi.org/10.1200/JCO.2017.35.15_suppl.2584

76 Funck-Brentano T , Nilsson KH , Brommage R , Henning P , Lerner UH , Koskela A , et al. Porcupine inhibitors impair trabecular and cortical bone mass and strength in mice. J Endocrinol. 2018;238(1):13–23. doi:.https://doi.org/10.1530/JOE-18-0153

77 Moon J , Zhou H , Zhang LS , Tan W , Liu Y , Zhang S , et al. Blockade to pathological remodeling of infarcted heart tissue using a porcupine antagonist. Proc Natl Acad Sci USA. 2017;114(7):1649–54. doi:.https://doi.org/10.1073/pnas.1621346114

78 Säfholm A , Leandersson K , Dejmek J , Nielsen CK , Villoutreix BO , Andersson T . A formylated hexapeptide ligand mimics the ability of Wnt-5a to impair migration of human breast epithelial cells. J Biol Chem. 2006;281(5):2740–9. doi:.https://doi.org/10.1074/jbc.M508386200

79 Soerensen PG , Andersson T , Buhl U , Moelvadgaard T , Jensen PB , Brunner N , et al. Phase I dose-escalating study to evaluate the safety, tolerability, and pharmacokinetic and pharmacodynamic profiles of Foxy-5 in patients with metastatic breast, colorectal, or prostate cancer. J Clin Oncol. 2014;32(15_suppl):TPS1140. doi:.https://doi.org/10.1200/jco.2014.32.15_suppl.tps1140

80 Andersson T , Axelsson L , Mohapatra P , Prasad C , Soerensen PG , Mau-Soerensen M , et al. Abstract A116: Targeting the Wnt-5a signaling pathway as a novel anti-metastatic therapy. Mol Cancer Ther. 2015;14(12, Supplement 2):A116. 

81WntResearch. A commentary on the interim Foxy-5 phase 1 study report. 2015. Available at: https://www.wntresearch.com/wp-content/uploads/2019/03/a-commentary-on-the-interim-foxy-5-phase-1-study-report-final-101115.pdf

82 Gang EJ , Hsieh Y-T , Pham J , Zhao Y , Nguyen C , Huantes S , et al. Small-molecule inhibition of CBP/catenin interactions eliminates drug-resistant clones in acute lymphoblastic leukemia. Oncogene. 2014;33(17):2169–78. doi:.https://doi.org/10.1038/onc.2013.169

83 He K , Xu T , Xu Y , Ring A , Kahn M , Goldkorn A . Cancer cells acquire a drug resistant, highly tumorigenic, cancer stem-like phenotype through modulation of the PI3K/Akt/β-catenin/CBP pathway. Int J Cancer. 2014;134(1):43–54. doi:.https://doi.org/10.1002/ijc.28341

84 Wend P , Fang L , Zhu Q , Schipper JH , Loddenkemper C , Kosel F , et al. Wnt/β-catenin signalling induces MLL to create epigenetic changes in salivary gland tumours. EMBO J. 2013;32(14):1977–89. doi:.https://doi.org/10.1038/emboj.2013.127

85 Cha JY , Jung J-E , Lee K-H , Briaud I , Tenzin F , Jung HK , et al. Anti-Tumor Activity of Novel Small Molecule Wnt Signaling Inhibitor, CWP232291, In Multiple Myeloma. Blood. 2010;116(21):3038. 

86 El-Khoueiry AB , Ning Y , Yang D , Cole S , Kahn M , Zoghbi M , et al. A phase I first-in-human study of PRI-724 in patients (pts) with advanced solid tumors. J Clin Oncol. 2013;31(15_suppl):2501. 

87 Foundation FS . 19th Congress of the European Hematology Association, Milan, Italy, June 12–15, 2014. Haematologica. 2014;99(supplement 1):1–796. 

88 Ko AH , Chiorean EG , Kwak EL , Lenz H-J , Nadler PI , Wood DL , et al. Final results of a phase Ib dose-escalation study of PRI-724, a CBP/beta-catenin modulator, plus gemcitabine (GEM) in patients with advanced pancreatic adenocarcinoma (APC) as second-line therapy after FOLFIRINOX or FOLFOX. J Clin Oncol. 2016;34(15_suppl):e15721. doi:.https://doi.org/10.1200/JCO.2016.34.15_suppl.e15721

89 Nishikawa K , Osawa Y , Kimura K . Wnt/β-Catenin Signaling as a Potential Target for the Treatment of Liver Cirrhosis Using Antifibrotic Drugs. Int J Mol Sci. 2018;19(10):3103. doi:.https://doi.org/10.3390/ijms19103103

90 Ahmed K , Shaw HV , Koval A , Katanaev VL . A Second WNT for Old Drugs: Drug Repositioning against WNT-Dependent Cancers. Cancers (Basel). 2016;8(7):66. doi:.https://doi.org/10.3390/cancers8070066

91 Harb J , Lin P-J , Hao J . Recent Development of Wnt Signaling Pathway Inhibitors for Cancer Therapeutics. Curr Oncol Rep. 2019;21(2):12. doi:.https://doi.org/10.1007/s11912-019-0763-9

92 Ahmed K , Koval A , Xu J , Bodmer A , Katanaev VL . Towards the first targeted therapy for triple-negative breast cancer: Repositioning of clofazimine as a chemotherapy-compatible selective Wnt pathway inhibitor. Cancer Lett. 2019;449:45–55. doi:.https://doi.org/10.1016/j.canlet.2019.02.018

93 Koval AV , Vlasov P , Shichkova P , Khunderyakova S , Markov Y , Panchenko J , et al. Anti-leprosy drug clofazimine inhibits growth of triple-negative breast cancer cells via inhibition of canonical Wnt signaling. Biochem Pharmacol. 2014;87(4):571–8. doi:.https://doi.org/10.1016/j.bcp.2013.12.007

94 Koval A , Pieme CA , Queiroz EF , Ragusa S , Ahmed K , Blagodatski A , et al. Tannins from Syzygium guineense suppress Wnt signaling and proliferation of Wnt-dependent tumors through a direct effect on secreted Wnts. Cancer Lett. 2018;435:110–20. doi:.https://doi.org/10.1016/j.canlet.2018.08.003

95 Blagodatski A , Cherepanov V , Koval A , Kharlamenko VI , Khotimchenko YS , Katanaev VL . High-throughput targeted screening in triple-negative breast cancer cells identifies Wnt-inhibiting activities in Pacific brittle stars. Sci Rep. 2017;7(1):11964. doi:.https://doi.org/10.1038/s41598-017-12232-7

96 Sheridan C . Wnt is back in drugmakers’ sights, but is it druggable? Nat Biotechnol. 2018;36(11):1028–9. doi:.https://doi.org/10.1038/nbt1118-1028

Notes

*

Contributed equally to the manuscript

Disclosure statement

No financial support and no other potential conflict of interest relevant to this article was reported.