Next Article in Journal
The Effects of Breastfeeding on Retinoblastoma Development: Results from an International Multicenter Retinoblastoma Survey
Next Article in Special Issue
Hedgehog Signaling in Myeloid Malignancies
Previous Article in Journal
Molecular Characterization of Prostate Cancers in the Precision Medicine Era
Previous Article in Special Issue
Hedgehog/GLI Signaling Pathway: Transduction, Regulation, and Implications for Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of the Hedgehog Pathway in Cholangiocarcinoma

1
Core Research Laboratory, Institute for Cancer Research and Prevention (ISPRO), 50139 Florence, Italy
2
Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy
*
Authors to whom correspondence should be addressed.
Equal contribution.
Cancers 2021, 13(19), 4774; https://doi.org/10.3390/cancers13194774
Submission received: 25 August 2021 / Revised: 17 September 2021 / Accepted: 21 September 2021 / Published: 24 September 2021
(This article belongs to the Special Issue The Role of Hedgehog Pathway in Cancer)

Abstract

:

Simple Summary

Cholangiocarcinoma (CCA) is one of the most refractory malignancies with a high mortality rate. Among all the pathways involved in CCA development, emerging evidence highlights Hedgehog (HH) signaling as a substantial player in CCA-genesis and development. The pro-tumoral function of HH provides potential therapeutic implications, and recently the use of HH inhibitors has paved the way for clinical application in various solid tumors. Targeting HH members, namely Hedgehog ligands, SMO transmembrane protein and GLI transcription factors may thus confer therapeutic options for the improvement of CCA treatment outcome.

Abstract

Cholangiocarcinoma (CCA) is a poorly treatable type of cancer and, along with hepatocellular carcinoma (HCC), is the predominant type of primitive liver cancer in adults. The lack of understanding of CCA biology has slowed down the identification of novel targets and the development of effective treatments. While tumors share some general characteristics, detailed knowledge of specific features is essential for the development of effectively tailored therapeutic approaches. The Hedgehog (HH) signaling cascade regulates stemness biology, embryonal development, tissue homeostasis, and cell proliferation and differentiation. Its aberrant activation has been associated with a variety of solid and hematological human malignancies. Several HH-inhibiting compounds have been indeed developed as potential anticancer agents in different types of tumors, with Smoothened and GLI inhibitors showing the most promising results. Beside its well-established function in other tumors, findings regarding the HH signaling in CCA are still controversial. Here we will give an overview of the most important clinical and molecular features of cholangiocarcinoma, and we will discuss the available evidence of the crosstalk between the HH signaling pathway and the cholangiocarcinoma cell biology.

1. Introduction

Primary liver cancer (PLC) is one of the most common cancers worldwide and is the second leading cause of cancer-related mortality [1,2]. Primary liver tumors are grossly classified in hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). HCC accounts for approximately 90% of all PLC, while CCA, a rare tumor but with an increasing global incidence, is the second most common form and accounts for about 5% of all PLC [1,2]. CCA is a highly heterogeneous disease arising from neoplastic transformation of intra- and extra-hepatic biliary epithelial cells (cholangiocytes), and it is characterized by a very poor prognosis [3,4].
The high mortality rate of CCA may depend on its nonspecific silent clinical features, which make it difficult to diagnose [5]. Currently, diagnosis is based on a combination of modalities, but, so far, no specific markers have been identified [1,2].
Another aspect contributing to the very poor survival rate of this tumor is its unresponsiveness to conventional therapies [3,4]. Currently, the standard-of-care treatment for CCA is limited to surgical resection, with 5-year survival of 20 to 40% [1,2]. Unfortunately, CCAs are generally asymptomatic in early stages and are usually diagnosed at an advanced unresectable stage and, although chemotherapy improves the quality of life of these patients, it remains only a palliative treatment [1,2]. Most patients with unresectable CCA undergo a rapid decline in clinical condition and die within 12 months of the onset of symptoms. To improve the outlook for individuals with CCA, both clinical and bench science are therefore imperative.
The goal of this review is to highlight the importance of the Hedgehog signaling pathway in the cholangio-carcinogenesis as a novel druggable candidate for this disease.

2. Cholangiocarcinoma Overview

Cholangiocarcinoma (CCA), a heterogeneous group of malignancies, occurs at any location along the biliary tree. It is anatomically classified as intrahepatic (iCCA), perihilar (pCCA), and distal extrahepatic (dCCA) [1,2,6,7]. On the other hand, iCCA can also be morphologically subcategorized into mass-forming (MF-iCCA), periductular-infiltrating (PI-iCCA), or intraductular growing (IG-iCCA). The MF-iCCA represents the most frequent form, whereas IG-iCCA is the least common form but with a more favorable prognosis [1].
During the past two decades, the incidence of iCCA as well as its mortality rate has been increasing worldwide, leading to the development of a growing scientific interest for this dismal malignancy. By contrast, the rate of eCCA is stable or even decreasing [1].
iCCA and eCCA, besides showing opposite epidemiological behaviors, are associated with different risk factors and histological features; diverse clinical outcomes; and dissimilar background in terms of expression profiling, pattern of genetic mutations, and epigenetic changes [1,6,7,8,9].
Some of the most prevalent CCA genetic alterations affect well-known cancer drivers such as tumor suppressor TP53, which regulates cell cycle, and KRAS, a tyrosine kinase signaling member [1]. About 20% of CCA showed TP53 mutation, whereas 7–54% of both iCCA and eCCA retained activating KRAS mutations [1,10]. Mutations in both genes were described as early events for CCA onset [1]. Moreover, 22% of iCCA presented proto-oncogene BRAF mutations, while inactivation of SMAD4 were described in 35% of iCCAs and 50% of eCCAs. Indeed, it has been hypothesized that there is a tumor suppressive role of SMAD4 by cell cycle regulation and the TGF-β pathway [1]. Taken together, these data indicate that genomic instability, TGF-β signaling, and RAS/RAF pathways may be driver for CCA onset.
Recent studies of next generation sequencing (NGS) have highlighted a highly mutated group of genes, including MLL3, RNF43, PEG3, ROBO2, BAP1, ARID1A, PBRM1, IDH1, and IDH2 [1,11], which are involved in histone modifier deactivation, G-proteins activation, and gain of genomic instability [11]. Notably, IDH1 and IDH2 mutations occurred together with an increased TP53 level [7]. Conversely, most of the tumors with mutations in the H3K4-specific methyltransferase MLL3 do not harbor TP53, KRAS, or SMAD4 mutations [1]. Interestingly, PEG3 and RNF43 are ubiquitin ligases and regulators of TP53 with the role of suppressing p53-mediated apoptosis. Moreover, RNF43 is even involved in the Wnt signaling pathway, targeting frizzled receptors for degradation. In Ov-related iCCAs, for example, it has been demonstrated as an upregulation of Wnt3a, Wnt5a, and Wnt7b [1]. ARID1A, PBRM1, and BAP1 together constitute the SWI/SNF complex, which is involved in nucleosome remodeling, mediating ATP-dependent chromatin remodeling processes. These tumor suppressor genes are frequently mutated and consequently silenced in CCAs [1].
Several observations underline how genetic variations may change according to etiology and CCA anatomical locations. BAP1 and IDH are more frequently mutated in non-liver–fluke-related CCAs, whereas TP53 mutations occur with increased frequency in liver–fluke-related tumors [10]. Moreover, although IDH1/2 and BRAF mutations are significantly predominant in the intrahepatic form of CCA, KRAS and TP53 mutations are relatively common in all CCA subtypes [1,6].
Others key mediators of cholangiocarcinogenesis are COX-2, MET, interleukin IL-6, and iNOS, as well as Notch, Hedgehog, and ErbB receptor kinase family members. In CCA, the soluble ligand EGF operates through the EGF receptor (EGFR), the founding member of the ErbB family, and increase CCA growth [1]. EGFR is deregulated in 32% of CCA patients, including both iCCA (11–27%) and eCCA (5–19%) [1,11]. Mutations in the tyrosine kinase domain of EGFR correlate with increased phosphorylation of AKT or p42/44 MAPK and p38 MAPK [1,12]. Human epidermal growth factor receptor 2 (ERBB2) is another member of the EGFR family responsible for CCA progression [11].
MAPK cascade can be activated even by MET, the receptor of hepatocyte growth factor (HGF), or IL-6, which represents a key cytokine in the CCA pathogenesis [11]. The binding of HGF to MET activates several downstream signaling pathways (RAS/MAPK, PI3K/AKT, and JAK/STAT) that drive tumor aggressiveness. MET over-expression has been described in both iCCA (20–60%) and eCCA (0–70%) [1]. In addition to the activation of MAPK cascade, IL-6 mediates the overexpression of EGFR as well as the STAT3-dependent upregulation of antiapoptotic protein MCL-1 [1].
In addition to intra-tumoral heterogeneity due to genetic alterations, heritable epigenetic alterations also generate phenotypical variability within tumors. Indeed, several epigenetic alterations, such as promoter hypermethylation and microRNA (miRNA) dysregulation, have also been linked to CCA development. Specifically, IDH mutations are associated with DNA hypermethylation of CpG shores, which suggests a global transcriptional deregulation. Importantly, the resulting deregulation of HNF4α blocks hepatocytic differentiation and promotes bile duct cancer development [1,13]. Moreover, in CCA, hypermethylated promoters silence tumor suppressor genes such as CDKN2A or p16INK4A, SOCS3, RASSF1A, and APC [11]. In parallel miRNAs also play an important role in CCA onset. For example, over-expressed miR21 has tumorigenic effects, by inhibiting programmed cell death 4 and tissue inhibitor of matrix metalloproteinase 3. Moreover, miR21 is able to regulate PI3K signaling, affecting CCA chemosensitivity [7,11].
Few studies have assessed the roles of chromosome aberrations such as copy number variations and gene fusions in the pathogenesis of human CCA. Notably, a study of copy number variation iCCA (n = 149) samples showed a range of chromosomal alterations, comprising gains at 1q and 7p and losses at 3p, 4q, 6q, 9pq, 13q, 14q, 8p, 17p, and 21q [7].
The advent of NGS approaches has further enriched the genomic landscape of CCA, reinforcing our understanding regarding its pathogenetic processes. Recent studies show involvement of fibroblast growth factor receptor 2 (FGFR2) and ROS proto-oncogene 1 (ROS1) fusions, together with an FGFR2–periphilin 1 (PPHLN1) fusion product. FGFR2 (chromosome 10q26) is one of the members of the fibroblast growth factor receptor family (FGFR1-4). FGFR2 alterations have primarily been detected in 6–50% of iCCA and 0–5% of eCCA, thus representing a putative critical diagnostic marker, as these rearrangements are identified almost exclusively in iCCA. FGFR2 fusions are the product of the combination of FGFR2 (exons 1–19) with disparate partners (i.e., BICC1, AHCYL1, MGEA5, TACC3, KCTD1, and TXLNA) [1]. The resulting fusion protein is activated through enforced dimerization, followed by the phosphorylation of the intracellular tyrosine kinase domain and the activation of downstream signaling pathways, including JAK/STAT pathways, PIK3/AKT/mTOR, and MAPKs [1]. Remarkably, a significant correlation has been observed between the presence of FGFR2–PPHLN1 and FGFR2–BICC1 fusion genes and the occurrence of mutations in KRAS genes, suggesting a possible cooperative role in coordinating iCCA pathogenesis [6]. FGFR2 fusions might thus represent the most prevailing targetable alteration in CCA; however, the sensitivity of the different FGFR2 fusion proteins to specific FGFR2 inhibitors remains undefined and should be broadly investigated in future studies.
Furthermore, 8.7% of CCA patients retained the FIG-ROS fusion gene that activates downstream signaling molecules, including STAT3 and AKT [1], thus accelerating tumorigenesis in a mouse model of iCCA holding KRAS and TP53 mutations [6].
Notably integrative genomic analysis identified two major iCCA types: inflammatory (STAT3 activation; cytokeratins overexpression) and proliferative (RAS, MAPK, and c-MET activation; KRAS and BRAF mutations) [1,9].
All these studies highlight the CCA heterogeneity which mirrors the underlying variability in molecular profile, drug-response, and outcome.

3. Current Therapeutic Intervention in Cholangiocarcinoma

3.1. Surgery

To date, radical surgical resection is the preferred therapeutic option for CCA, offering a curative chance. However, because of the challenging anatomical location and of the clinically “silencing” of this malignancy until later stages, only approximately 35% of patients with CCA undergo surgery [14]. Moreover, bilateral, multifocal disease, metastases, high post-surgical recurrence rates (generally 1–2 years) as well as comorbidities related to surgical risk, effectively decrease the real expected surgical benefits [15]. Frequent post-surgical relapse led to an implementation of several studies of adjuvant therapy. The BilCap study showed increased overall survival (OS) after surgical resection followed by Capecitabine treatment (51 months in the treated arm vs. 36 months in the observation arm) in iCCA. On the other hand, the resection of biliary tract cancer followed by adjuvant chemotherapy with gemcitabine and oxaliplatin (GEMOX), three months later, did not increase recurrence-free survival of CCA patients (PRODIGE12 study) [16]. Recently, the liver transplantation option has been considered in some specific disease conditions. For example, liver transplantation in unresectable pCCA preceded by neoadjuvant chemoradiotherapy showed admirable long-term disease-free survival rates [17,18]. Recently it has been demonstrated that very early iCCA patients after liver transplantation present good oncologic and survival outcome, although more confirmations are needed, such as the clarification of the systemic neoadjuvant regimens to be associated [19,20].

3.2. Systemic Chemotherapy

Patients with advanced or unresectable CCA require systemic chemotherapies as curative opportunity, although they have limited effectiveness and they are mostly palliative. The current standard of care is gemcitabine coupled with cisplatin [21]. The ABC-02 Phase III trial, which randomized 410 patients to gemcitabine alone or cisplatin/gemcitabine, demonstrated that cisplatin/gemcitabine, compared with gemcitabine monotherapy, prolongs OS by 3.6 months (11.7 months vs. 8.1 months, respectively) [22]. First-line treatment agents are associated with several side effects such as nausea, vomiting, and anorexia, and often fall into the development of chemoresistance. Recently, large randomization trials elucidated the possible benefits of second-line treatments. The phase III ABC-06 clinical trial randomly assigned 162 patients with advanced biliary tract cancer (72% CCA) progressing on first-line cisplatin-gemcitabine to either active symptom control (81 patients) or active symptom control with FOLFOX (folinic acid, 5-FU, and oxaliplatin) second-line chemotherapy (81 patients) [23]. Results of the trial showed a modest 0.9-month OS benefit in patients treated with FOLFOX, and clinically meaningful differences in survival at 6 and 12 months, suggesting that FOLFOX can be considered a good candidate in the second-line setting. Recently, results from a phase II clinical trial showed that treatment with nab-paclitaxel plus gemcitabine-cisplatin provided advantages in terms of median progression-free survival and overall survival as compared to controls treated with gemcitabine-cisplatin alone [24]. The gemcitabine-cisplatin-nab paclitaxel is now being compared in phase III setting to gem/cis as a new first-line standard (SWOG clinical trial number S1815).

3.3. Other Specific in Loco Therapies

Locoregional therapies based on the focalized delivery of chemotherapy and radiotherapy are also considered valid options for patients with localized unresectable CCA [25,26]. Increased survival and improved local control in locally advanced-metastatic iCCAs have been achieved with the following techniques: transarterial chemoembolization, transarterial radioembolization, hepatic arterial-based therapies, radiofrequency ablation, and photodynamic therapy. However, recurrence rates are still elevated [27].

3.4. Molecular Targeted Therapy

The high genetic variability of CCA represents a major challenge for effective pharmacological treatment; indeed, currently it remains one of the most dismal tumors with very limited therapeutic options. In recent years, deeper genomic studies and the advent of the next generation sequencing showed that nearly 45% of patients present potentially targetable genetic alterations, opening the possibility for new therapeutic opportunities [28,29]. The recent results of the MOSCATO-01 trial, which represent the first large-scale evaluation of precision medicine in hard-to-treat cancers, further support this evidence. In the trial, 68% of the comprised 43 biliary tract cancer (BTC) cases (29 iCCA, 10 eCCA, and 4 gallbladder cancer) showed actionable alterations, making this tumor one of the most potentially targetable malignancies [30,31]. The proper targeted therapies increased progression-free survival (PFS) in BTC patients as compared to those patients that could not be included in such targeted therapy trials. Thus, precision medicine seems to really have a good potential to improve BTC therapy. Indeed, the high requirement of introducing new therapeutic strategies in cholangiocarcinoma is completely revolutionizing the standard treatment algorithms for this disease, and many specific therapies are entering clinical practice in cholangiocarcinoma. The most significant targeted therapies associated with cholangiocarcinoma actionable molecular alterations currently in clinical trial development are briefly described below.

3.5. FGFR Inhibitors

Approximately 15–28% of patients with iCCA have tumors with FGFR2 rearrangements [28,29,32]. Pemigatinib (Pemazyre), a reversible adenosine triphosphate–competitive FGFR kinase inhibitor, was FDA-approved in 2020 as a targeted therapy for cholangiocarcinoma based on the results of the FIGHT-202 trial (NCT02924376) [33]. In this trial, 107 patients with CCA harboring FGFR2 rearrangements who were previously treated with chemotherapy were treated with pemigatinib once daily. An impressive 36% objective response rate and a median PFS of 6.9 months was observed. Futibatinib, an irreversible FGFR inhibitor also showed impressive activity in FGFR2-rearranged iCCA (NCT02052778) [34]. Importantly, futibatinib has been shown to overcome acquired resistance to reversible FGFR inhibitors, thus opening the potential to offer prolonged benefit by sequential FGFR inhibitor therapy [35]. Very recently, FDA granted the accelerated approval to Infigratinib for adults with previously treated, unresectable locally advanced or metastatic CCA with a fibroblast growth factor receptor 2 (FGFR2) fusion or other rearrangement [36]. Some studies with FGFR inhibitors (i.e., the FIGHT-302 study (NCT03656536) and the PROOF study (NCT03773302) are currently being considered in first-line setting [1,37].

3.6. IDH1/2 Inhibitors

Pathogenic mutations in isocitrate dehydrogense IDH1 and 2, a metabolic enzyme involved in the conversion of isocitrate to α-ketoglutarate, are among the most recurrent mutations, occurring in approximately 25% of iCCA patients [28,38,39]. Many IDH-selective inhibitors have been developed for cholangiocarcinoma patients in the last year, with AG-120 (ivosidenib, Agios) being the most evolved. This is an oral IDH1 inhibitor, FDA approved for the treatment of IDH1-mutated acute myeloid leukemia and very recently for advanced or metastatic CCA. In the phase III trial ClarIDHy (NCT02989857), 185 patients (91% IHCC) with IDH1-mutated cholangiocarcinoma who had received 1 or 2 lines of prior therapy were treated with either ivosidenib or placebo. Results showed a similar objective response rate (2.4% vs. 0%, with ivosidenib and placebo, respectively) but a modest benefit in PFS (2.7 months compared with 1.4 months for patients treated with placebo) and limited adverse toxicities [40,41]. Recently emerging combination strategies with IDH1 inhibitors are being tested in clinical trials with the intent to improve benefits, limiting toxicities, and possibly overcoming acquired resistance mechanisms. Based on recent preclinical evidence that IDH1-mutated malignancies present alteration in homologous recombination pathway and an increase in PARP inhibitor sensitivity, some preclinical studies and clinical trials investigations are undergoing in IDH mutated-CCA testing the drug combination between IDH and PARP inhibitors [42,43,44].
Moreover, a combination of IDH inhibitors with systemic chemotherapy or immunotherapy is another evaluable therapeutic opportunity [39].

3.7. BRAF-Directed Therapy

BRAF V600E mutations are present in approximately 3% of iCCA [28,29]. A single-arm, multicenter phase 2 ROAR trial (NCT02034110) elucidated the activity of the BRAF/MEK inhibitor combination in the context of CCA by evaluating the BRAF inhibitor dabrafenib (Tafinlar) combined with the MEK inhibitor trametinib (Mekinist) [45]. Results showed that the combination had a 47% objective response in patients with BRAF V600E–mutated cholangiocarcinoma. Patients with CCA enrolled in this trial had a median PFS of 9.2 months and median OS of 11.7 months [45].

3.8. Other Molecular Targets

Several other targetable molecular abnormalities occur in CCA. Notably, many reports highlighted the presence of cell cycle dysregulations and abnormalities, DNA damage response (DDR) pathway deficiency, and genomic instability [46,47]. Mutations in the most studied DDR genes BRCA1/2 fluctuates from 1% to 7% in patients with CCA; thus, after their success in pancreatic cancer, a plethora of studies aimed at investigating the response of CCA patients to PARP inhibitors, both in monotherapy and in combination with chemo and immunotherapy, are ongoing [43,48]. Other targetable DDR and cell cycle regulators of potential interest for CCA are being evaluated prevalently in preclinical setting (i.e., CDK4/6, Wee1, and ATR) [49,50,51]. Nonetheless, mutations in the epigenetic factors BAP1 and ARID1A are also emerging in CCA as potential actionable targets in synthetic lethality with PARP inhibitors and agents inducing DSB [28,52,53]. Other translocations such as ALK, ROS1, and NTRK and HER2 amplifications and mutations are present at low frequencies in CCA [28,54,55]. In summary, a plethora of targetable molecular pathways involved in CCA biology may contribute to ameliorate CCA therapeutic options. The Hedgehog pathway is among these pathways, but its role and mechanisms of action in this disease are still not fully understood [56,57].

4. The Hedgehog Signaling Pathway

The Hedgehog (HH) signaling cascade was first discovered as a key morphogenetic pathway in driving the embryonal development of fruit fly larva [58]. Its function is highly conserved in mammals, where it crucially participates in the regulation of essential cellular processes, including cell proliferation and differentiation, stemness, metabolism, tissue regeneration, and homeostasis [59,60]. Being fundamental in controlling such processes, the dysregulation of the HH cascade has been associated with diverse developmental disorders and cancer diseases [61].
A Hedgehog ligand is required to initiate the intracellular signaling, which culminates with the activation of the final effectors of the entire cascade, which are the GLI transcription factors. In mammals, three different Hedgehog ligands stimulate Hh-responsive cells. All three operate as morphogen proteins in regulating different aspects of the embryonal development, and they share comparable physiological effects, but with a different expression pattern and differential cellular responses, depending on ligand concentration [61,62,63]. Sonic Hedgehog (Shh) is the predominantly expressed Hh ligand in mammals [64,65], with Indian Hedgehog (Ihh) being partially redundant with Shh [61,66,67,68] and Desert Hedgehog (Dhh) mostly limited to the gonads, where its signal is important for Sertoli cells and granulosa cells [69,70,71].
In the absence of any Hh ligand, the 12-pass transmembrane receptor Patched 1 (PTCH1) occupies the base of the primary cilium, a specialized organelle required for the Hedgehog machinery, hindering the activity of the main HH transducer Smoothened (SMO) [72,73]. PKA, CK1, and GSK3β kinases phosphorylate GLI2 and GLI3, which are subsequently recognized by the F-box protein β-transducing-repeat-containing protein (β-TrCP) and sequestered outside the nucleus through a physical interaction with Suppressor of Fused (SuFu) [74,75,76]. Within the cytoplasm, GLI2 and GLI3 undergo proteasomal partial cleavage, reverting to their C-terminally truncated repressor forms (GLIR) and suppressing the transcription of HH target genes [77]. Conversely, GLI1 is predominantly regulated at the transcriptional level and through the ubiquitin-proteasome system (UPS)-dependent recognition of two degradation signals, which are degron DC and degron DN, located at the GLI1 C-terminus and the N-terminus, respectively [78].
Once secreted, the Hh ligand specifically binds the 12-pass transmembrane receptor PTCH1, which is therefore rapidly internalized and degraded [72,73]. The 7-pass transmembrane G-coupled receptor Smoothened is now released from PTCH1 inhibitory activity and translocated into the tip of the primary cilium, being therefore phosphorylated and activated by CK1α and GPCR kinase 2. SMO initiates an intracellular cascade, which prevents GLI suppressive modifications, promoting the migration of full-length GLI1, GLI2, and GLI3 into the nucleus as active forms (GLIA), in order to stimulate the transcription of HH target genes [79,80,81]. Among them, there are several factors implicated in controlling cell proliferation, survival, invasiveness, and stemness, together with GLI1 and PTCH1, thus creating a feedback loop, which further modulates the entire cascade itself [82,83] (Figure 1).
Beside the canonical process that turns on the Hedgehog signaling, different mechanisms are responsible for an aberrant activation of the cascade in cancer cells. Genetic alterations affecting HH fundamental members, i.e., loss of function (LOF) mutations in PTCH1, gain of function (GOF) mutations in SMO, and amplifications of GLI transcription factors, as well as dysregulated ligand-dependent stimulations (autocrine, paracrine, reverse paracrine), can be responsible for a non-canonical induction of the cascade [83,84,85,86,87]. In addition, an intricate network of intracellular interactions with other oncogenic pathways enables to bypass the entire HH upstream regulation by directly modulating the function of GLI transcription factors, both at a transcriptional and a post-translational level. Hence, a cooperative integration with RAS/RAF/MEK/ERK, PI3K/AKT, TGF-β cascades, among others, promotes the uncontrolled pro-tumoral activity of the GLI transcription factors in different types of human malignancies, including those of skin and lung, and also brain, gastrointestinal, and haematological tumors [59,88,89,90,91,92].

4.1. The Hedgehog Signaling in the Liver

The HH signaling remains mainly quiescent during adult life, where its activity is required for tissue regeneration. Therefore, as mentioned, an aberrant reactivation of the pathway has been found to be responsible for cancer initiation and progression in a variety of both solid and hematological human malignancies [53,93,94,95,96].
Several studies suggest that the Hedgehog cascade plays major roles in regulating both embryonal development and adult repair of the liver [97,98,99,100,101]. Namely, in the adult hepatic physiology, the HH signaling is nearly dormant, due to poor production and secretion of Hedgehog ligands by liver cells [102,103,104] and, on the other hand, to strong expression of Hedgehog inhibitors (i.e., Hhip) by liver sinusoidal cells, such as endothelial cells and quiescent hepatic stellate cells (HSCs) [105,106,107,108]. Furthermore, the HH signaling activity is gradually suppressed throughout the maturation and differentiation of liver epithelial cells [99].
Nonetheless, the HH signaling can be strongly reactivated during liver repair response upon the exposure to different types of injuries [98,100]. Hence, failing in proper regulation of the balance between latent/active HH cascade has been broadly associated with chronic liver disease and liver transformation [100,101,109,110].
Concerning liver tumors, an aberrant upregulation of the HH pathway has been demonstrated to be involved in both initiation and progression of HCC, hepatoblastoma, gallbladder cancer, and CCA [57,111,112,113,114,115,116,117,118].

4.2. Hedgehog and Cholangiocarcinoma

Being cholangiocarcinoma the second most frequent type of liver cancer, with very limited available therapeutic strategies and poor outcomes, the necessity to delve into understanding the molecular landscape of this biliary malignancy becomes explicit. Even though the function and the mechanisms of action of the HH pathway in CCA biology are still poorly explored and not completely understood, available data suggest a prominent role for HH in supporting cholangiocarcinoma malignant properties.

4.2.1. Hedgehog in CCA Patients

As mentioned before, one of the mechanisms responsible for driving an aberrant HH signaling in tumor cells concerns genetic alterations involving HH pathway key components. Both mutation and copy number alterations (CNA) affecting Hedgehog pathway main members, namely GLI1, GLI2, GLI3, SMO, PTCH1/2, SUFU, and DHH, have been observed in mixed cholangiocarcinoma patients with different frequencies (Table 1) [119,120]. GLI3 is one of the most frequently mutated genes (4.6%), together with Desert Hedgehog (5.6%), whose function in human cancer is still poorly investigated. Nonetheless, DHH expression has been observed in gastric cancer [121], correlates with advanced tumor grades in breast cancer [122], and has been supposed as a prognostic indicator for clear cell renal carcinoma [123] and for a specific subgroup of pediatric AML [124]. By the way, the biological significance of those HH alterations identified in CCA patients is still not characterized. Amplifications detected in GLI1 and DHH may assume gain of function effects, whereas, for missense mutations, we can only hypothesize inactivating alterations for the negative regulators PTCH1, PTCH2, and SUFU on one hand, and activating alterations for GLI transcription factors, SMO and Hedgehog ligands, on the other, as occurs in other types of cancer [85,125,126,127,128,129,130,131,132].
The expression of Hedgehog signaling members has been analyzed in a cohort of 50 human CCA tissues, where GLI1 and PTCH1 have been found overexpressed in approximately 50% and 30% of cases, respectively, together with SHH, detecting a significant activation of Hedgehog in almost 50% of cases [56]. Furthermore, a significant correlation of HH pathway activation with tumor progression and prognosis has been documented in CCA. GLI1 overexpression has been associated with malignant lymph node status, and SHH expression is enriched in undifferentiated tumors (grade 2 and 3), compared to highly differentiated tumors (grade 1) [133,134,135]. In a group of 200 patients with intrahepatic cholangiocarcinoma, SHH, IHH, PTCH1, GLI1, GLI2, and SMO have been observed overexpressed in more than 65% of clinical iCCA specimen, compared to non-malignant biliary epithelium, with SHH, SMO, and GLI2 being the most frequently upregulated HH members (>80%). Interestingly, in iCCA tissues, high GLI1 and/or GLI2 expression are significantly associated with intrahepatic metastasis and patient adverse disease-free-survival and overall survival [113,136], which has also been correlated with IHH and PTCH1 expression [112]. The Hedgehog cascade has been related also to CDH3 expression, which is associated with CCA progression to more aggressive stages and thereupon is correlated to limited overall survival [137]. Altogether, the reported studies provide a role for the Hedgehog-GLI signaling pathway, particularly for GLI1 and GLI2, as reliable prognostic factor for cholangiocarcinoma.

4.2.2. Hedgehog Aberrant Activation in CCA Cells

Early evidence of HH pathway aberrant activation in BTC came from an analysis of mRNA expression in a panel of gastrointestinal cancer cell lines, including cholangiocarcinoma. Herein, SHH, IHH, GLI, and PTCH mRNA expression was widely detected [138]. GLI1, as in other tumors, was found to promote iCCA survival, growth, and EMT reprogramming [113,139], together with GLI3, which directly binds to the promoter of death receptor 4 (DR4), repressing its transcription and thus preventing TRAIL-dependent cholangiocarcinoma cell death [140]. Furthermore, the Hedgehog signaling was shown to stimulate the intracellular production of miR-25, a microRNA that is functionally involved in the protection of CCA cells towards TRAIL-determined cell death [141].
In CCA cell lines, the stimulation of the pathway with recombinant SHH ligand resulted as being very effective in activating HH-dependent transcriptional activity and in promoting in vitro cholangiocarcinoma cell growth, which were both negatively affected by the administration of the 5E1 Hedgehog-neutralizing antibody and by the genetic modulation of GLI1 [138]. The responsiveness of CCA cells to ligand stimulation could suggest a requirement for the canonical activation of HH in CCA cells. Nevertheless, cholangiocarcinoma cells often lack the ability to express cilia [57,142], which are cellular structures required for the transduction of the canonical pathway, and other studies have indeed provided evidences concerning a cilium and/or GLI-independent modality of HH activation in non-ciliated CCA cell lines [57,139]. The precise role of the primary cilium is currently thought to be context-dependent [143,144,145] and in cancer we can find a multitude of diverse aspects that can drive the non-canonical route. Thus, additional studies appear to be required, with the aim of elucidating the network of non-canonical regulation of Hedgehog in CCA cells.
Beside sustaining growth and proliferative processes, the HH pathway acts as a key mediator in coordinating many other cancer properties in a variety of human cancers, including survival, stemness, migration, invasion, deregulated metabolism, genome instability, angiogenesis, and pro-tumoral inflammation [146]. Hypoxia, which is an important element in contributing to malignant transformation and cancer progression, as well as chemoresistance [147,148,149], induces the activity of HIF-1α, which positively regulates the HH signaling with malignant outcomes in different types of human cancers [150,151], including cholangiocarcinoma, where hypoxia-induced HIF-1α promotes stemness features and invasive behavior by modulating SHH, SMO, and GLI1 [121]. HIF-1α appears to be preferentially expressed in CCA tissues, rather than in the neighboring normal biliary epithelium [152], and being such a relevant element in sustaining tumor chemoresistance and immune evasion, many hypoxia-targeting strategies are now under investigation for cancer therapy [153]. An accurate comprehension of the relationship between HH and hypoxia in CCA would thus be beneficial for the designing of powerful combinatorial targeted strategies.
Cholangiocarcinoma is characterized by a strong desmoplastic reaction, prompted by both fibroblastic and immunological cellular subsets, with HSC-derived myofibroblasts being one of the most predominantly represented stromal components. The intricate network that develops between tumor and the stromal counterpart appears to be relevant in sustaining malignant properties in cancer cells [154]. As many signaling molecules are released by stromal cells and then recognized by cancer cells, the CCA desmoplastic environment could represent an important factor in stimulating Hedgehog signaling in tumor cells in a paracrine way. Indeed, the suppressive effect of the HH signaling on TRAIL-induced cell death has been demonstrated to be mediated by myofibroblasts-secreted PDFG-BB, which signals in a paracrine manner in cancer cells, where it activates the Hedgehog cascade [155,156]. In this context, in the absence of any HH ligand stromal PDGF-BB induces the trafficking of SMO in a PKA-dependent manner, promoting the translocation of GLI2 into the nucleus and, ultimately, its transcriptional activity [157]. Once activated, the HH pathway protects CCA cells from TRAIL-induced apoptosis via, at least in part, a PLK2-mediated mechanism [126,127,155,156]. That Hedgehog-conditioned interplay between cholangiocarcinoma cells and the stromal counterpart is crucial, not only in preventing cancer cell death, but also in stimulating in vitro cancer cell proliferation, migration, and invasion [112]. In addition, the presence of a functional stromal microenvironment enhances tumor growth in vivo, along with activating the angiogenetic process in an HH-dependent manner and with making CCA cells more sensitive to SMO-antagonizing compounds [112]. This could be due to the fact that, beside cancer cells, the stromal cellular portion also displays an activation of the HH signaling, with Shh stimulating HSC-derived CAFs viability in an autocrine fashion [108]. A reciprocal HH-dependent regulation could thus be feasible in cholangiocarcinoma, as occurs in other types of gastrointestinal cancers [158,159]. Beside CAFs, CCA microenvironment results enriched different immunological cell subsets [154]. Among them, M2 macrophages have been demonstrated to sustain CCA growth in vivo in a WNT-dependent manner. Interestingly, the depletion of macrophages results in reduced CCA growth upon the downregulation of many signaling proteins, including SMO and PTCH1 [160], suggesting that this HH-conditioned interplay with CCA cells may involve not only CAFs, but also immunological cells.

4.2.3. Hedgehog Targeting Strategies in CCA

Blocking the HH signaling by chemically targeting upstream or downstream HH mediators displayed promising effects in hindering CCA growth and progression. The currently available HH inhibiting compounds can be subdivided into 2 main groups: SMO and GLI inhibitors. The natural alkaloyd cyclopamine is the first discovered SMO inhibitor and has been largely used in CCA studies, providing early favorable data concerning the efficacy of targeting HH in this biliary tract neoplasia. Suppressing the activity of SMO with cyclopamine, alone or in combination with other appropriate targeted therapies, for instance MAPKs inhibitors [161], resulted as being strongly effective in repressing CCA growth and survival, but also in preventing cholangiocarcinoma invasion and metastasis [57,113,133,138,155]. Nevertheless, the emergence of limited oral solubility and serious side effects in mice restricts cyclopamine availability to pre-clinical studies and denotes the necessity to involve different HH-antagonizing compounds to select for putative clinical studies.
Vismodegib (GDC-0449) is a second generation cyclopamine derivative and the first SMO blocking agent to be FDA approved for clinical administration in advanced and metastatic BCC patients [162]. Vismodegib has also been found to be powerful as an anticancer agent in cholangiocarcinoma, reducing in vivo cholangiocarcinoma initiation, growth, and metastatic spreading [57], suggesting new potential strategies beyond cyclopamine to target the Hedgehog signaling in cholangiocarcinoma. Notwithstanding, the onset of mutations in the drug binding pocket of SMO that interrupt the responsiveness to vismodegib administration is frequent in BCC and MB patients [87,163,164]. Thus, despite the strong preclinical efficacy of targeting HH with cyclopamine derivatives in CCA, engaging novel SMO antagonizing structures that could offer more powerful mechanisms of target binding and biological action [165,166,167,168,169,170,171,172,173,174] should be properly considered for CCA studies.
Hedgehog-antagonizing agents have also been demonstrated to be beneficial in combination with conventional chemotherapy to target biliary tract tumors. Intriguingly, the administration of the SMO inhibitor BMS-833923 in mouse xenograft models of cholangiocarcinoma potentiates the effects of the chemotherapeutic agent gemcitabine in reducing in vivo tumor volume [56]. Similarly, the combination of cisplatin with the GLI inhibitor GANT61 displayed synergistic effects in hampering in vitro cholangiocarcinoma cell growth and survival [134], providing interesting novel insights concerning the opportunity to target the non-canonical cascade by acting directly on GLI transcription factors, even in BTC (Figure 2). This therapeutic approach is attractive because, beside the outbreak of mutations in SMO, resistance to SMO inhibitors can be also driven by downstream compensatory mechanisms, which are responsible for the activation of the GLI (reviewed in [87]).

5. Concluding Remarks and Future Perspectives

HH signaling is an evolutionarily conserved pathway, being essential for coordinating normal development and differentiation in vertebrates. However, aberrant HH pathway activation is widely known to be closely correlated with the development and malignant progression of several human cancers. The available preclinical experimental data indicate that dysregulation of this pathway is a determinant of cholangiocarcinoma progression and metastasis (Figure 2). Nevertheless, in CCA, many aspects concerning the specific molecular mechanisms that drive the pro-tumoral function of the Hedgehog signaling still remain poorly understood. Trying to delve into the specific function of each member of the HH cascade, as well as comprehending the biological implications of related genetic alterations detected in CCA patients, thus becomes conceivable, in order to orientate the selection of HH targeting agents for further preclinical studies and hypothetical future clinical enrollment. Furthermore, there are many elements related to the crosstalk of HH signaling with other CCA-driving oncogenic pathways that are thus far open to question. In addition, in cancer, more than one non-canonical mode of activation can often be present, and canonical and non-canonical activation can coexist, shaping an intricate and sophisticated molecular landscape. Defining and elucidating the weight of this particular aspect of non-canonical activation, together with establishing the molecular mechanisms of action of HH targeting agents in CCA, could therefore become challenging, with the perspective of combining Hedgehog inhibitors with other targeted strategies, which appears to represent a promising system from available CCA preclinical studies. Because cholangiocarcinoma management is still grounded predominantly on systemic approaches, building up novel strategies based on blocking tumor-specific deregulated mediators should be seen as imperative. Furthermore, preclinical studies indicate that HH pathway inhibitors improve the efficacy of chemotherapeutic regimen. Lastly, a special consideration should be addressed to the profound interconnection between CCA cells and the stromal microenvironment, which not merely supports and sustains tumor progression, but also functions in affecting and restricting therapy responsiveness. For that reason, different strategies to target cancer microenvironments are currently under preclinical and clinical considerations in different human neoplasias [175], even though using TME-targeting strategies alone cannot reasonably ensure a complete eradication of the tumor. In CCA, some of the TME-driven mechanisms that promote malignant progression function in a Hedgehog-dependent manner; hence, a combinatorial targeting of HH and stromal function may be contemplated. In conclusion, this review aims to clarify the current knowledge of Hedgehog signaling in cholangiocarcinoma and highlights the possibility of promoting the study of innovative anti-CCA strategies based on HH-inhibiting approaches.

Author Contributions

Conceptualization, F.M. and C.R.; writing—original draft preparation, G.A. and L.C.; writing—review and editing, B.S.; supervision, F.M. and C.R.; funding acquisition, F.M. and C.R. All authors have read and agreed to the published version of the manuscript.

Funding

Funding for this work was partially provided by Italian Foundation of Cancer Research award (IG23117) to Raggi and (IG17786) to Marra. Raggi is a member of the European Network for the Study of Cholangiocarcinoma (ENSCCA) and participates in the initiative COST Action EURO-CHOLANGIO-NET granted by the COST Association (CA18122).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable. No new data were created or analyzed in this work.

Acknowledgments

Figures were created with BioRender.com. Agreement numbers: IB22ZOHL2Y, VZ22ZOE5DW.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next horizon in mechanisms and management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef] [PubMed]
  2. Banales, J.M.; Cardinale, V.; Carpino, G.; Marzioni, M.; Andersen, J.B.; Invernizzi, P.; Lind, G.E.; Folseraas, T.; Forbes, S.J.; Fouassier, L.; et al. Expert consensus document: Cholangiocarcinoma: Current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 261–280. [Google Scholar] [CrossRef]
  3. Andersen, J.B.; Spee, B.; Blechacz, B.R.; Avital, I.; Komuta, M.; Barbour, A.; Conner, E.A.; Gillen, M.C.; Roskams, T.; Roberts, L.R.; et al. Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology 2012, 142, 1021–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Blechacz, B.; Gores, G.J. Cholangiocarcinoma: Advances in pathogenesis, diagnosis, and treatment. Hepatology 2008, 48, 308–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Blechacz, B.; Komuta, M.; Roskams, T.; Gores, G.J. Clinical diagnosis and staging of cholangiocarcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 512–522. [Google Scholar] [CrossRef] [Green Version]
  6. Moeini, A.; Sia, D.; Bardeesy, N.; Mazzaferro, V.; Llovet, J.M. Molecular Pathogenesis and Targeted Therapies for Intrahepatic Cholangiocarcinoma. Clin. Cancer Res. 2016, 22, 291–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Sia, D.; Tovar, V.; Moeini, A.; Llovet, J.M. Intrahepatic cholangiocarcinoma: Pathogenesis and rationale for molecular therapies. Oncogene 2013, 32, 4861–4870. [Google Scholar] [CrossRef] [Green Version]
  8. Cardinale, V.; Carpino, G.; Reid, L.; Gaudio, E.; Alvaro, D. Multiple cells of origin in cholangiocarcinoma underlie biological, epidemiological and clinical heterogeneity. World J. Gastrointest. Oncol. 2012, 4, 94–102. [Google Scholar] [CrossRef]
  9. Raggi, C.; Invernizzi, P.; Andersen, J.B. Impact of microenvironment and stem-like plasticity in cholangiocarcinoma: Molecular networks and biological concepts. J. Hepatol. 2015, 62, 198–207. [Google Scholar] [CrossRef] [Green Version]
  10. Rizvi, S.; Borad, M.J.; Patel, T.; Gores, G.J. Cholangiocarcinoma: Molecular pathways and therapeutic opportunities. Semin. Liver Dis. 2014, 34, 456–464. [Google Scholar] [CrossRef] [Green Version]
  11. Rizvi, S.; Gores, G.J. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 2013, 145, 1215–1229. [Google Scholar] [CrossRef] [Green Version]
  12. Leone, F.; Cavalloni, G.; Pignochino, Y.; Sarotto, I.; Ferraris, R.; Piacibello, W.; Venesio, T.; Capussotti, L.; Risio, M.; Aglietta, M. Somatic mutations of epidermal growth factor receptor in bile duct and gallbladder carcinoma. Clin. Cancer Res. 2006, 12, 1680–1685. [Google Scholar] [CrossRef] [Green Version]
  13. Saha, S.K.; Parachoniak, C.A.; Ghanta, K.S.; Fitamant, J.; Ross, K.N.; Najem, M.S.; Gurumurthy, S.; Akbay, E.A.; Sia, D.; Cornella, H.; et al. Corrigendum: Mutant IDH inhibits HNF-4α to block hepatocyte differentiation and promote biliary cancer. Nature 2015, 528, 152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rizvi, S.; Khan, S.A.; Hallemeier, C.L.; Kelley, R.K.; Gores, G.J. Cholangiocarcinoma—Evolving concepts and therapeutic strategies. Nat. Rev. Clin. Oncol. 2018, 15, 95–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cillo, U.; Fondevila, C.; Donadon, M.; Gringeri, E.; Mocchegiani, F.; Schlitt, H.J.; Ijzermans, J.N.M.; Vivarelli, M.; Zieniewicz, K.; Damink, S.O.; et al. Surgery for cholangiocarcinoma. Liver Int. 2019, 39 (Suppl. S1), 143–155. [Google Scholar] [CrossRef] [Green Version]
  16. Kelley, R.K.; Bridgewater, J.; Gores, G.J.; Zhu, A.X. Systemic therapies for intrahepatic cholangiocarcinoma. J. Hepatol. 2020, 72, 353–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Gringeri, E.; Gambato, M.; Sapisochin, G.; Ivanics, T.; Lynch, E.N.; Mescoli, C.; Burra, P.; Cillo, U.; Russo, F.P. Cholangiocarcinoma as an Indication for Liver Transplantation in the Era of Transplant Oncology. J. Clin. Med. 2020, 9, 1353. [Google Scholar] [CrossRef]
  18. Lehrke, H.D.; Heimbach, J.K.; Wu, T.T.; Jenkins, S.M.; Gores, G.J.; Rosen, C.B.; Mounajjed, T. Prognostic Significance of the Histologic Response of Perihilar Cholangiocarcinoma to Preoperative Neoadjuvant Chemoradiation in Liver Explants. Am. J. Surg. Pathol. 2016, 40, 510–518. [Google Scholar] [CrossRef]
  19. Sapisochin, G.; Facciuto, M.; Rubbia-Brandt, L.; Marti, J.; Mehta, N.; Yao, F.Y.; Vibert, E.; Cherqui, D.; Grant, D.R.; Hernandez-Alejandro, R.; et al. Liver transplantation for “very early” intrahepatic cholangiocarcinoma: International retrospective study supporting a prospective assessment. Hepatology 2016, 64, 1178–1188. [Google Scholar] [CrossRef]
  20. Sapisochin, G.; de Lope, C.R.; Gastaca, M.; de Urbina, J.O.; López-Andujar, R.; Palacios, F.; Ramos, E.; Fabregat, J.; Castroagudín, J.F.; Varo, E.; et al. Intrahepatic cholangiocarcinoma or mixed hepatocellular-cholangiocarcinoma in patients undergoing liver transplantation: A Spanish matched cohort multicenter study. Ann. Surg. 2014, 259, 944–952. [Google Scholar] [CrossRef]
  21. Valle, J.W.; Furuse, J.; Jitlal, M.; Beare, S.; Mizuno, N.; Wasan, H.; Bridgewater, J.; Okusaka, T. Cisplatin and gemcitabine for advanced biliary tract cancer: A meta-analysis of two randomised trials. Ann. Oncol. 2014, 25, 391–398. [Google Scholar] [CrossRef] [PubMed]
  22. Valle, J.; Wasan, H.; Palmer, D.H.; Cunningham, D.; Anthoney, A.; Maraveyas, A.; Madhusudan, S.; Iveson, T.; Hughes, S.; Pereira, S.P.; et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N. Engl. J. Med. 2010, 362, 1273–1281. [Google Scholar] [CrossRef] [Green Version]
  23. Lamarca, A.; Palmer, D.H.; Wasan, H.S.; Ross, P.J.; Ma, Y.T.; Arora, A.; Falk, S.; Gillmore, R.; Wadsley, J.; Patel, K.; et al. ABC-06|A randomised phase III, multi-centre, open-label study of active symptom control (ASC) alone or ASC with oxaliplatin / 5-FU chemotherapy (ASC+mFOLFOX) for patients (pts) with locally advanced / metastatic biliary tract cancers (ABC) previously-treated with cisplatin/gemcitabine (CisGem) chemotherapy. J. Clin. Oncol. 2019, 37, 4003. [Google Scholar] [CrossRef]
  24. Shroff, R.T.; Javle, M.M.; Xiao, L.; Kaseb, A.O.; Varadhachary, G.R.; Wolff, R.A.; Raghav, K.P.S.; Iwasaki, M.; Masci, P.; Ramanathan, R.K.; et al. Gemcitabine, Cisplatin, and nab-Paclitaxel for the Treatment of Advanced Biliary Tract Cancers: A Phase 2 Clinical Trial. JAMA Oncol. 2019, 5, 824–830. [Google Scholar] [CrossRef]
  25. Seidensticker, R.; Ricke, J.; Seidensticker, M. Integration of chemoembolization and radioembolization into multimodal treatment of cholangiocarcinoma. Best Pract. Res. Clin. Gastroenterol. 2015, 29, 319–332. [Google Scholar] [CrossRef] [PubMed]
  26. Cercek, A.; Boerner, T.; Tan, B.R.; Chou, J.F.; Gönen, M.; Boucher, T.M.; Hauser, H.F.; Do, R.K.G.; Lowery, M.A.; Harding, J.J.; et al. Assessment of Hepatic Arterial Infusion of Floxuridine in Combination with Systemic Gemcitabine and Oxaliplatin in Patients with Unresectable Intrahepatic Cholangiocarcinoma: A Phase 2 Clinical Trial. JAMA Oncol. 2020, 6, 60–67. [Google Scholar] [CrossRef] [PubMed]
  27. Blechacz, B. Cholangiocarcinoma: Current Knowledge and New Developments. Gut Liver 2017, 11, 13–26. [Google Scholar] [CrossRef] [PubMed]
  28. Lowery, M.A.; Ptashkin, R.; Jordan, E.; Berger, M.F.; Zehir, A.; Capanu, M.; Kemeny, N.E.; O’Reilly, E.M.; El-Dika, I.; Jarnagin, W.R.; et al. Comprehensive Molecular Profiling of Intrahepatic and Extrahepatic Cholangiocarcinomas: Potential Targets for Intervention. Clin. Cancer Res. 2018, 24, 4154–4161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Jusakul, A.; Cutcutache, I.; Yong, C.H.; Lim, J.Q.; Huang, M.N.; Padmanabhan, N.; Nellore, V.; Kongpetch, S.; Ng, A.W.T.; Ng, L.M.; et al. Whole-Genome and Epigenomic Landscapes of Etiologically Distinct Subtypes of Cholangiocarcinoma. Cancer Discov. 2017, 7, 1116–1135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Massard, C.; Michiels, S.; Ferté, C.; Le Deley, M.C.; Lacroix, L.; Hollebecque, A.; Verlingue, L.; Ileana, E.; Rosellini, S.; Ammari, S.; et al. High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers: Results of the MOSCATO 01 Trial. Cancer Discov. 2017, 7, 586–595. [Google Scholar] [CrossRef] [Green Version]
  31. Verlingue, L.; Malka, D.; Allorant, A.; Massard, C.; Ferté, C.; Lacroix, L.; Rouleau, E.; Auger, N.; Ngo, M.; Nicotra, C.; et al. Precision medicine for patients with advanced biliary tract cancers: An effective strategy within the prospective MOSCATO-01 trial. Eur. J. Cancer 2017, 87, 122–130. [Google Scholar] [CrossRef]
  32. Valle, J.W.; Lamarca, A.; Goyal, L.; Barriuso, J.; Zhu, A.X. New Horizons for Precision Medicine in Biliary Tract Cancers. Cancer Discov. 2017, 7, 943–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Abou-Alfa, G.K.; Sahai, V.; Hollebecque, A.; Vaccaro, G.; Melisi, D.; Al-Rajabi, R.; Paulson, A.S.; Borad, M.J.; Gallinson, D.; Murphy, A.G.; et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: A multicentre, open-label, phase 2 study. Lancet. Oncol. 2020, 21, 671–684. [Google Scholar] [CrossRef]
  34. Goyal, L.; Meric-Bernstam, F.; Hollebecque, A.; Valle, J.W.; Morizane, C.; Karasic, T.B.; Abrams, T.A.; Furuse, J.; He, Y.; Soni, N.; et al. FOENIX-CCA2: A phase II, open-label, multicenter study of futibatinib in patients (pts) with intrahepatic cholangiocarcinoma (iCCA) harboring FGFR2 gene fusions or other rearrangements. J. Clin. Oncol. 2020, 38, 108. [Google Scholar] [CrossRef]
  35. Goyal, L.; Shi, L.; Liu, L.Y.; Fece de la Cruz, F.; Lennerz, J.K.; Raghavan, S.; Leschiner, I.; Elagina, L.; Siravegna, G.; Ng, R.W.; et al. TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion-Positive Intrahepatic Cholangiocarcinoma. Cancer Discov. 2019, 9, 1064–1079. [Google Scholar] [CrossRef] [Green Version]
  36. Infigratinib Approved for Cholangiocarcinoma. Cancer Discov. 2021, 11, Of5. [CrossRef]
  37. Bekaii-Saab, T.S.; Valle, J.W.; Van Cutsem, E.; Rimassa, L.; Furuse, J.; Ioka, T.; Melisi, D.; Macarulla, T.; Bridgewater, J.; Wasan, H.; et al. FIGHT-302: First-line pemigatinib vs gemcitabine plus cisplatin for advanced cholangiocarcinoma with FGFR2 rearrangements. Future Oncol. 2020, 16, 2385–2399. [Google Scholar] [CrossRef]
  38. Molenaar, R.J.; Maciejewski, J.P.; Wilmink, J.W.; van Noorden, C.J.F. Wild-type and mutated IDH1/2 enzymes and therapy responses. Oncogene 2018, 37, 1949–1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Rizzo, A.; Ricci, A.D.; Brandi, G. IDH inhibitors in advanced cholangiocarcinoma: Another arrow in the quiver? Cancer Treat. Res. Commun. 2021, 27, 100356. [Google Scholar] [CrossRef]
  40. Abou-Alfa, G.K.; Macarulla, T.; Javle, M.M.; Kelley, R.K.; Lubner, S.J.; Adeva, J.; Cleary, J.M.; Catenacci, D.V.; Borad, M.J.; Bridgewater, J.; et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): A multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet. Oncol. 2020, 21, 796–807. [Google Scholar] [CrossRef]
  41. Abou-Alfa, G.K.; Macarulla Mercade, T.; Javle, M.; Kelley, R.K.; Lubner, S.; Adeva, J.; Cleary, J.M.; Catenacci, D.V.; Borad, M.J.; Bridgewater, J.A.; et al. LBA10_PR—ClarIDHy: A global, phase III, randomized, double-blind study of ivosidenib (IVO) vs placebo in patients with advanced cholangiocarcinoma (CC) with an isocitrate dehydrogenase 1 (IDH1) mutation. Ann. Oncol. 2019, 30, v872–v873. [Google Scholar] [CrossRef]
  42. Sulkowski, P.L.; Corso, C.D.; Robinson, N.D.; Scanlon, S.E.; Purshouse, K.R.; Bai, H.; Liu, Y.; Sundaram, R.K.; Hegan, D.C.; Fons, N.R.; et al. 2-Hydroxyglutarate produced by neomorphic IDH mutations suppresses homologous recombination and induces PARP inhibitor sensitivity. Sci. Transl. Med. 2017, 9, eaal2463. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ricci, A.D.; Rizzo, A.; Bonucci, C.; Tober, N.; Palloni, A.; Mollica, V.; Maggio, I.; Deserti, M.; Tavolari, S.; Brandi, G. PARP Inhibitors in Biliary Tract Cancer: A New Kid on the Block? Medicines 2020, 7, 54. [Google Scholar] [CrossRef] [PubMed]
  44. IDH-Mutant Tumors Vulnerable to PARP Inhibition. Cancer Discov. 2017, 7, Of4. [CrossRef] [Green Version]
  45. Subbiah, V.; Lassen, U.; Élez, E.; Italiano, A.; Curigliano, G.; Javle, M.; de Braud, F.; Prager, G.W.; Greil, R.; Stein, A.; et al. Dabrafenib plus trametinib in patients with BRAF(V600E)-mutated biliary tract cancer (ROAR): A phase 2, open-label, single-arm, multicentre basket trial. Lancet. Oncol. 2020, 21, 1234–1243. [Google Scholar] [CrossRef]
  46. Chae, H.; Kim, D.; Yoo, C.; Kim, K.P.; Jeong, J.H.; Chang, H.M.; Lee, S.S.; Park, D.H.; Song, T.J.; Hwang, S.; et al. Therapeutic relevance of targeted sequencing in management of patients with advanced biliary tract cancer: DNA damage repair gene mutations as a predictive biomarker. Eur. J. Cancer 2019, 120, 31–39. [Google Scholar] [CrossRef]
  47. Lamarca, A.; Barriuso, J.; McNamara, M.G.; Valle, J.W. Biliary Tract Cancer: State of the Art and potential role of DNA Damage Repair. Cancer Treat. Rev. 2018, 70, 168–177. [Google Scholar] [CrossRef] [PubMed]
  48. Ahn, D.H.; Bekaii-Saab, T. Biliary tract cancer and genomic alterations in homologous recombinant deficiency: Exploiting synthetic lethality with PARP inhibitors. Chin. Clin. Oncol. 2020, 9, 6. [Google Scholar] [CrossRef]
  49. Nam, A.R.; Jin, M.H.; Park, J.E.; Bang, J.H.; Oh, D.Y.; Bang, Y.J. Therapeutic Targeting of the DNA Damage Response Using an ATR Inhibitor in Biliary Tract Cancer. Cancer Res. Treat. 2019, 51, 1167–1179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Nam, A.R.; Jin, M.H.; Bang, J.H.; Oh, K.S.; Seo, H.R.; Oh, D.Y.; Bang, Y.J. Inhibition of ATR Increases the Sensitivity to WEE1 Inhibitor in Biliary Tract Cancer. Cancer Res. Treat. 2020, 52, 945–956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Song, X.; Liu, X.; Wang, H.; Wang, J.; Qiao, Y.; Cigliano, A.; Utpatel, K.; Ribback, S.; Pilo, M.G.; Serra, M.; et al. Combined CDK4/6 and Pan-mTOR Inhibition Is Synergistic Against Intrahepatic Cholangiocarcinoma. Clin. Cancer Res. 2019, 25, 403–413. [Google Scholar] [CrossRef] [Green Version]
  52. Jiao, Y.; Pawlik, T.M.; Anders, R.A.; Selaru, F.M.; Streppel, M.M.; Lucas, D.J.; Niknafs, N.; Guthrie, V.B.; Maitra, A.; Argani, P.; et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat. Genet. 2013, 45, 1470–1473. [Google Scholar] [CrossRef]
  53. Pak, E.; Segal, R.A. Hedgehog Signal Transduction: Key Players, Oncogenic Drivers, and Cancer Therapy. Dev. Cell 2016, 38, 333–344. [Google Scholar] [CrossRef] [Green Version]
  54. Kheder, E.S.; Hong, D.S. Emerging Targeted Therapy for Tumors with NTRK Fusion Proteins. Clin. Cancer Res. 2018, 24, 5807–5814. [Google Scholar] [CrossRef] [Green Version]
  55. Law, L.Y. Dramatic response to trastuzumab and paclitaxel in a patient with human epidermal growth factor receptor 2-positive metastatic cholangiocarcinoma. J. Clin. Oncol. 2012, 30, e271–e273. [Google Scholar] [CrossRef]
  56. Riedlinger, D.; Bahra, M.; Boas-Knoop, S.; Lippert, S.; Bradtmöller, M.; Guse, K.; Seehofer, D.; Bova, R.; Sauer, I.M.; Neuhaus, P.; et al. Hedgehog pathway as a potential treatment target in human cholangiocarcinoma. J. Hepato-Biliary-Pancreat. Sci. 2014, 21, 607–615. [Google Scholar] [CrossRef] [PubMed]
  57. Razumilava, N.; Gradilone, S.A.; Smoot, R.L.; Mertens, J.C.; Bronk, S.F.; Sirica, A.E.; Gores, G.J. Non-canonical Hedgehog signaling contributes to chemotaxis in cholangiocarcinoma. J. Hepatol. 2014, 60, 599–605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Nüsslein-Volhard, C.; Wieschaus, E. Mutations affecting segment number and polarity in Drosophila. Nature 1980, 287, 795–801. [Google Scholar] [CrossRef] [PubMed]
  59. Pandolfi, S.; Stecca, B. Cooperative integration between HEDGEHOG-GLI signalling and other oncogenic pathways: Implications for cancer therapy. Expert Rev. Mol. Med. 2015, 17, e5. [Google Scholar] [CrossRef]
  60. Pietrobono, S.; Gagliardi, S.; Stecca, B. Non-canonical Hedgehog Signaling Pathway in Cancer: Activation of GLI Transcription Factors Beyond Smoothened. Front. Genet. 2019, 10, 556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Briscoe, J.; Thérond, P.P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 2013, 14, 416–429. [Google Scholar] [CrossRef] [PubMed]
  62. McMahon, A.P.; Ingham, P.W.; Tabin, C.J. Developmental roles and clinical significance of hedgehog signaling. Curr. Top. Dev. Biol. 2003, 53, 1–114. [Google Scholar] [CrossRef] [PubMed]
  63. Sagai, T.; Hosoya, M.; Mizushina, Y.; Tamura, M.; Shiroishi, T. Elimination of a long-range cis-regulatory module causes complete loss of limb-specific Shh expression and truncation of the mouse limb. Development 2005, 132, 797–803. [Google Scholar] [CrossRef] [Green Version]
  64. Ingham, P.W.; McMahon, A.P. Hedgehog signaling in animal development: Paradigms and principles. Genes Dev. 2001, 15, 3059–3087. [Google Scholar] [CrossRef] [Green Version]
  65. Varjosalo, M.; Taipale, J. Hedgehog: Functions and mechanisms. Genes Dev. 2008, 22, 2454–2472. [Google Scholar] [CrossRef] [Green Version]
  66. Dyer, M.A.; Farrington, S.M.; Mohn, D.; Munday, J.R.; Baron, M.H. Indian hedgehog activates hematopoiesis and vasculogenesis and can respecify prospective neurectodermal cell fate in the mouse embryo. Development 2001, 128, 1717–1730. [Google Scholar] [CrossRef] [PubMed]
  67. Vortkamp, A.; Lee, K.; Lanske, B.; Segre, G.V.; Kronenberg, H.M.; Tabin, C.J. Regulation of rate of cartilage differentiation by Indian hedgehog and PTH-related protein. Science 1996, 273, 613–622. [Google Scholar] [CrossRef] [PubMed]
  68. St-Jacques, B.; Hammerschmidt, M.; McMahon, A.P. Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev. 1999, 13, 2072–2086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Bitgood, M.J.; Shen, L.; McMahon, A.P. Sertoli cell signaling by Desert hedgehog regulates the male germline. Curr. Biol. 1996, 6, 298–304. [Google Scholar] [CrossRef] [Green Version]
  70. Yao, H.H.; Whoriskey, W.; Capel, B. Desert Hedgehog/Patched 1 signaling specifies fetal Leydig cell fate in testis organogenesis. Genes Dev. 2002, 16, 1433–1440. [Google Scholar] [CrossRef] [Green Version]
  71. Wijgerde, M.; Ooms, M.; Hoogerbrugge, J.W.; Grootegoed, J.A. Hedgehog signaling in mouse ovary: Indian hedgehog and desert hedgehog from granulosa cells induce target gene expression in developing theca cells. Endocrinology 2005, 146, 3558–3566. [Google Scholar] [CrossRef]
  72. Denef, N.; Neubüser, D.; Perez, L.; Cohen, S.M. Hedgehog induces opposite changes in turnover and subcellular localization of patched and smoothened. Cell 2000, 102, 521–531. [Google Scholar] [CrossRef] [Green Version]
  73. Rohatgi, R.; Milenkovic, L.; Scott, M.P. Patched1 regulates hedgehog signaling at the primary cilium. Science 2007, 317, 372–376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kogerman, P.; Grimm, T.; Kogerman, L.; Krause, D.; Undén, A.B.; Sandstedt, B.; Toftgård, R.; Zaphiropoulos, P.G. Mammalian suppressor-of-fused modulates nuclear-cytoplasmic shuttling of Gli-1. Nat. Cell Biol. 1999, 1, 312–319. [Google Scholar] [CrossRef]
  75. Wang, B.; Li, Y. Evidence for the direct involvement of {β}TrCP in Gli3 protein processing. Proc. Natl. Acad. Sci. USA 2006, 103, 33–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Niewiadomski, P.; Kong, J.H.; Ahrends, R.; Ma, Y.; Humke, E.W.; Khan, S.; Teruel, M.N.; Novitch, B.G.; Rohatgi, R. Gli protein activity is controlled by multisite phosphorylation in vertebrate Hedgehog signaling. Cell Rep. 2014, 6, 168–181. [Google Scholar] [CrossRef] [Green Version]
  77. Pan, Y.; Bai, C.B.; Joyner, A.L.; Wang, B. Sonic hedgehog signaling regulates Gli2 transcriptional activity by suppressing its processing and degradation. Mol. Cell. Biol. 2006, 26, 3365–3377. [Google Scholar] [CrossRef] [Green Version]
  78. Huntzicker, E.G.; Estay, I.S.; Zhen, H.; Lokteva, L.A.; Jackson, P.K.; Oro, A.E. Dual degradation signals control Gli protein stability and tumor formation. Genes Dev. 2006, 20, 276–281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Chen, Y.; Sasai, N.; Ma, G.; Yue, T.; Jia, J.; Briscoe, J.; Jiang, J. Sonic Hedgehog dependent phosphorylation by CK1α and GRK2 is required for ciliary accumulation and activation of smoothened. PLoS Biol. 2011, 9, e1001083. [Google Scholar] [CrossRef] [Green Version]
  80. Corbit, K.C.; Aanstad, P.; Singla, V.; Norman, A.R.; Stainier, D.Y.; Reiter, J.F. Vertebrate Smoothened functions at the primary cilium. Nature 2005, 437, 1018–1021. [Google Scholar] [CrossRef] [PubMed]
  81. Kovacs, J.J.; Whalen, E.J.; Liu, R.; Xiao, K.; Kim, J.; Chen, M.; Wang, J.; Chen, W.; Lefkowitz, R.J. β-arrestin-mediated localization of smoothened to the primary cilium. Science 2008, 320, 1777–1781. [Google Scholar] [CrossRef] [Green Version]
  82. Katoh, Y.; Katoh, M. Hedgehog target genes: Mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr. Mol. Med. 2009, 9, 873–886. [Google Scholar] [CrossRef] [PubMed]
  83. Gupta, S.; Takebe, N.; Lorusso, P. Targeting the Hedgehog pathway in cancer. Ther. Adv. Med Oncol. 2010, 2, 237–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ng, J.M.; Curran, T. The Hedgehog’s tale: Developing strategies for targeting cancer. Nat. Rev. Cancer 2011, 11, 493–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kinzler, K.W.; Bigner, S.H.; Bigner, D.D.; Trent, J.M.; Law, M.L.; O’Brien, S.J.; Wong, A.J.; Vogelstein, B. Identification of an amplified, highly expressed gene in a human glioma. Science 1987, 236, 70–73. [Google Scholar] [CrossRef]
  86. Dierks, C.; Grbic, J.; Zirlik, K.; Beigi, R.; Englund, N.P.; Guo, G.R.; Veelken, H.; Engelhardt, M.; Mertelsmann, R.; Kelleher, J.F.; et al. Essential role of stromally induced hedgehog signaling in B-cell malignancies. Nat. Med. 2007, 13, 944–951. [Google Scholar] [CrossRef] [PubMed]
  87. Pietrobono, S.; Stecca, B. Targeting the Oncoprotein Smoothened by Small Molecules: Focus on Novel Acylguanidine Derivatives as Potent Smoothened Inhibitors. Cells 2018, 7, 272. [Google Scholar] [CrossRef] [Green Version]
  88. Samatar, A.A.; Poulikakos, P.I. Targeting RAS-ERK signalling in cancer: Promises and challenges. Nat. Rev. Drug Discov. 2014, 13, 928–942. [Google Scholar] [CrossRef] [PubMed]
  89. Rovida, E.; Stecca, B. Mitogen-activated protein kinases and Hedgehog-GLI signaling in cancer: A crosstalk providing therapeutic opportunities? Semin. Cancer Biol. 2015, 35, 154–167. [Google Scholar] [CrossRef]
  90. Buonamici, S.; Williams, J.; Morrissey, M.; Wang, A.; Guo, R.; Vattay, A.; Hsiao, K.; Yuan, J.; Green, J.; Ospina, B.; et al. Interfering with resistance to smoothened antagonists by inhibition of the PI3K pathway in medulloblastoma. Sci. Transl. Med. 2010, 2, 51ra70. [Google Scholar] [CrossRef] [Green Version]
  91. Dennler, S.; André, J.; Alexaki, I.; Li, A.; Magnaldo, T.; ten Dijke, P.; Wang, X.J.; Verrecchia, F.; Mauviel, A. Induction of sonic hedgehog mediators by transforming growth factor-β: Smad3-dependent activation of Gli2 and Gli1 expression in vitro and in vivo. Cancer Res. 2007, 67, 6981–6986. [Google Scholar] [CrossRef] [Green Version]
  92. Johnson, R.W.; Nguyen, M.P.; Padalecki, S.S.; Grubbs, B.G.; Merkel, A.R.; Oyajobi, B.O.; Matrisian, L.M.; Mundy, G.R.; Sterling, J.A. TGF-β promotion of Gli2-induced expression of parathyroid hormone-related protein, an important osteolytic factor in bone metastasis, is independent of canonical Hedgehog signaling. Cancer Res. 2011, 71, 822–831. [Google Scholar] [CrossRef] [Green Version]
  93. O’Toole, S.A.; Swarbrick, A.; Sutherland, R.L. The Hedgehog signalling pathway as a therapeutic target in early breast cancer development. Expert Opin. Ther. Targets 2009, 13, 1095–1103. [Google Scholar] [CrossRef] [PubMed]
  94. Scales, S.J.; de Sauvage, F.J. Mechanisms of Hedgehog pathway activation in cancer and implications for therapy. Trends Pharmacol. Sci. 2009, 30, 303–312. [Google Scholar] [CrossRef] [PubMed]
  95. Teglund, S.; Toftgård, R. Hedgehog beyond medulloblastoma and basal cell carcinoma. Biochim. Biophys. Acta 2010, 1805, 181–208. [Google Scholar] [CrossRef] [PubMed]
  96. McMillan, R.; Matsui, W. Molecular pathways: The hedgehog signaling pathway in cancer. Clin. Cancer Res. 2012, 18, 4883–4888. [Google Scholar] [CrossRef] [Green Version]
  97. Hirose, Y.; Itoh, T.; Miyajima, A. Hedgehog signal activation coordinates proliferation and differentiation of fetal liver progenitor cells. Exp. Cell Res. 2009, 315, 2648–2657. [Google Scholar] [CrossRef]
  98. Jeng, K.S.; Chang, C.F.; Lin, S.-S. Sonic Hedgehog Signaling in Organogenesis, Tumors, and Tumor Microenvironments. Int. J. Mol. Sci. 2020, 21, 758. [Google Scholar] [CrossRef] [Green Version]
  99. Sicklick, J.K.; Li, Y.X.; Melhem, A.; Schmelzer, E.; Zdanowicz, M.; Huang, J.; Caballero, M.; Fair, J.H.; Ludlow, J.W.; McClelland, R.E.; et al. Hedgehog signaling maintains resident hepatic progenitors throughout life. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 290, G859–G870. [Google Scholar] [CrossRef]
  100. Machado, M.V.; Diehl, A.M. Hedgehog signalling in liver pathophysiology. J. Hepatol. 2018, 68, 550–562. [Google Scholar] [CrossRef] [Green Version]
  101. Omenetti, A.; Choi, S.; Michelotti, G.; Diehl, A.M. Hedgehog signaling in the liver. J. Hepatol. 2011, 54, 366–373. [Google Scholar] [CrossRef]
  102. Omenetti, A.; Popov, Y.; Jung, Y.; Choi, S.S.; Witek, R.P.; Yang, L.; Brown, K.D.; Schuppan, D.; Diehl, A.M. The hedgehog pathway regulates remodelling responses to biliary obstruction in rats. Gut 2008, 57, 1275–1282. [Google Scholar] [CrossRef] [PubMed]
  103. Omenetti, A.; Yang, L.; Li, Y.X.; McCall, S.J.; Jung, Y.; Sicklick, J.K.; Huang, J.; Choi, S.; Suzuki, A.; Diehl, A.M. Hedgehog-mediated mesenchymal-epithelial interactions modulate hepatic response to bile duct ligation. Lab. Investig. J. Tech. Methods Pathol. 2007, 87, 499–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Jung, Y.; McCall, S.J.; Li, Y.X.; Diehl, A.M. Bile ductules and stromal cells express hedgehog ligands and/or hedgehog target genes in primary biliary cirrhosis. Hepatology 2007, 45, 1091–1096. [Google Scholar] [CrossRef] [PubMed]
  105. Choi, S.S.; Omenetti, A.; Witek, R.P.; Moylan, C.A.; Syn, W.K.; Jung, Y.; Yang, L.; Sudan, D.L.; Sicklick, J.K.; Michelotti, G.A.; et al. Hedgehog pathway activation and epithelial-to-mesenchymal transitions during myofibroblastic transformation of rat hepatic cells in culture and cirrhosis. Am. J. Physiol. Gastrointest. Liver Physiol. 2009, 297, G1093–G1106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Choi, S.S.; Witek, R.P.; Yang, L.; Omenetti, A.; Syn, W.K.; Moylan, C.A.; Jung, Y.; Karaca, G.F.; Teaberry, V.S.; Pereira, T.A.; et al. Activation of Rac1 promotes hedgehog-mediated acquisition of the myofibroblastic phenotype in rat and human hepatic stellate cells. Hepatology 2010, 52, 278–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Sicklick, J.K.; Li, Y.X.; Choi, S.S.; Qi, Y.; Chen, W.; Bustamante, M.; Huang, J.; Zdanowicz, M.; Camp, T.; Torbenson, M.S.; et al. Role for hedgehog signaling in hepatic stellate cell activation and viability. Lab. Investig. J. Tech. Methods Pathol. 2005, 85, 1368–1380. [Google Scholar] [CrossRef] [PubMed]
  108. Yang, L.; Wang, Y.; Mao, H.; Fleig, S.; Omenetti, A.; Brown, K.D.; Sicklick, J.K.; Li, Y.-X.; Diehl, A.M. Sonic hedgehog is an autocrine viability factor for myofibroblastic hepatic stellate cells. J. Hepatol. 2008, 48, 98–106. [Google Scholar] [CrossRef] [Green Version]
  109. Shackel, N.A.; McGuinness, P.H.; Abbott, C.A.; Gorrell, M.D.; McCaughan, G.W. Identification of novel molecules and pathogenic pathways in primary biliary cirrhosis: cDNA array analysis of intrahepatic differential gene expression. Gut 2001, 49, 565–576. [Google Scholar] [CrossRef]
  110. Omenetti, A.; Diehl, A.M. The adventures of sonic hedgehog in development and repair. II. Sonic hedgehog and liver development, inflammation, and cancer. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 294, G595–G598. [Google Scholar] [CrossRef] [PubMed]
  111. Razumilava, N.; Gores, G.J. Cholangiocarcinoma. Lancet 2014, 383, 2168–2179. [Google Scholar] [CrossRef] [Green Version]
  112. Kim, Y.; Kim, M.O.; Shin, J.S.; Park, S.H.; Kim, S.B.; Kim, J.; Park, S.C.; Han, C.J.; Ryu, J.K.; Yoon, Y.B.; et al. Hedgehog signaling between cancer cells and hepatic stellate cells in promoting cholangiocarcinoma. Ann. Surg. Oncol. 2014, 21, 2684–2698. [Google Scholar] [CrossRef]
  113. Tang, L.; Tan, Y.X.; Jiang, B.G.; Pan, Y.F.; Li, S.X.; Yang, G.Z.; Wang, M.; Wang, Q.; Zhang, J.; Zhou, W.P.; et al. The prognostic significance and therapeutic potential of hedgehog signaling in intrahepatic cholangiocellular carcinoma. Clin. Cancer Res. 2013, 19, 2014–2024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Eichenmüller, M.; Gruner, I.; Hagl, B.; Häberle, B.; Müller-Höcker, J.; von Schweinitz, D.; Kappler, R. Blocking the hedgehog pathway inhibits hepatoblastoma growth. Hepatology 2009, 49, 482–490. [Google Scholar] [CrossRef]
  115. Li, Y.-C.; Deng, Y.-H.; Guo, Z.-H.; Zhang, M.-M.; Zhu, J.; Pu, C.-L.; Xiang, C.-P.; Guo, C.-B.; Li, Y.-C.; Deng, Y.-H.; et al. Prognostic value of hedgehog signal component expressions in hepatoblastoma patients. Eur. J. Med. Res. 2010, 15, 468–474. [Google Scholar] [CrossRef] [Green Version]
  116. Matsushita, S.; Onishi, H.; Nakano, K.; Nagamatsu, I.; Imaizumi, A.; Hattori, M.; Oda, Y.; Tanaka, M.; Katano, M. Hedgehog signaling pathway is a potential therapeutic target for gallbladder cancer. Cancer Sci. 2014, 105, 272–280. [Google Scholar] [CrossRef]
  117. Takebe, N.; Yang, S.X. Sonic hedgehog signaling pathway and gallbladder cancer: Targeting with precision medicine approach. Chin. Clin. Oncol. 2016, 5, 1. [Google Scholar] [CrossRef]
  118. Dixit, R.; Pandey, M.; Tripathi, S.K.; Dwivedi, A.N.; Shukla, V.K. Comparative Analysis of Mutational Profile of Sonic hedgehog Gene in Gallbladder Cancer. Dig. Dis. Sci. 2017, 62, 708–714. [Google Scholar] [CrossRef]
  119. Cerami, E.; Gao, J.; Dogrusoz, U.; Gross, B.E.; Sumer, S.O.; Aksoy, B.A.; Jacobsen, A.; Byrne, C.J.; Heuer, M.L.; Larsson, E.; et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2, 401–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 2013, 6, pl1. [Google Scholar] [CrossRef] [Green Version]
  121. Ohta, H.; Aoyagi, K.; Fukaya, M.; Danjoh, I.; Ohta, A.; Isohata, N.; Saeki, N.; Taniguchi, H.; Sakamoto, H.; Shimoda, T.; et al. Cross talk between hedgehog and epithelial-mesenchymal transition pathways in gastric pit cells and in diffuse-type gastric cancers. Br. J. Cancer 2009, 100, 389–398. [Google Scholar] [CrossRef]
  122. Riaz, S.K.; Khan, J.S.; Shah, S.T.A.; Wang, F.; Ye, L.; Jiang, W.G.; Malik, M.F.A. Involvement of hedgehog pathway in early onset, aggressive molecular subtypes and metastatic potential of breast cancer. Cell Commun. Signal. CCS 2018, 16, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Jäger, W.; Thomas, C.; Fazli, L.; Hurtado-Coll, A.; Li, E.; Janssen, C.; Gust, K.M.; So, A.I.; Hainz, M.; Schmidtmann, I.; et al. DHH is an independent prognosticator of oncologic outcome of clear cell renal cell carcinoma. J. Urol. 2014, 192, 1842–1848. [Google Scholar] [CrossRef] [PubMed]
  124. Masetti, R.; Togni, M.; Astolfi, A.; Pigazzi, M.; Manara, E.; Indio, V.; Rizzari, C.; Rutella, S.; Basso, G.; Pession, A.; et al. DHH-RHEBL1 fusion transcript: A novel recurrent feature in the new landscape of pediatric CBFA2T3-GLIS2-positive acute myeloid leukemia. Oncotarget 2013, 4, 1712–1720. [Google Scholar] [CrossRef] [Green Version]
  125. Taylor, M.D.; Liu, L.; Raffel, C.; Hui, C.C.; Mainprize, T.G.; Zhang, X.; Agatep, R.; Chiappa, S.; Gao, L.; Lowrance, A.; et al. Mutations in SUFU predispose to medulloblastoma. Nat. Genet. 2002, 31, 306–310. [Google Scholar] [CrossRef] [PubMed]
  126. Xie, J.; Murone, M.; Luoh, S.M.; Ryan, A.; Gu, Q.; Zhang, C.; Bonifas, J.M.; Lam, C.W.; Hynes, M.; Goddard, A.; et al. Activating Smoothened mutations in sporadic basal-cell carcinoma. Nature 1998, 391, 90–92. [Google Scholar] [CrossRef]
  127. Northcott, P.A.; Nakahara, Y.; Wu, X.; Feuk, L.; Ellison, D.W.; Croul, S.; Mack, S.; Kongkham, P.N.; Peacock, J.; Dubuc, A.; et al. Multiple recurrent genetic events converge on control of histone lysine methylation in medulloblastoma. Nat. Genet. 2009, 41, 465–472. [Google Scholar] [CrossRef]
  128. Reifenberger, J.; Wolter, M.; Knobbe, C.B.; Köhler, B.; Schönicke, A.; Scharwächter, C.; Kumar, K.; Blaschke, B.; Ruzicka, T.; Reifenberger, G. Somatic mutations in the PTCH, SMOH, SUFUH and TP53 genes in sporadic basal cell carcinomas. Br. J. Dermatol. 2005, 152, 43–51. [Google Scholar] [CrossRef]
  129. Dahmane, N.; Lee, J.; Robins, P.; Heller, P.; Ruiz i Altaba, A. Activation of the transcription factor Gli1 and the Sonic hedgehog signalling pathway in skin tumours. Nature 1997, 389, 876–881. [Google Scholar] [CrossRef]
  130. McGarvey, T.W.; Maruta, Y.; Tomaszewski, J.E.; Linnenbach, A.J.; Malkowicz, S.B. PTCH gene mutations in invasive transitional cell carcinoma of the bladder. Oncogene 1998, 17, 1167–1172. [Google Scholar] [CrossRef] [Green Version]
  131. Skoda, A.M.; Simovic, D.; Karin, V.; Kardum, V.; Vranic, S.; Serman, L. The role of the Hedgehog signaling pathway in cancer: A comprehensive review. Bosn. J. Basic Med. Sci. 2018, 18, 8–20. [Google Scholar] [CrossRef]
  132. Kool, M.; Jones, D.T.; Jäger, N.; Northcott, P.A.; Pugh, T.J.; Hovestadt, V.; Piro, R.M.; Esparza, L.A.; Markant, S.L.; Remke, M.; et al. Genome sequencing of SHH medulloblastoma predicts genotype-related response to smoothened inhibition. Cancer Cell 2014, 25, 393–405. [Google Scholar] [CrossRef] [Green Version]
  133. El Khatib, M.; Kalnytska, A.; Palagani, V.; Kossatz, U.; Manns, M.P.; Malek, N.P.; Wilkens, L.; Plentz, R.R. Inhibition of hedgehog signaling attenuates carcinogenesis in vitro and increases necrosis of cholangiocellular carcinoma. Hepatology 2013, 57, 1035–1045. [Google Scholar] [CrossRef]
  134. Kiesslich, T.; Mayr, C.; Wachter, J.; Bach, D.; Fuereder, J.; Wagner, A.; Alinger, B.; Pichler, M.; Di Fazio, P.; Ocker, M.; et al. Activated hedgehog pathway is a potential target for pharmacological intervention in biliary tract cancer. Mol. Cell. Biochem. 2014, 396, 257–268. [Google Scholar] [CrossRef] [PubMed]
  135. Al-Bahrani, R.; Nagamori, S.; Leng, R.; Petryk, A.; Sergi, C. Differential Expression of Sonic Hedgehog Protein in Human Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. Pathol. Oncol. Res. 2015, 21, 901–908. [Google Scholar] [CrossRef]
  136. Nanashima, A.; Hatachi, G.; Tsuchiya, T.; Matsumoto, H.; Arai, J.; Abo, T.; Murakami, G.; Tominaga, T.; Takagi, K.; Nagayasu, T. Clinical significances of cancer stem cells markers in patients with intrahepatic cholangiocarcinoma who underwent hepatectomy. Anticancer Res. 2013, 33, 2107–2114. [Google Scholar]
  137. Peng, Y.; Meng, G.; Sheng, X.; Gao, H. Transcriptome and DNA methylation analysis reveals molecular mechanisms underlying intrahepatic cholangiocarcinoma progression. J. Cell. Mol. Med. 2021, 25, 6373–6387. [Google Scholar] [CrossRef]
  138. Berman, D.M.; Karhadkar, S.S.; Maitra, A.; Montes De Oca, R.; Gerstenblith, M.R.; Briggs, K.; Parker, A.R.; Shimada, Y.; Eshleman, J.R.; Watkins, D.N.; et al. Widespread requirement for Hedgehog ligand stimulation in growth of digestive tract tumours. Nature 2003, 425, 846–851. [Google Scholar] [CrossRef]
  139. Guo, L.; Zhou, Y.; Chen, Y.; Sun, H.; Wang, Y.; Qu, Y. LncRNA ASAP1-IT1 positively modulates the development of cholangiocarcinoma via hedgehog signaling pathway. Biomed. Pharmacother. 2018, 103, 167–173. [Google Scholar] [CrossRef]
  140. Kurita, S.; Mott, J.L.; Almada, L.L.; Bronk, S.F.; Werneburg, N.W.; Sun, S.Y.; Roberts, L.R.; Fernandez-Zapico, M.E.; Gores, G.J. GLI3-dependent repression of DR4 mediates hedgehog antagonism of TRAIL-induced apoptosis. Oncogene 2010, 29, 4848–4858. [Google Scholar] [CrossRef] [Green Version]
  141. Razumilava, N.; Bronk, S.F.; Smoot, R.L.; Fingas, C.D.; Werneburg, N.W.; Roberts, L.R.; Mott, J.L. miR-25 targets TNF-related apoptosis inducing ligand (TRAIL) death receptor-4 and promotes apoptosis resistance in cholangiocarcinoma. Hepatology 2012, 55, 465–475. [Google Scholar] [CrossRef]
  142. Mansini, A.P.; Peixoto, E.; Thelen, K.M.; Gaspari, C.; Jin, S.; Gradilone, S.A. The cholangiocyte primary cilium in health and disease. Biochim. Biophys. Acta. Mol. Basis Dis. 2018, 1864, 1245–1253. [Google Scholar] [CrossRef]
  143. Wheway, G.; Nazlamova, L.; Hancock, J.T. Signaling through the Primary Cilium. Front. Cell Dev. Biol. 2018, 6, 8. [Google Scholar] [CrossRef]
  144. Wong, S.Y.; Seol, A.D.; So, P.L.; Ermilov, A.N.; Bichakjian, C.K.; Epstein, E.H., Jr.; Dlugosz, A.A.; Reiter, J.F. Primary cilia can both mediate and suppress Hedgehog pathway-dependent tumorigenesis. Nat. Med. 2009, 15, 1055–1061. [Google Scholar] [CrossRef] [Green Version]
  145. Han, Y.G.; Kim, H.J.; Dlugosz, A.A.; Ellison, D.W.; Gilbertson, R.J.; Alvarez-Buylla, A. Dual and opposing roles of primary cilia in medulloblastoma development. Nat. Med. 2009, 15, 1062–1065. [Google Scholar] [CrossRef] [Green Version]
  146. Hanna, A.; Shevde, L.A. Hedgehog signaling: Modulation of cancer properies and tumor mircroenvironment. Mol. Cancer 2016, 15, 24. [Google Scholar] [CrossRef] [Green Version]
  147. Moeller, B.J.; Richardson, R.A.; Dewhirst, M.W. Hypoxia and radiotherapy: Opportunities for improved outcomes in cancer treatment. Cancer Metastasis Rev. 2007, 26, 241–248. [Google Scholar] [CrossRef]
  148. Shannon, A.M.; Bouchier-Hayes, D.J.; Condron, C.M.; Toomey, D. Tumour hypoxia, chemotherapeutic resistance and hypoxia-related therapies. Cancer Treat. Rev. 2003, 29, 297–307. [Google Scholar] [CrossRef]
  149. Ellis, L.M.; Hicklin, D.J. Resistance to Targeted Therapies: Refining Anticancer Therapy in the Era of Molecular Oncology. Clin. Cancer Res. 2009, 15, 7471–7478. [Google Scholar] [CrossRef] [Green Version]
  150. Tang, Y.A.; Chen, Y.F.; Bao, Y.; Mahara, S.; Yatim, S.; Oguz, G.; Lee, P.L.; Feng, M.; Cai, Y.; Tan, E.Y.; et al. Hypoxic tumor microenvironment activates GLI2 via HIF-1α and TGF-β2 to promote chemoresistance in colorectal cancer. Proc. Natl. Acad. Sci. USA 2018, 115, e5990–e5999. [Google Scholar] [CrossRef] [Green Version]
  151. Chen, S.; Zhang, M.; Xing, L.; Wang, Y.; Xiao, Y.; Wu, Y. HIF-1α contributes to proliferation and invasiveness of neuroblastoma cells via SHH signaling. PLoS ONE 2015, 10, e0121115. [Google Scholar] [CrossRef]
  152. Vanichapol, T.; Leelawat, K.; Hongeng, S. Hypoxia enhances cholangiocarcinoma invasion through activation of hepatocyte growth factor receptor and the extracellular signal-regulated kinase signaling pathway. Mol. Med. Rep. 2015, 12, 3265–3272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Jing, X.; Yang, F.; Shao, C.; Wei, K.; Xie, M.; Shen, H.; Shu, Y. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 2019, 18, 157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Gentilini, A.; Pastore, M.; Marra, F.; Raggi, C. The Role of Stroma in Cholangiocarcinoma: The Intriguing Interplay between Fibroblastic Component, Immune Cell Subsets and Tumor Epithelium. Int. J. Mol. Sci. 2018, 19, 2885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Fingas, C.D.; Bronk, S.F.; Werneburg, N.W.; Mott, J.L.; Guicciardi, M.E.; Cazanave, S.C.; Mertens, J.C.; Sirica, A.E.; Gores, G.J. Myofibroblast-derived PDGF-BB promotes Hedgehog survival signaling in cholangiocarcinoma cells. Hepatology 2011, 54, 2076–2088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Fingas, C.D.; Mertens, J.C.; Razumilava, N.; Sydor, S.; Bronk, S.F.; Christensen, J.D.; Rizvi, S.H.; Canbay, A.; Treckmann, J.W.; Paul, A.; et al. Polo-like kinase 2 is a mediator of hedgehog survival signaling in cholangiocarcinoma. Hepatology 2013, 58, 1362–1374. [Google Scholar] [CrossRef]
  157. Milenkovic, L.; Scott, M.P. Not lost in space: Trafficking in the hedgehog signaling pathway. Sci. Signal. 2010, 3, pe14. [Google Scholar] [CrossRef] [Green Version]
  158. Walter, K.; Omura, N.; Hong, S.M.; Griffith, M.; Vincent, A.; Borges, M.; Goggins, M. Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clin. Cancer Res. 2010, 16, 1781–1789. [Google Scholar] [CrossRef] [Green Version]
  159. Theunissen, J.W.; de Sauvage, F.J. Paracrine Hedgehog signaling in cancer. Cancer Res. 2009, 69, 6007–6010. [Google Scholar] [CrossRef] [Green Version]
  160. Boulter, L.; Guest, R.V.; Kendall, T.J.; Wilson, D.H.; Wojtacha, D.; Robson, A.J.; Ridgway, R.A.; Samuel, K.; Van Rooijen, N.; Barry, S.T.; et al. WNT signaling drives cholangiocarcinoma growth and can be pharmacologically inhibited. J. Clin. Investig. 2015, 125, 1269–1285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Jinawath, A.; Akiyama, Y.; Sripa, B.; Yuasa, Y. Dual blockade of the Hedgehog and ERK1/2 pathways coordinately decreases proliferation and survival of cholangiocarcinoma cells. J. Cancer Res. Clin. Oncol. 2007, 133, 271–278. [Google Scholar] [CrossRef] [PubMed]
  162. Sekulic, A.; Migden, M.R.; Oro, A.E.; Dirix, L.; Lewis, K.D.; Hainsworth, J.D.; Solomon, J.A.; Yoo, S.; Arron, S.T.; Friedlander, P.A.; et al. Efficacy and safety of vismodegib in advanced basal-cell carcinoma. N. Engl. J. Med. 2012, 366, 2171–2179. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Dong, X.; Wang, C.; Chen, Z.; Zhao, W. Overcoming the resistance mechanisms of Smoothened inhibitors. Drug Discov. Today 2018, 23, 704–710. [Google Scholar] [CrossRef]
  164. Yauch, R.L.; Dijkgraaf, G.J.; Alicke, B.; Januario, T.; Ahn, C.P.; Holcomb, T.; Pujara, K.; Stinson, J.; Callahan, C.A.; Tang, T.; et al. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 2009, 326, 572–574. [Google Scholar] [CrossRef] [Green Version]
  165. Tao, H.; Jin, Q.; Koo, D.I.; Liao, X.; Englund, N.P.; Wang, Y.; Ramamurthy, A.; Schultz, P.G.; Dorsch, M.; Kelleher, J.; et al. Small molecule antagonists in distinct binding modes inhibit drug-resistant mutant of smoothened. Chem. Biol. 2011, 18, 432–437. [Google Scholar] [CrossRef] [Green Version]
  166. Kim, J.; Aftab, B.T.; Tang, J.Y.; Kim, D.; Lee, A.H.; Rezaee, M.; Kim, J.; Chen, B.; King, E.M.; Borodovsky, A.; et al. Itraconazole and arsenic trioxide inhibit Hedgehog pathway activation and tumor growth associated with acquired resistance to smoothened antagonists. Cancer Cell 2013, 23, 23–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Chen, B.; Trang, V.; Lee, A.; Williams, N.S.; Wilson, A.N.; Epstein, E.H., Jr.; Tang, J.Y.; Kim, J. Posaconazole, a Second-Generation Triazole Antifungal Drug, Inhibits the Hedgehog Signaling Pathway and Progression of Basal Cell Carcinoma. Mol. Cancer Ther. 2016, 15, 866–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Wang, J.; Mook, R.A., Jr.; Lu, J.; Gooden, D.M.; Ribeiro, A.; Guo, A.; Barak, L.S.; Lyerly, H.K.; Chen, W. Identification of a novel Smoothened antagonist that potently suppresses Hedgehog signaling. Bioorgan. Med. Chem. 2012, 20, 6751–6757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Kumar, V.; Chaudhary, A.K.; Dong, Y.; Zhong, H.A.; Mondal, G.; Lin, F.; Kumar, V.; Mahato, R.I. Design, Synthesis and Biological Evaluation of novel Hedgehog Inhibitors for treating Pancreatic Cancer. Sci. Rep. 2017, 7, 1665. [Google Scholar] [CrossRef] [Green Version]
  170. Lin, P.; He, Y.; Chen, G.; Ma, H.; Zheng, J.; Zhang, Z.; Cao, B.; Zhang, H.; Zhang, X.; Mao, X. A novel hedgehog inhibitor for the treatment of hematological malignancies. Anti-Cancer Drugs 2018, 29, 995–1003. [Google Scholar] [CrossRef]
  171. Manetti, F.; Faure, H.; Roudaut, H.; Gorojankina, T.; Traiffort, E.; Schoenfelder, A.; Mann, A.; Solinas, A.; Taddei, M.; Ruat, M. Virtual screening-based discovery and mechanistic characterization of the acylthiourea MRT-10 family as smoothened antagonists. Mol. Pharmacol. 2010, 78, 658–665. [Google Scholar] [CrossRef] [Green Version]
  172. Pietrobono, S.; Santini, R.; Gagliardi, S.; Dapporto, F.; Colecchia, D.; Chiariello, M.; Leone, C.; Valoti, M.; Manetti, F.; Petricci, E.; et al. Targeted inhibition of Hedgehog-GLI signaling by novel acylguanidine derivatives inhibits melanoma cell growth by inducing replication stress and mitotic catastrophe. Cell Death Dis. 2018, 9, 142. [Google Scholar] [CrossRef]
  173. Chiarenza, A.; Manetti, F.; Petricci, E.; Ruat, M.; Naldini, A.; Taddei, M.; Carraro, F. Novel Acylguanidine Derivatives Targeting Smoothened Induce Antiproliferative and Pro-Apoptotic Effects in Chronic Myeloid Leukemia Cells. PLoS ONE 2016, 11, e0149919. [Google Scholar] [CrossRef] [Green Version]
  174. Bernardini, G.; Geminiani, M.; Gambassi, S.; Orlandini, M.; Petricci, E.; Marzocchi, B.; Laschi, M.; Taddei, M.; Manetti, F.; Santucci, A. Novel smoothened antagonists as anti-neoplastic agents for the treatment of osteosarcoma. J. Cell. Physiol. 2018, 233, 4961–4971. [Google Scholar] [CrossRef] [PubMed]
  175. Valkenburg, K.C.; de Groot, A.E.; Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018, 15, 366–381. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The canonical Hedgehog-GLI signaling. In the absence of any Hedgehog ligand stimulation (a), the transmembrane receptor PTCH1 inhibits the G-coupled protein Smoothened (SMO). GLI transcription factors are phosphorylated by PKA, CK1, and GSK3β and are thus recognized by SuFu, which sequesters them outside the nucleus. Herein, GLI2 and GLI3 are processed into the repressive forms, which hinder the transcription of Hedgehog target genes. GLI1 is degraded through the ubiquitin-proteasome system (UPS). The presence of a Hedgehog ligand that specifically binds PTCH1 (b) releases the inhibitory regulation of SMO. Accordingly, SMO initiates an intracellular cascade, which terminates with the translocation of the GLI as active forms into the nucleus, promoting the transcription of Hedgehog target genes.
Figure 1. The canonical Hedgehog-GLI signaling. In the absence of any Hedgehog ligand stimulation (a), the transmembrane receptor PTCH1 inhibits the G-coupled protein Smoothened (SMO). GLI transcription factors are phosphorylated by PKA, CK1, and GSK3β and are thus recognized by SuFu, which sequesters them outside the nucleus. Herein, GLI2 and GLI3 are processed into the repressive forms, which hinder the transcription of Hedgehog target genes. GLI1 is degraded through the ubiquitin-proteasome system (UPS). The presence of a Hedgehog ligand that specifically binds PTCH1 (b) releases the inhibitory regulation of SMO. Accordingly, SMO initiates an intracellular cascade, which terminates with the translocation of the GLI as active forms into the nucleus, promoting the transcription of Hedgehog target genes.
Cancers 13 04774 g001
Figure 2. Schematic representation of the Hedgehog pathway and its functions in CCA. The HH signaling regulates growth, survival, stemness, and invasiveness in cholangiocarcinoma cells. HH pathway antagonists used in CCA preclinical studies are acting at the level of SHH (5E1 anti-SHH antibody), SMO (vismodegib, cyclopamine, BMS-833923), and the GLI (GANT61). They have been tested alone or in combination with other targeted molecules (U0126) or with chemotherapeutic drugs (gemcitabine, cisplatin) in vitro and in vivo. Proteins that are illustrated as HH downstream targets have been demonstrated to be modulated directly by transcriptional binding of GLI (DR4, PLK2) or as a final effect of the pharmacological inhibition of the HH cascade in CCA cells.
Figure 2. Schematic representation of the Hedgehog pathway and its functions in CCA. The HH signaling regulates growth, survival, stemness, and invasiveness in cholangiocarcinoma cells. HH pathway antagonists used in CCA preclinical studies are acting at the level of SHH (5E1 anti-SHH antibody), SMO (vismodegib, cyclopamine, BMS-833923), and the GLI (GANT61). They have been tested alone or in combination with other targeted molecules (U0126) or with chemotherapeutic drugs (gemcitabine, cisplatin) in vitro and in vivo. Proteins that are illustrated as HH downstream targets have been demonstrated to be modulated directly by transcriptional binding of GLI (DR4, PLK2) or as a final effect of the pharmacological inhibition of the HH cascade in CCA cells.
Cancers 13 04774 g002
Table 1. Genetic alterations affecting Hedgehog signaling pathway in CCA patients.
Table 1. Genetic alterations affecting Hedgehog signaling pathway in CCA patients.
GeneMissenseNonsenseIn/DelSplicingCNAFrequency (%)
GLI1Q867K, A108D, A941T, P821S, S89L, A670S, P842S, E1015D G274Afs*6 AMP2.5
GLI2P386LE431 * 2.7
GLI3P575T, D848G, Y1223F, N1203K, R875C, S1025I, G1001S, E729K, L364I, G221=, R1010W, R220P, V778I, R1189H, R1010Q, G371D, P791L, A1005D, L690I, I254V, N1203K, D428N, T167M, E1014K, E551DR667 * X158_splice 4.6
SMOS785L, G402V, T307I, A631S L23dup 0.7
PTCH1G880V, D301N, L775M, E405K, P1315L, A412T, D878H 1.1
PTCH2A396V 2.9
SUFUV323I, L68S 0.4
DHHS384P AMP5.6
Data were obtained from cBioportal public databases. The frequency reported represents the sum of frequency in point mutations and copy number alterations. *: stop codon.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Anichini, G.; Carrassa, L.; Stecca, B.; Marra, F.; Raggi, C. The Role of the Hedgehog Pathway in Cholangiocarcinoma. Cancers 2021, 13, 4774. https://doi.org/10.3390/cancers13194774

AMA Style

Anichini G, Carrassa L, Stecca B, Marra F, Raggi C. The Role of the Hedgehog Pathway in Cholangiocarcinoma. Cancers. 2021; 13(19):4774. https://doi.org/10.3390/cancers13194774

Chicago/Turabian Style

Anichini, Giulia, Laura Carrassa, Barbara Stecca, Fabio Marra, and Chiara Raggi. 2021. "The Role of the Hedgehog Pathway in Cholangiocarcinoma" Cancers 13, no. 19: 4774. https://doi.org/10.3390/cancers13194774

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