Next Article in Journal
Aquaporins: More Than Functional Monomers in a Tetrameric Arrangement
Next Article in Special Issue
Targeting the Oncoprotein Smoothened by Small Molecules: Focus on Novel Acylguanidine Derivatives as Potent Smoothened Inhibitors
Previous Article in Journal
Molecular Basis of Aquaporin-7 Permeability Regulation by pH
Previous Article in Special Issue
Targeting the Multidrug Transporter Ptch1 Potentiates Chemotherapy Efficiency
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells

Soonchunhyang Institute of Medi-bio Science, Soonchunhyang University, Cheonan 31151, Korea
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
Cells 2018, 7(11), 208; https://doi.org/10.3390/cells7110208
Submission received: 22 October 2018 / Revised: 3 November 2018 / Accepted: 5 November 2018 / Published: 10 November 2018
(This article belongs to the Collection Hedgehog Signal Transduction in Physiology and Disease)

Abstract

:
The Hedgehog (Hh) pathway is a signaling cascade that plays a crucial role in many fundamental processes, including embryonic development and tissue homeostasis. Moreover, emerging evidence has suggested that aberrant activation of Hh is associated with neoplastic transformations, malignant tumors, and drug resistance of a multitude of cancers. At the molecular level, it has been shown that Hh signaling drives the progression of cancers by regulating cancer cell proliferation, malignancy, metastasis, and the expansion of cancer stem cells (CSCs). Thus, a comprehensive understanding of Hh signaling during tumorigenesis and development of chemoresistance is necessary in order to identify potential therapeutic strategies to target various human cancers and their relapse. In this review, we discuss the molecular basis of the Hh signaling pathway and its abnormal activation in several types of human cancers. We also highlight the clinical development of Hh signaling inhibitors for cancer therapy as well as CSC-targeted therapy.

1. Introduction

The Hedgehog (Hh) signaling pathway was first discovered in 1980 by Nusslein-Volhard and Wieschaus in a large-scale genetic screening to find mutations that affect larval body segment development in the fruit fly, Drosophila melanogaster (D. melanogaster) [1]. The Hh signal transduction pathway is an evolutionarily conserved pathway [2] that plays an important role in the regulation of a variety of developmental and physiological processes involving normal stem cell differentiation and proliferation, as well as proper segregation in invertebrates and vertebrates [3,4,5]. However, misregulation of the Hh pathway during embryonic development has been found to cause a variety of birth defects [6,7,8,9,10,11] as well as several neoplastic transformations and malignant tumors [10,12,13,14,15,16,17,18]. Moreover, as the Hh pathway plays an essential role in the maintenance of somatic stem cells and pluripotent cells, it is involved in the regenerative proliferation of epithelial stem cells in the lung [19], tooth [20], liver [21], prostate [22], and bladder [23], and this signaling pathway may be correlated with the maintenance of cancer stem cells (CSCs). Thus, targeting CSCs through inhibition of the Hh signaling pathway is a potential treatment that could effectively improve the clinical outcomes of cancer patients. This review describes the signaling pathway of Hh and its aberrant activation in several cancers and highlights the current clinical trials involving Hh inhibitors for cancer therapy.

2. Elements of the Hh Signaling Pathway

Even though there are several important differences between Drosophila and other higher organisms, the mechanisms of the Hh signaling pathway are highly conserved [24]. In humans, the Hh pathway has several main components: (1) three Hh homologs, (2) Patched1 (PTCH1 in humans, Ptch1 in mice, and Ptc in Drosophila), (3) a G-protein-coupled receptor (GPCR)-like receptor Smoothened (SMO in humans and Smo in mice/Drosophila), and (4) three transcription factors (GLI1, GLI2, and GLI3) named from the correlation of GLI1 and glioma [25].

2.1. Hh Proteins

Hh proteins are one of the secreted signaling protein families that can be diffused and establish gradients in target tissues after secretion, and thus are responsible for cell interactions during embryonic development [26]. Hh proteins can act as either morphogens or mitogens which control developmental processes during multiple developmental stages in many different tissues in a dose-dependent manner [26]. Hh signaling plays an important role in postnatal bone homeostasis [27,28] as well as in regulating the development and function of theca cells [29] and Leydig cells [30], indicating that Hh ligands not only participate in embryonic patterning but are also involved in the maintenance of tissue homeostasis over the course of a lifetime. Hh proteins undergo maturation by post-translational processing and multiple covalent modifications before the active ligand is released and activates the Hh pathway [27,31]. The N-terminal signal sequence is removed after being translated, and the Hh protein is then autocatalytically cleaved internally [32]. During this process, the C-terminal domain acts as an intramolecular cholesterol transferase to promote the covalent attachment of a molecule of cholesterol to the N-terminal signaling domain [33], while a palmitic acid moiety is added to the N-terminal cysteine residue by the acyltransferase Skinny Hedgehog (Ski) to form a mature Hh protein dually modified by cholesteryl and palmitoyl adducts [34]. Surprisingly, this unusual dual lipid modification plays a vital role in how Hh ligands can move far away from the organizing centers where Hh is specifically expressed and acts as a morphogen [35,36]. Therefore, this dual lipid modification could facilitate the control of the long-range activity of Hh ligands in cancers even where the local Hh proteins are limited [37].
In mammals, there are three Hh gene family members: Sonic hedgehog (SHH), Indian hedgehog (IHH), and Desert hedgehog (DHH). They are all considered to be similar in their physiological effects, but their roles in development are different in many organ systems due to the diversely regulated patterns of expression [1]. Of all the Hh ligands, SHH is the most studied ligand and acts as a cell–cell signaling factor, modulating cell fates through an autocrine or paracrine mechanism. During embryogenesis, SHH has been shown to be expressed in embryonic structures and involved in pattern formation in the embryo, including the development of the limb at the zone of polarizing activity (ZPA), notochord, or the floor plate of the neural tube [38,39] In addition, SHH is also expressed in the development of the lung [40,41], teeth [42,43], brain [44], hair follicle [45], pancreas [46], and intestine [47]. IHH is involved in the differentiation and cell fate decision of several specific tissues, including the differentiation of colonic epithelial cells [48,49] and the decision of neuroectodermal cell fate by activating the development of the earliest hemato-vascular system [50]. It is implicated in bone formation due to its role in regulating the rate of chondrocyte differentiation [51,52] and promoting human cartilage endplate (CEP) degeneration [53]. Interestingly, SHH [54,55] and IHH [56] were shown to be involved in the commitment and differentiation of the T cell lineage, as well as the proliferation and survival of developing T cells. It was also reported that SHH regulates the B-lineage commitment of hematopoietic progenitor cells and the development of B cells [57]. Whereas SHH and IHH are closely related to each other, DHH is the closest homolog to D. melanogaster of all the discovered Hh ligands [58]. DHH expression is mainly restricted to gonads, such as Sertoli cells [58] and Leydig cells [30] in the testis and granulosa cells of growing follicles in the ovaries [29], where it plays an important role in gametogenesis and steroidogenesis. Besides this, DHH could negatively regulate erythrocyte differentiation at multiple stages in both the spleen and bone marrow [59].

2.2. PTCH

The Hh/SHH receptor is PTCH [60,61], a 12-pass transmembrane protein that has two large extracellular loops and two large intracellular loops [62,63]. Two mammalian PTCH homologs have been identified: Patched1 (PTCH1) and Patched2 (PTCH2). It was shown that they bind the three Hh ligands with equal affinity and inhibit the activity of the SMO protein [18]. While PTCH1 is primarily expressed in mesenchymal cells throughout the embryo and plays a role as the primary mediator for most SHH activities, PTCH2 is specifically expressed in skin cells and spermatocytes; it is therefore likely to participate in the function of DHH in germ cells as DHH is mainly expressed in the testis [64]. Mutations of the Patched gene have been demonstrated in several diseases such as basal cell nevus syndrome (BCNS), nevoid basal cell carcinoma syndrome, sporadic basal cell carcinomas, and medulloblastomas [65,66,67].

2.3. SMO

SMO is a seven-pass integral membrane protein that is a member of the Frizzled (FzD) class of G-protein-coupled receptors (GPCRs) and functions as a positive regulator of the Hh signaling pathway because of its physical characteristics and position in Hh signaling by acting downstream of or in parallel to Patched [68]. SMO has an extracellular cysteine-rich domain (CRD), which binds to small-molecule modulators and is therefore indispensable for SMO function in the Hh signaling pathway [69]. It has been indicated that SMO does not directly bind SHH [70]; Hh binds specifically to PTCH without any help from SMO and consequently promotes the conformational change resulting in the releasing of SMO [71]. Moreover, SMO can form a physical complex with PTCH1, which indirectly inhibits SMO activity [61]; the mechanism is still not clear, but possibly involves changes in the distribution or concentration of a small, unknown molecule [72]. In addition, SMO is induced by Hh through the phosphorylation by protein kinase A (PKA) and casein kinase I (CKI), which regulate its cell-surface accumulation and signaling activity [73].

2.4. GLI

The human Gli gene is located at chromosome 12 (q13 to q14.3) and was identified by Vogelstein in 1987 because of its gene amplification of more than 50-fold in glioblastoma multiforme (GBM) and its derived cell line [74]. In mammals, three members of the Gli gene family have been identified—GLI (or GLI1), GLI2, and GLI3, which have five successive repeats of highly conserved zinc finger DNA-binding domains, characterized as members in the Kruppel family of zinc-finger-containing transcription factors. Moreover, they require the carboxyl-terminal amino acids 1020–1091, which include an 18-amino-acid herpes simplex viral protein 16-like activation domain, to act as transcription factors in the vertebrate SHH–Patched signaling pathway [75]. These findings support the hypothesis that GLI proteins are the terminal evolutionarily conserved transcription factors of the Hh signaling pathway and directly bind to the promoters of their target genes [76]. After being translated, GLI proteins mainly undergo nuclear localization and bind their DNA binding site with high affinity to protect a 23- to 24-bp region, including the 9-base-pair consensus sequence 5′-GACCACCCA-3′ [77].
Among the three GLI family members, the Drosophila Ci is more closely functionally related to mammalian GLI2 and GLI3, as they can activate transcription and undergo proteolysis to generate both activating and repressing domains, whereas GLI1 cannot be modified post-translationally and thus only plays a role as a transcriptional activator of the Hh signaling pathway [78]. GLI1 has been demonstrated to be a mediator of the SHH signal in vertebrates [79] by binding to the promoter region of several target genes involved in different cellular processes, such as G1 cell cycle progression, tumor formation, and progression [80]. On the other hand, GLI2 primarily acts as a transcriptional activator (GLIA) with activity in patterning the ventral regions of the spinal cord, whereas GLI3 mainly functions as a transcriptional repressor (GLIR) of the Hh signaling pathway, and plays a role in patterning the intermediate spinal cord [81]. Mechanistically, the three mammalian GLI proteins share a similar DNA-binding domain including five zinc finger repeats and an activation domain in the C-terminal, but only GLI2 and GLI3 have the repression domain at the N-terminus even though GLI2 acts only as a weak transcriptional activator [82,83,84].

3. Cascades of Canonical Hh Signaling

In the absence of an Hh ligand, PTCH1, a 12-pass transmembrane receptor, localizes at the base of the primary cilium (PC), a subcellular membrane extension that is important for the distribution and function of most of the components in this pathway. PTCH1 then destabilizes SMO by repressing its ciliary accumulation and consequently inhibits SMO activity, which is an essential step for activating this pathway in mammals (but not in Drosophila) [85]. The full-length GLI is therefore sequentially phosphorylated at multiple sites in the C-terminal region by protein kinase A (PKA), glycogen synthase kinase-3 (GSK3), and casein kinase 1 (CK1), [86]. The phosphorylated GLI protein directly binds β-transducin-repeat-containing protein (β-TrCP) in the Skp1-Cul1-F-box protein (SCF) ubiquitin–ligase complex via two binding sites; after that, it is subsequently ubiquitinated and degraded by the proteasome [87]. The truncated N-terminal GLI repressor (GLIR) then translocates to the nucleus and binds to Hh target gene promoters and switches off their expression in a sustained manner [88].
In the canonical Hh signaling pathway, Hh regulates the GLI family of transcription factors. Upon Hh binding to PTCH1, PTCH1 leaves the cilia by internalizing together with its ligand SHH, and both proteins are degraded in lysosomes [89]. This binding therefore relieves the inhibition of SMO. SMO is then accumulated and translocated into the PC [90]. The Hh signaling is subsequently activated and transmitted via a protein complex, which includes kinesin protein (Kif7) and Suppressor of fused (Sufu). In the absence of Hh, Sufu restrains GLI protein in the cytoplasm, whereas binding of Hh to its receptor activates the release of GLI from Sufu and allows it to enter the nucleus [91]. In addition, Kif7 has also been involved in the regulation of the Hh signaling pathway as a ciliary motor in which the activation of the Hh pathway promotes its ciliary trafficking from the base to the tip of the PC [92]. In the absence of Hh, Kif7 accumulates at the base of the PC, and Kif7 physically interacts and prevents GLI protein accumulation within the cilia by controlling their proteolysis and stability [93]. Upon Hh binding, Kif7 localizes at the distal tip of the PC and prevents the cleavage of GLI3 into a transcription repressor [94]. In the end, GLIFL is then converted to its active form GLIA and migrates to the nucleus to activate the expression of a number of target genes including PTCH1 [95]. A schematic of the activation of the canonical Hh signaling pathway is shown in Figure 1.

4. Noncanonical Hedgehog Pathway

However, emerging evidence over the past few years has indicated that Hh proteins can also signal through GLI-independent mechanisms, the so-called “noncanonical” Hh signaling [5]. In this pathway, all the responses to Hh ligands are not through transcriptional changes mediated by the transcription factors of the Gli family [96]. Interestingly, Hedgehog signaling in CSCs is noncanonical PTCH1-dependent, which is a positive regulator of WNT signaling, whereas canonical SMO-dependent Hedgehog signaling is involved in the downregulation of WNT signaling in normal and differentiated cancer cells through nuclear localization of GLI1 [97]. Noncanonical Hh signaling has been classified into two types: (1) SMO-independent and exclusively through PTCH, and (2) SMO-downstream, mediated by activation of small GTPases, but independent of GLI transcription factors. The SMO-independent GLI activation pathway has additionally been shown to be noncanonical Hh signaling [98,99].
The SMO-independent noncanonical signaling pathway, which operates via PTCH and three Hh proteins (SHH, IHH, and DHH), can inhibit the activation of caspase-3 and promote cell survival through inactivating the PTCH1 pro-apoptotic activity in an SMO-independent manner [100]. Surprisingly, it is indicated that the autocrine Shh–Ptch–Smo signaling is not required in pancreatic ductal cells for the progression of pancreatic ductal adenocarcinoma (PDAC), as the deletion of SMO in the pancreas does not affect the multistage development of PDAC tumors [98]; instead, GLI1 is regulated by TGF-β and KRAS, and Gli–NF-κB oncogenic activation is required for the Kras-dependent transformation of cultured PDAC cancer cells [101]. Besides this, Smo- and Gli-independent Hedgehog also participate in the regulation of the cell cycle through Cyclin B1 and G-protein receptor kinase-2 (GRK2) [102].
In the noncanonical signaling pathway, which works through functions or activities of SMO, it has been indicated that via the rapid stimulation of Rac1 and RhoA small GTPases by SMO, heterotrimeric Gi Proteins are required for SHH-induced fibroblast migration in human endothelial cells [103]. Additionally, cholangiocarcinoma cells exhibit a GLI- and cilia-independent noncanonical Hh signaling pathway, which contributes to mammalian tumor cell engraftment and chemotaxis, including cytoskeleton remodeling and cell migration [104]. Moreover, arachidonic acid (ARA) metabolism through the lipoxygenase pathway plays an important role in Gli-independent Hh signaling in fibroblast migration [105] and neuronal development, particularly in the synthesis of leukotrienes [106]. In addition, the noncanonical Hh signaling pathway is also involved in controlling axon guidance in a Smo-dependent manner by inducing phosphorylation and activation of Src family kinases (SFKs) to alter axon trajectories [107].

5. Types of Aberrant Activation of Hedgehog Signaling in Diseases and Cancers

Accumulating evidence suggests that aberrant activation of the Hh signaling pathway by deregulation of any component within may be involved in the development and progression of cancers and diseases. In various cancer types, three types of proposed mechanisms of uncontrolled activation of Hh signaling are explained below, as summarized in Table 1.

5.1. Type I—Ligand Independent

The first evidence of a link between aberrant Hh signaling and cancer is the rare condition Gorlin syndrome (also known as Naevoid Basal Cell Carcinoma Syndrome or Basal Cell Naevus Syndrome), which is caused by the activating mutation in the Patched gene [108]. Gorlin syndrome is characterized by developmental abnormalities and a distinct postnatal occurrence of cancers that are also known to be caused by the aberrant activation of Hh signaling [66,109]. Cancers in patients with Gorlin syndrome include basal cell carcinomas (BCCs) [66,110], the most common cancer in the Western world; medulloblastoma (MB) [111,112], the most frequent malignant pediatric brain tumor; and rhabdomyosarcoma (RMS), the most common type of soft tissue cancer in children [113].
The tumorigenesis is ligand independent among all these cases, in which the pathway is constitutively activated in the absence of the ligand through mutations in components of the Hh pathway, including activating mutations in the SMO and inactivating mutations in the PTCH1 or Sufu. In Gorlin syndrome, a PTCH1 mutation on chromosome 9 was found [114]. Interestingly, either mutational inactivation of PTCH1 (approximately 30% of cases) or activating mutations of SMO are identified in sporadic BCCs [65,114,115]. In addition, children with MB carry inactivation mutations in PTCH1 or Sufu [112,116,117]. Mutations in Sufu are also found in sporadic BCC, although rarely [118]. Moreover, GLI2 amplifications are found in children (older than 3 years old) with MB as well [119]. Similar to MB, inactivation mutations in either PTCH1 or Sufu are involved in the development of RMS [113]. Apart from these diseases, loss of heterozygosity or somatic mutations in the PTCH1 gene are observed in invasive transitional cell carcinoma of the bladder [120], esophageal squamous cell carcinoma [121], and trichoepitheliomas [122].
These observations have been confirmed by experiments in a variety of preclinical and clinical models. As heritable mutations in BCNS patients and a somatic mutation in a sporadic BCC were found in the Ptch1 gene [66], Ptch1 heterozygous knockout mice developed BCC after being induced with ultraviolet and ionizing radiation or through an inducible Keratin6 promoter [123,124]. In addition, BCC-like skin abnormalities were identified in a Smo constitutively overexpressing skin-specific transgenic mouse model, as activating somatic missense mutations in the Smo gene were found in sporadic BCCs [115]. Moreover, mice carrying heterozygous loss-of-function mutations in the Sufu gene also develop a skin phenotype similar to that in Gorlin syndrome such as basaloid changes and jaw keratocysts [125].

5.2. Type II—Ligand-Dependent Autocrine/Juxtacrine Signaling

Besides activation via the mutations in components of the Hh pathway without ligands, Hh signaling could also be ligand-dependent autocrine/juxtacrine-activated, in which the Hh ligand is profusely released and taken up, activating the same or surrounding tumor cells. In addition to the overexpression of the Hh ligand, other tumors display a high level of PTCH1 and GLI expression.
As the Hh ligand is known to be widely expressed in the foregut endoderm and involved in the regulation of branching morphogenesis in the mammalian lung [126], Hh signaling is proven to participate in the malignant phenotypes of airway epithelial progenitors and small-cell lung tumors, and the first Hh-overexpressing tumor was identified [27]. The activation of the Hh signaling pathway in a ligand-dependent manner was later observed in a variety of different cancers, including digestive tract [127], colorectal [128], prostate [129], liver [130], breast [131], ovarian [132], and brain [133] cancer and melanoma [134]. To prove the autocrine mechanism of the Hh signaling pathway, an Hh-neutralizing antibody or exogenously added Hh ligand was used [127]. Moreover, cyclopamine, a Hedgehog pathway inhibitor, also enhanced the apoptosis and suppressed the proliferation of tumor cells as well as the growth of tumor xenografts after treatment, which strengthened the hypothesis that the Hh pathway could be activated via Hh ligands released by the same or surrounding tumor cells [128,135,136].

5.3. Type III—Ligand-Dependent Paracrine Signaling

In addition to autocrine/juxtacrine activation of the Hh signaling pathway, the Hh pathway can be activated through a paracrine manner in stromal cells as well. In this type, Hh ligands released by cancer cells will bind to the PTCH1 receptor and switch on the Hh signaling in stromal cells. These stromal cells will secrete paracrine growth signals such as vascular endothelial growth factor (VEGF), insulin-like growth factor (IGF), interleukin-6 (IL-6), Wnt, Platelet-derived growth factor (PDGF), and Bone morphogenetic proteins (BMP) to induce the growth of the tumor [12]. The paracrine activation of the Hh pathway has been shown in several cancers such as prostate [137], pancreatic [138], and colorectal cancers and in subsets of esophageal cancers [139]. For instance, in prostate cancer specimens, the expression of SHH is also detected in the tumor epithelium while GLI1 expression is found in the tumor stromal cells, which suggests that Hh signaling is induced in stroma cells by tumor cells to induce the growth of the tumor through secreting paracrine signals [137]. Similarly, the SHH transcript is localized to tumor tissue whereas GLI1 and PTCH1 are detected in both the tumor and the stroma, proving the hypothesis that SHH and its target genes function in a paracrine manner to regulate the development of subsets of esophageal cancers [139].
Moreover, this type of Hh signaling pathway could also be in a reverse paracrine manner, in which the cancer cells take up the Hh ligands released by stromal cells and promote their growth. For example, Hh ligands released by bone-marrow, nodal, and splenic stroma could activate Hh signaling, which plays an important role in the survival of B and plasma cell malignancies [140]. Moreover, the source of the SHH ligand in the activation of Hh signaling pathway for the pathogenesis of multiple myeloma was also found from bone marrow stromal cells [141]. Interestingly, it was also demonstrated that intact SHH-producing microenvironments or neurosphere conditions were required for GLI activation in gliomas [142].

6. Hedgehog Signaling in Cancers and Its Inhibitors

As previously mentioned, while a controlled Hh signaling pathway contributes to numerous biological processes throughout embryonic development of normal tissues and organs as well as to tissue repair and tissue homeostasis, uncontrolled activation of the pathway may promote tumorigenesis [143], such as medulloblastoma, rhabdomyosarcoma, basal cell carcinoma, and lung, colon, stomach, pancreas, ovarian, breast, and malignant hematological cancers [10,12,13,14,15,16,17,18]. Moreover, accumulating evidence suggests that Hedgehog signaling plays a role in the properties of CSCs in several cancers. For instance, the expression of Hedgehog signaling components such as PTCH1, GLI1, and GLI2 is up-regulated in normal human mammary stem/progenitor cells of mammospheres and the Hh signaling pathway is induced in human breast CSCs [144]. Also, the Hh pathway is critical for the maintenance and self-renewal of normal and neoplastic stem cells of the hematopoietic system, including CSCs in myeloid leukemia [145], as well as in solid cancers such as melanoma and pancreatic cancer [146,147]. Some cancer cells surviving after chemotherapeutic treatment such as gemcitabine harbor stem-cell-like properties and display characteristics associated with epithelial–mesenchymal transition (EMT) [148]. Surprisingly, Hh signaling also controls the expression of ABC transporter proteins such as multi-drug resistance protein-1, leading to the chemoresistance of CSCs [149]. Besides this, as CSCs are known to undergo EMT, Hh pathway inhibitors such as cyclopamine and GANT-61 [150] or panobinostat [151] can be used as a potential novel treatment strategy to suppress the EMT process and distant metastasis in cancers. Interestingly, recent evidence has indicated that Hh signaling plays an essential role in tumor angiogenesis through VEGF-A induction in stromal perivascular cells [152], particularly in triple-negative breast cancer [153]. Thus, many inhibitors of the Hh pathway have been developed and tried clinically with promising results (Table 2 and Table 3). However, emerging data indicate that cancer cells may also develop resistance to Hh inhibitors [154,155,156,157,158,159]. For example, in medulloblastoma and BCC, treatment with vismodegib (GDC-0449) and sonidegib (LDE-225), two drugs that have shown significant results in clinical trials, caused acquired drug resistance in the residing cells of these cancers [154,155]. Using genomic analysis, Sharpe et al. indicated that acquired-drug-resistant cells employed SMO mutations in BCC, which further drove drug resistance to SMO inhibitors [155]. The SMO-D4738H mutation was found in 42.5% of BCC patients that acquired drug resistance [156]. This mutation blocked the binding of vismodegib and sonigedib to the mutant SMO protein [156]. While the new generation of SMO inhibitors is undergoing clinical trials, a recent study by Li et al. demonstrated that benzimidazole derivates named HH-1, HH-13, and HH-20 are potent novel SMO inhibitors [160,161,162,163]. Moreover, HH-13 and HH-20 showed prompt inhibition of acquired-drug-resistance SMO-D37738H cells as well [163]. In this section, we discuss the importance of Hh signaling in several cancers, including how Hh signaling affects the survival, EMT, metastasis, and acquired drug resistance of cancer cells and CSC expansion. Also, we discuss the development of the Hh inhibitors that are being clinically trialed to eradicate bulk tumors and CSCs, as summarized in Table 2 and Table 3.

6.1. Basal Cell Carcinoma (BCC)

Basal cell carcinoma (BCC) is the first group of cancers where the oncogenic potential of deregulated Hh signaling was identified. The pioneer study by Hanh et al. and Jonhson et al. indicated that patients with Gorlin’s syndrome had a strong tendency to develop BCC [66,108]. It is believed that the upregulation of Hh signaling is the ace and driver of all BCC malignancies. Hh signaling in the BCC belongs to the ligand-independent type I Hh signaling [66]. About 90% of BCC patients have an identifiable loss-of-function mutation in at least one allele of PTCH1, and the remaining 10% have activating gain-of-function mutations in SMO [115]. In line with these findings, an in vivo study using various models of BCC showed that dysregulation of the Hh signaling pathway mediated uncontrolled proliferation of basal cells [115,124,186]. Besides its function in the promotion of tumorigenesis, Hh signaling has also been reported to be involved in the BCC tumor microenvironment, especially cancer immunity [187]. A study by Otsuka et al. showed that Hh signaling inhibition led to enhanced adaptive immune responses in BCC [187]. Hh components were known to be highly expressed in the thymus, and its upregulation led to the inhibition of T cell activation [188,189,190]. IL-4 upregulation in the tumor microenvironment was reported to enhance tumorigenesis as well as inhibit antitumor response [191,192]. Furmanski et al. also showed that IL-4 is a transcriptional target of Hh signaling in T cells [193]. In addition, Hh signaling inhibition was also shown to increase interferon-gamma (IFNγ) expression [193,194]. Taken together, this suggests that targeting Hh signaling in BCC would increase the effectiveness of cancer therapy by modulating the antitumor activity of the immune cells.
Cyclopamine, an Hh signaling inhibitor, is the first drug that has been used to treat BCC [195]. Since then, several other synthetic cyclopamine derivatives have continuously been developed as Hh pathway inhibitors, with better pharmacological and inhibitory properties than those of cyclopamine. Cur-61414, one of the synthetic SMO inhibitors, potentially prevented the formation of BCC-like cells and BCC-like lesions without affecting surrounding normal cells [17,196]. Because of its safety, Cur-61414 was considered for preclinical models and was the first class of Hh antagonist to enter phase I clinical trials for BCC patient treatment [196]. However, it failed to either meet the patient’s response or reduce GLI1 expression [196]. Vismodegib (GDC-0449) was then developed as a second SMO inhibitor with a better ability to inhibit BCC progression and with more favorable pharmaceutical properties than cyclopamine. Vismodegib showed great antitumor activity in preclinical models [158,197,198]. A phase I study of vismodegib that demonstrated antitumor activity in patients with BCC and medulloblastoma was conducted in 2009 [158,197]. Based on these promising results, vismodegib has recently entered phase II trials in advanced BCC. Another SMO inhibitor that also entered clinical trials is sonidegib (LDE225), which is used for treating recurring BCC after surgery and radiation therapy [197]. In addition, saridegib (IPI-926) is another SMO inhibitor that has tolerability in advanced solid tumors, including BCC [199]. Despite the successes of these drugs, it is likely that many tumors acquire clinical resistance during therapy [200].
CD200+CD45 cells have been identified as a CSC population in BCC. This population was resistant to chemotherapy and expressed a high level of multidrug resistance protein 1 (MDR1), whose expression gradually increased upon drug exposure. CD200+ BCC also expressed a high level of GLI1 and relied on GLI1 for their survival, but not GLI2 [164,201]. In an in vivo study, Colmont et al. found that CD200+CD45 cells were able to initiate BCC tumor growth with identical histology to the parental cells and showed the upregulation of SHH signaling genes. Inhibiting Hh signaling by Smo antagonists was successful in eradicating BCC CSCs. Thus, this suggested a hypothesis that dual therapy using available anti CD200+ neutralizing antibody in combination with Smo antagonists would be a novel strategy for a complete inhibition of BCC progression [164].

6.2. Colon Cancer

The Hedgehog (Hh) signaling pathway is essential for cell growth as well as the development of gastrointestinal tracts [202]. Accumulating evidence suggests that Hh signaling plays important roles in the development of colon cancer [203,204]. Hh signaling in the CRC belongs to the ligand-dependent type II Hh signaling [128]. Even though each colorectal cancer (CRC) has a different pattern of Hh signaling expression, most of them highly express the Hh mRNA or protein [49]. Moreover, the expression levels of PTCH and SMO were gradually increased as the colon cancer progressed from a benign to more malignant tumor [203]. In addition, other components of Hh signaling, such as SHH and GLI1, were also reported as being upregulated in colon cancer [205]. Dysregulation of Hh signaling contributed to gene mutation, metastasis, and angiogenesis in colon cancer [206], and termination of Hh signaling through GLI1 inhibition resulted in the inhibition of proliferation in colon cancer [207,208]. Considering the importance of Hh signaling in the progression of colon cancer, cyclopamine appeared to inhibit the Hh signal and suppressed the survival and proliferation of colon cancer. Cyclopamine treatment resulted in the inhibition of Hh, SMO, and PTCH mRNA, which were highly expressed in the colon cancer cell lines [209]. In addition, cyclopamine inhibited E-cadherin expression in benign or malignant colorectal tumor cells, and reduced their invasive capacity [210]. Vismodegib (GDC-0449), which has been approved for treating BCC, was also utilized for Hh-targeted therapy for colon cancer patients [12,25]. However, drug resistance to vismodegib was acquired over time. In addition, side effects were observed in the patients that underwent vismodegib treatment [25,155,158]. Therefore, a recent study by Chen et al. suggested that Hh003, an SMO antagonist with a new scaffold, induced a greater effect on the elimination of human colon and pancreatic cancer compared with vismodegib. Hh003 exerted stronger antitumor effects compared to vismodegib by activating caspase8-dependent apoptosis [211].
It has been reported that Hh signaling interacts with other signaling pathways, such as Wnt/b-catenin and PI3K pathways in several cancers, including in colon cancer [212]. Dual targeting of Wnt/b-catenin and Hh-GLI1 signaling resulted in a better elimination of colon cancer cells compared with inhibition of GLI1 alone [97,212]. In addition, dual targeting of GLI1 by arsenic trioxide and PI3K inhibitor LY294002 synergistically downregulated both the GLI1 protein level and GLI1 target genes BCL2 and CCND1, which resulted in decreased proliferarion of colon cancer cells [213].
CD133+ was identified as a marker of colon CSCs, and Hh-GLI1 signaling was also reported to play important roles in colon CSC survival and expansion [128]. Treatment with cyclopamine showed a reduction in colon cancer progression and metastasis, and inhibition of Hh signaling through GLI1-targeted shRNA consistently displayed dramatically lower colony forming ability in colon CSCs [128]. A study by Batsaikhan et al. showed that the expression of EMT markers and stem cell markers was higher in the colon cancer spheres than in the parental cell line. Treatment of colon CSCs with cyclopamine decreased the expression of SHH and its downstream targets in a dose-dependent manner [214]. Noncanonical PTCH-1-dependent Hh signaling is required for the survival of colon CSCs, especially by its regulation in the activation of the Wnt/b-catenin signaling pathway. It acts as a positive regulator of Wnt/b-catenin signaling to maintain colon CSCs in a quiescent state [97,212]. Treatment of colon CSCs with PTCH1 inhibitor RU-SKI 43 significantly decreased Wnt activity, whereas vismodegib treatment failed to achieve the same outcome [97].

6.3. Breast Cancer

Aberrant activation of Hh signaling has been observed in the several types of breast cancers. As we discussed earlier, Hh signaling alteration in cancer arises due to reasons such as overexpression of Hh ligands, which results in the activation of either autocrine or paracrine signaling, loss of function of the receptor, somatic mutation of Hh components such as GLI1 and PTCH1, and the overexpression of the Hh components [215]. In breast cancer, Hh signaling is dependent on the autocrine manner of the Hh ligands of the tumor cells (type II Hh signaling), which in turn acts as a self-activating signal for the Hh amplification [131]. The receptor loss of function normally occurs in PTCH receptors, whereas gain of function was shown mostly in SMO receptors [216,217]. SMO and GLI1 were highly expressed in triple negative breast cancer (TNBC), and their excessive expression was correlated with metastasis, poor prognosis, and recurrence of TNBC [218]. Moreover, in breast cancer, Hh signaling has been shown to induce angiogenesis independently of VEGF activation [219]. Recently, a comprehensive immunohistochemistry (IHC) study by Kurebayashi et al. showed that SHH expression was positively correlated with tumor size, Ki-67 labeling index, and HER2 positivity, but negatively correlated with estrogen receptor (ER+) and progesterone receptor (PR+) breast cancer [220]. In addition, a GLI1 isoform called truncated GLI1 (tGLI1) has also been reported to induce the activation of genes related to proliferation, migration, and angiogenesis of breast cancer. Even though the tGLI1 mechanism of action was different from that of GLI1, it functioned similarly in promoting breast cancer growth [197].
Hh inhibitor sonidegib was used in the treatment of several cancers and has been used clinically to treat breast cancer patients, either independently or in a combination with docetaxel [221]. Cyclopamine treatment inhibited PTCH1 expression and cell cycle progression in breast cancer cells [222]. GLI1 inhibitors such as pirfenidone and imiquimod have been reported to have promising antitumor activity in breast cancer [223,224]. Topical application of imiquimod was well tolerated and decreased the cancer progression [224]. GANT61 is a small GLI1 antagonist that was also reported to have antitumor activity in breast cancer. GANTS61 had high specificity to reduce GLI1 expression and blocked the binding of GLI to the target DNA sequences [225]. Breast cancer treatment with GDC-0449 and LDE225 SMO inhibitors has also been studied in clinical trials [197].
CD44+/CD24/Lin is a subpopulation of breast CSCs that has the ability to produce large numbers of tumors having self-renewal function. CSCs expressing CD44+/CD24/Lin have increased PTCH1, GLI1, and GLI2 mRNA expression [144,226]. P63, a sister homolog of p53, which has been reported to interact with Hh signaling, is also highly expressed in breast CSCs compared to normal cells [216]. In addition, GLI1 is highly expressed in breast CSCs, and a study in an orthotopic xenografts model showed that low expression of GLI1 reduces tumor growth, self-renewal, motility, and viability of breast CSCs [227,228]. Hh signaling inactivation in breast CSCs downregulates the representative stem cell markers OCT4, NESTIN, and NANOG. Through in vitro study, cyclopamine showed the capacity to inhibit mammosphere formation of mammary gland stem cells [144,165].

6.4. Pancreatic Cancer

Pancreatic ductal adenocarcinoma (PDAC) is thought to be one of the leading causes of cancer death with 5-year survival rates of 1 to 5% [229]. It has been reported that uncontrolled activation of Hh signaling is efficient in maintaining tumor growth and progression of the pancreatic tumor [230]. Using Pdx–Shh mice, in which sonic hedgehog (Shh) overexpression was driven by the pancreatic-specific Pdx1 promoter, a study by Thayer et al. showed that mouse pancreas developed abnormal tubular structures similar to the phenotype of human pancreatic intraepithelial neoplasia (PanIN-1 and -2) [135]. Not only genetic mutation but also epigenetic modification is involved in pancreatic cancer tumorigenesis. Hedgehog-interacting protein (HHIP), a negative regulator of Hh signaling which is normally downregulated or absent in pancreatic cancer, was considered to be the target of genetic and epigenetic modifications. The HHIP promoter underwent partial and complete methylation in several pancreatic cancer cell lines, primary pancreatic cancers, and pancreatic cancer xenografts, whereas no methylation was found in normal pancreata. This suggests that methylation of the HHIP promoter contributes to the upregulated Hedgehog signaling in pancreatic neoplasms [231].
Hh signaling in pancreatic cancer belongs to type III ligand-dependent signaling [98]. Among the components of Hh signaling, it has been reported that deletion of SMO, a component of SHH–PTCH1–SMO autocrine signaling, did not affect multistage tumor development of PDAC [98]. Furthermore, a study by Yauch et al. showed a ligand-dependent activation of the Hh pathway in the stromal microenvironment of pancreatic cancer [198]. They showed that inhibition of the Hh pathway using a Hh pathway antagonist downregulated the canonical Hh target genes in the stromal microenvironment and inhibited pancreatic tumor growth in xenograft models [198]. Taken together, this suggests that the paracrine ligand for Hh signaling but not autocrine signaling is required for PDAC tumor progression in pancreatic ductal cells [98].
Various studies have shown the ability of cyclopamine to induce apoptosis in a variety of tumor cell lines and to inhibit pancreatic cancer [135,232]. Cyclopamine not only induced apoptosis and inhibited proliferation in pancreatic cancer cell lines both in vitro and in vivo, but also inhibited systemic metastases in spontaneously metastatic orthotopic xenograft models [233]. Similarly, IPI-269609, a Hh-targeted small molecule, had a similar effect to that of cyclopamine in inhibiting pancreatic cancer progression [135,232]. It has been shown that Hh signaling inhibition by cyclopamine significantly extended median survival in the transgenic mouse model [233]. Hh inhibition by cyclopamine led to the down-regulation of SNAIL, up-regulation of E-CADHERIN, and inhibition of the metastasis capability of pancreatic cancer [232]. Indeed, a combination of gemcitabine and cyclopamine showed a complete suppression and significant reduction in the primary tumor size [232]. However, the follow-up to the clinical trials did not show a significant effect. On the other hand, a phase II clinical trial of a IPI-926–gemcitabine combination was closed early because of the side effects of the treatment [234]. The combination of vismodegib with gemcitabine did not improve the response rate and overall survival of patients with metastatic pancreatic cancer. Also, preclinical and clinical results reported by Catenacci et al. showed that the combination treatment did not have a significantly different outcome from that of vismodegib treatment alone [235].
In pancreatic cancer, CD44+CD24+ESA+ cells were identified as a population of pancreatic CSC [232]. Hh signaling is known to be a major regulator of pancreatic CSC self-renewal [169]. Kim et al. showed that even though GDC-0449 led to a downregulation of GLI1 and PTCH1, it did not show any significant differences in the elimination of pancreatic CSCs. Moreover, GDC-0449 and gemcitabine combination therapy did not significantly abrogate the metastasis of pancreatic cancer more efficiently than gemcitabine alone. Therefore, development of a pancreatic CSC-targeted Hh inhibitor still needs to be explored [236]. Li et al. found that treatment with sulforaphane significantly reduced tumor growth and self-renewal of pancreatic CSCs both in vitro and in orthotopically implanted human-derived primary pancreatic CSCs [169]. Inhibition of Hh–GLI1 signaling by GANT-61 (GLI1 antagonist) showed an inhibition of pancreatic CSC growth in both in vitro and in vivo models [166]. Baicalein, an active compound in the QYHJ formula, also showed a great ability to impair pancreatic CSC self-renewal through the inhibition of the SHH signaling pathway [171]. Sanguinarine is another pancreatic-CSC-targeted drug whose usefulness has been challenged. Sanguinarine inhibits the SHH–GLI1 pathway, resulting in the downregulation of GLI1 target genes in pancreatic CSCs. It inhibits the proliferation and self-renewal of pancreatic CSCs by inducing oxidative damage, leading to cell apoptosis [170]. Gemcitabine-resistant pancreatic cancer cells displayed high expression of Hh signaling components as well as CSC markers [172]. Treatment of these resistant cells with cyclopamine reverted the acquired chemoresistance and decreased the expression of CSC markers [168,172]. In addition, treatment with cyclopamine derivate (CyT) along with focal irradiation showed an additive effect on tumor sphere formation in vitro, as well as inhibition of lymph node metastasis in an in vivo model [167]. Taken together, targeting Hh signaling in pancreatic CSCs showed a better outcome in the ablation of pancreatic cancer cells.

6.5. Medulloblastoma

Medulloblastoma, an aggressive childhood tumor of cerebellar origin, is a malignancy exhibiting a well-recognized reliance on aberrant Hh signaling. The Hh pathway of medulloblastoma belongs to the ligand-independent type I Hh signaling [17]. Abnormal Hh signaling and mutations in PTCH causing hyperactivation of Hh signaling have been observed in medulloblastoma [112,117,237]. In murine medulloblastoma cells, cyclopamine treatment suppressed proliferation in vitro and caused loss of neuronal stem-cell-like characteristics [238]. Also, itraconazole, a commonly used antifungal agent that works via inhibiting Hh signaling, was reported to inhibit the growth of medulloblastoma in a mouse allograft model, specifically through the inhibition of SMO [239]. Buonamici et al. reported that combination treatment of sonidegib with either PI3K inhibitor (NVP-BKM120) or the dual PI3K/mTOR inhibitor (NVP-BEZ235) significantly delayed medulloblastoma relapse [240]. Meanwhile, even though SMO inhibitor vismodegib (GDC-0449) was reported to have antitumor potency in early clinical studies of medulloblastoma, relapse appeared after treatment [154]. Indeed, the efficacy of vismodegib in combination with other drugs in treating medulloblastoma still remains unclear.
CD15 has been identified as a marker for medulloblastoma CSCs. Medulloblastoma with CD15+ cells expressed higher levels of Shh target genes, such as GLI1 and CYCLIN-D1, than that with CD15 cells [174,175]. The transcription factors GLI1/2 of the Hh pathway have been indicated to interact with stem-related factor NANOG. They bind to the specific cis-regulatory sequences of NANOG in both mouse and human stem cells [173]. When the Hh pathway was blocked by KAAD–cyclopamine, the expression of GLI1 and NANOG was reduced, and the self-renewal of medulloblastoma neurospheres was also significantly inhibited [173].

6.6. Leukemia

Hh pathway activity has a critical role in the maintenance of normal and neoplastic stem cells in the hematopoietic system and correlates with the chemotherapy-resistant phenotype of myeloid leukemia cells and disease recurrence [145,176,178]. However, Hh signaling has been reported to be less pronounced in chemosensitive cells [178]. In cancer with paracrine Hh signaling, such as pancreatic cancer, tumor cells secrete Hh ligands which then are received by remote cells in the stroma [198]. However, in leukemia, B-cell lymphomas, and multiple myelomas, the reverse paracrine signaling is reported. In this model, the Hh ligands are produced by the bone marrow stromal cells and are then received by the tumor cells [241]. These ligands support the proliferation and growth of surrounding cancerous tissue through upregulating the anti-apoptotic gene Bcl2 [241].
In a chronic myelogenous leukemia (CML) study, Chen et al. reported that a loss of Smoothened (SMO) damaged the renewal of hematopoietic stem cells and impaired induction of CML by the BCR-ABL1 oncoprotein [145]. Moreover, Sonic hedgehog (SHH), smoothened (SMO), and GLI1 are significantly increased in CML patients compared with in normal people, suggesting that activation of the Hh pathway is a critical inducer of CML progression [242]. Hh signaling was also found to be implicated in multidrug resistance in CML. Treatment of cyclopamine along with nilotinib, a tyrosine kinase inhibitor (TKI), resulted in a re-sensitization to chemotherapy and inhibited CML progression [178]. Similar to solid cancer, leukemia cells also contain a subpopulation of leukemia-initiating cells or leukemic stem cells (LSCs) that have been reported to have the capability to self-renew and drive hematopoietic malignancies [145,243]. Hh signaling was activated through upregulation of SMO in LSCs and pharmacological inhibition of SMO reduced LSCs in vivo and delayed relapse after the end of treatment [176]. Indeed, loss of SMO causes depletion of CML stem cells while the activation of SMO supplements CML stem cell number and accelerates the disease without an impairment of normal hematopoiesis [145,176]. Additionally, GLI1, a downstream target of Hh signaling, was significantly upregulated in CD34+ chronic phase (CP) CML cells compared to in normal CD34+ hematopoietic cells [145]. Tyrosine kinase inhibitors (TKIs) such as imatinib, nilotinib, and dasatinib have been shown to have a dramatic effect on the eradication of chronic-phase (CP) CML. However, quiescent CML stem cells were not effectively targeted by TKIs. Drugs targeting both bulk progenitor and stem cell populations have been investigated for several decades. Irvine et al. showed that a combination of Hh inhibitor sonidegib (LDE225) and nilotinib succeeded in eradicating both CML LSCs and progenitor cells [177]. Cyclopamine and nilotinib, as well as an SMO antagonist (PF-04449913) and dasatinib, also showed effective elimination of CML LSCs [176,179]. These findings show that the combination of Hh inhibitors and TKIs reduces LSC self-renewal and clinically improves survival [177].
In acute myeloid leukemia (AML), activation of Hh signaling has been shown in several human AML cells, especially primary CD34+ leukemic cells [181]. Not only expression levels but also epigenetic modifications of Hh signaling components have been observed to correlate with disease status in AML patients [244]. A high level of GLI1 expression resulted in poor prognosis for AML patients [245]. Moreover, Hh pathway activation increased both the survival and drug resistance capability of CD34+ leukemia cells. Inhibition of Hh signaling with Smoothened antagonist cyclopamine, endogenous Hh inhibitor Hh-interacting protein, or anti-Hh neutralizing antibody induced apoptosis of CD34+ leukemic cells, although these CD34+ cell lines have a resistance to cytarabine (Ara-C) [181]. Furthermore, the combination of cyclopamine and Ara-C significantly reduced the drug resistance of CD34+ leukemic cells. Ara-C treatment in combination with cyclopamine significantly enhanced the sensitivity to Ara-C, which resulted in the complete abrogation of bulk leukemia cells [181]. PF-04449913 treatment modulated the cell cycle regulation and self-renewal signaling, which in turn reduced the leukemia initiation potential of AML cells and enhanced sensitivity to chemotherapy, overcoming the resistance in the bone marrow microenvironment [182]. Recently, Kakiuchi et al. reported that a novel SMO inhibitor, PF-913, impaired the self-renewal signature and cell cycle regulation of leukemic-stem-cell-like properties from AML patients, suggesting that PF-913 induced cell cycling in quiescent leukemia stem cells [180].
In lymphoblastic leukemia, Hh pathway components are upregulated in precursor B-ALL, and SMO inhibitor has been reported to reduce self-renewal of ALL cells in vitro and in vivo [173,184]. Moreover, mutations of the Hh pathway have also been described in T-ALL [246,247,248,249]. PTCH1 mutations were the most common Hh signaling found in T-ALL patients [246]. A study using the zebrafish model indicated that PTCH1 mutations contributing to chemotherapy resistance in human T-ALL were driving mutations responsible for oncogenic transformation in T-ALL malignancy [246]. Treatment of SANT-1 or KAAD cyclopamine was reported to induce the cell death of T-ALL cells as well as to re-sensitize the cells to chemotherapy [183]. Treatment of cyclopamine or IPI-926 was shown to inhibit the expansion of B-ALL LSCs by reducing the capability of self-renewal [173,184]. In chronic lymphocytic leukemia (CLL), the expression of Hh signaling molecules, such as GLI1, GLI2, BCL2, and SUFU, was significantly upregulated and correlated with B-CLL progression [241]. Furthermore, cyclopamine treatment inhibited bone marrow stromal cells, which resulted in a lower level of survival of B-CLL cells, indicating an important role of Hh signaling in the survival of B-CLL cells [241].

7. Conclusions

The Hh pathway is evolutionarily conserved and essential for the regulation of normal development and differentiation in invertebrates and vertebrates. However, aberrant Hh pathway activation by either mutation or ligand overexpression is closely correlated with many types of human cancers. Thus, targeting the Hh pathway may provide therapeutic options for the treatment of numerous types of human cancers. Effective inhibition of the Hh signaling pathway can be obtained through several approaches, such as using anti-Hh antibodies to inhibit receptor activation or small-molecule antagonists that inhibit downstream Hh signaling, such as SMO and GLI1. Synthetic Hh antagonists are now available, and some are undergoing clinical evaluation (Table 3). In this review, we have discussed the importance of Hh signaling in several types of cancers, such as basal cell carcinoma (BCC), leukemia, and medulloblastoma, and in colon, breast, and pancreatic cancer. The contributions of Hh signaling to cancer progression are summarized in Figure 2. Several agents targeting the Hh signaling pathway to eradicate CSCs in the same cancer have been proposed to have anticancer activity either as stand-alone drugs or in combination with standard chemotherapy drugs, and some of them have entered clinical trials as novel treatment strategies to regulate CSC replication, survival, and differentiation. However, not many of them have been successful in the total eradication of CSCs. Moreover, the toxicity associated with these drugs is a major impediment to their applicability. Therefore, considering the substantial contributions of Hh signaling to tumorigenesis and drug resistance, understanding the fundamental Hh pathway regulation in a precise manner will help in the development of new therapeutics to tackle Hh-pathway-activated cancers and achieve better therapeutic outcomes for cancer therapy.

Funding

This work was supported by the Soonchunhyang University Research Fund and Basic Science Research Program through the National Research Foundation of Korea (NRF-2017R1A2B4001996).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Varjosalo, M.; Taipale, J. Hedgehog: Functions and mechanisms. Genes Dev. 2008, 22, 2454–2472. [Google Scholar] [CrossRef] [PubMed]
  2. Wilson, C.W.; Chuang, P.T. Mechanism and evolution of cytosolic Hedgehog signal transduction. Development 2010, 137, 2079–2094. [Google Scholar] [CrossRef] [PubMed]
  3. Briscoe, J.; Therond, 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]
  4. Jia, Y.; Wang, Y.; Xie, J. The Hedgehog pathway: Role in cell differentiation, polarity and proliferation. Arch. Toxicol. 2015, 89, 179–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Robbins, D.J.; Fei, D.L.; Riobo, N.A. The Hedgehog signal transduction network. Sci. Signal. 2012, 5, re6. [Google Scholar] [CrossRef] [PubMed]
  6. Haas, D.; Muenke, M. Abnormal sterol metabolism in holoprosencephaly. Am. J. Med. Genet. C Semin Med. Genet. 2010, 154C, 102–108. [Google Scholar] [CrossRef] [PubMed]
  7. Hill, R.E.; Heaney, S.J.; Lettice, L.A. Sonic hedgehog: Restricted expression and limb dysmorphologies. J. Anat. 2003, 202, 13–20. [Google Scholar] [CrossRef] [PubMed]
  8. 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] [PubMed]
  9. Muenke, M.; Beachy, P.A. Genetics of ventral forebrain development and holoprosencephaly. Curr. Opin. Genet. Dev. 2000, 10, 262–269. [Google Scholar] [CrossRef] [Green Version]
  10. Tostar, U.; Toftgard, R.; Zaphiropoulos, P.G.; Shimokawa, T. Reduction of human embryonal rhabdomyosarcoma tumor growth by inhibition of the hedgehog signaling pathway. Genes Cancer 2010, 1, 941–951. [Google Scholar] [CrossRef] [PubMed]
  11. Zhang, L.; Chen, X.M.; Sun, Z.J.; Bian, Z.; Fan, M.W.; Chen, Z. Epithelial expression of SHH signaling pathway in odontogenic tumors. Oral. Oncol. 2006, 42, 398–408. [Google Scholar] [CrossRef] [PubMed]
  12. Amakye, D.; Jagani, Z.; Dorsch, M. Unraveling the therapeutic potential of the Hedgehog pathway in cancer. Nat. Med. 2013, 19, 1410–1422. [Google Scholar] [CrossRef] [PubMed]
  13. Hui, M.; Cazet, A.; Nair, R.; Watkins, D.N.; O’Toole, S.A.; Swarbrick, A. The Hedgehog signalling pathway in breast development, carcinogenesis and cancer therapy. Breast Cancer Res. 2013, 15, 203. [Google Scholar] [CrossRef] [PubMed]
  14. Liao, X.; Siu, M.K.; Au, C.W.; Wong, E.S.; Chan, H.Y.; Ip, P.P.; Ngan, H.Y.; Cheung, A.N. Aberrant activation of hedgehog signaling pathway in ovarian cancers: Effect on prognosis, cell invasion and differentiation. Carcinogenesis 2009, 30, 131–140. [Google Scholar] [CrossRef] [PubMed]
  15. Ok, C.Y.; Singh, R.R.; Vega, F. Aberrant activation of the hedgehog signaling pathway in malignant hematological neoplasms. Am. J. Pathol. 2012, 180, 2–11. [Google Scholar] [CrossRef] [PubMed]
  16. 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]
  17. 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]
  18. Teglund, S.; Toftgard, R. Hedgehog beyond medulloblastoma and basal cell carcinoma. Biochim. Biophys. Acta 2010, 1805, 181–208. [Google Scholar] [CrossRef] [PubMed]
  19. Watkins, D.N.; Berman, D.M.; Burkholder, S.G.; Wang, B.; Beachy, P.A.; Baylin, S.B. Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer. Nature 2003, 422, 313–317. [Google Scholar] [CrossRef] [PubMed]
  20. Zhao, H.; Feng, J.; Seidel, K.; Shi, S.; Klein, O.; Sharpe, P.; Chai, Y. Secretion of shh by a neurovascular bundle niche supports mesenchymal stem cell homeostasis in the adult mouse incisor. Cell Stem Cell 2014, 14, 160–173. [Google Scholar] [CrossRef] [PubMed]
  21. Ochoa, B.; Syn, W.K.; Delgado, I.; Karaca, G.F.; Jung, Y.; Wang, J.; Zubiaga, A.M.; Fresnedo, O.; Omenetti, A.; Zdanowicz, M.; et al. Hedgehog signaling is critical for normal liver regeneration after partial hepatectomy in mice. Hepatology 2010, 51, 1712–1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Karhadkar, S.S.; Bova, G.S.; Abdallah, N.; Dhara, S.; Gardner, D.; Maitra, A.; Isaacs, J.T.; Berman, D.M.; Beachy, P.A. Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature 2004, 431, 707–712. [Google Scholar] [CrossRef] [PubMed]
  23. Shin, K.; Lee, J.; Guo, N.; Kim, J.; Lim, A.; Qu, L.; Mysorekar, I.U.; Beachy, P.A. Hedgehog/Wnt feedback supports regenerative proliferation of epithelial stem cells in bladder. Nature 2011, 472, 110–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Rubin, L.L.; de Sauvage, F.J. Targeting the Hedgehog pathway in cancer. Nat. Rev. Drug Discov. 2006, 5, 1026–1033. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, F.; Zhang, Y.; Sun, B.; McMahon, A.P.; Wang, Y. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem. Biol. 2017, 24, 252–280. [Google Scholar] [CrossRef] [PubMed]
  26. Ingham, P.W.; McMahon, A.P. Hedgehog signaling in animal development: Paradigms and principles. Genes Dev. 2001, 15, 3059–3087. [Google Scholar] [CrossRef] [PubMed]
  27. Mak, K.K.; Bi, Y.; Wan, C.; Chuang, P.T.; Clemens, T.; Young, M.; Yang, Y. Hedgehog signaling in mature osteoblasts regulates bone formation and resorption by controlling PTHrP and RANKL expression. Dev. Cell 2008, 14, 674–688. [Google Scholar] [CrossRef] [PubMed]
  28. Ohba, S.; Kawaguchi, H.; Kugimiya, F.; Ogasawara, T.; Kawamura, N.; Saito, T.; Ikeda, T.; Fujii, K.; Miyajima, T.; Kuramochi, A.; et al. Patched1 haploinsufficiency increases adult bone mass and modulates Gli3 repressor activity. Dev. Cell 2008, 14, 689–699. [Google Scholar] [CrossRef] [PubMed]
  29. 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] [PubMed]
  30. 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] [PubMed] [Green Version]
  31. King, P.J.; Guasti, L.; Laufer, E. Hedgehog signalling in endocrine development and disease. J. Endocrinol. 2008, 198, 439–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Mann, R.K.; Beachy, P.A. Novel lipid modifications of secreted protein signals. Annu. Rev. Biochem. 2004, 73, 891–923. [Google Scholar] [CrossRef] [PubMed]
  33. Porter, J.A.; Young, K.E.; Beachy, P.A. Cholesterol modification of hedgehog signaling proteins in animal development. Science 1996, 274, 255–259. [Google Scholar] [CrossRef] [PubMed]
  34. Buglino, J.A.; Resh, M.D. Hhat is a palmitoylacyltransferase with specificity for N-palmitoylation of Sonic Hedgehog. J. Biol. Chem. 2008, 283, 22076–22088. [Google Scholar] [CrossRef] [PubMed]
  35. Chen, M.H.; Li, Y.J.; Kawakami, T.; Xu, S.M.; Chuang, P.T. Palmitoylation is required for the production of a soluble multimeric Hedgehog protein complex and long-range signaling in vertebrates. Genes Dev. 2004, 18, 641–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Gallet, A.; Ruel, L.; Staccini-Lavenant, L.; Therond, P.P. Cholesterol modification is necessary for controlled planar long-range activity of Hedgehog in Drosophila epithelia. Development 2006, 133, 407–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Goetz, J.A.; Suber, L.M.; Zeng, X.; Robbins, D.J. Sonic Hedgehog as a mediator of long-range signaling. Bioessays 2002, 24, 157–165. [Google Scholar] [CrossRef] [PubMed]
  38. Echelard, Y.; Epstein, D.J.; St-Jacques, B.; Shen, L.; Mohler, J.; McMahon, J.A.; McMahon, A.P. Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity. Cell 1993, 75, 1417–1430. [Google Scholar] [CrossRef]
  39. Weed, M.; Mundlos, S.; Olsen, B.R. The role of sonic hedgehog in vertebrate development. Matrix Biol. 1997, 16, 53–58. [Google Scholar] [CrossRef]
  40. Bellusci, S.; Furuta, Y.; Rush, M.G.; Henderson, R.; Winnier, G.; Hogan, B.L. Involvement of Sonic hedgehog (Shh) in mouse embryonic lung growth and morphogenesis. Development 1997, 124, 53–63. [Google Scholar] [PubMed]
  41. Litingtung, Y.; Lei, L.; Westphal, H.; Chiang, C. Sonic hedgehog is essential to foregut development. Nat. Genet. 1998, 20, 58–61. [Google Scholar] [CrossRef] [PubMed]
  42. Hardcastle, Z.; Mo, R.; Hui, C.C.; Sharpe, P.T. The Shh signalling pathway in tooth development: Defects in Gli2 and Gli3 mutants. Development 1998, 125, 2803–2811. [Google Scholar] [PubMed]
  43. Seppala, M.; Fraser, G.J.; Birjandi, A.A.; Xavier, G.M.; Cobourne, M.T. Sonic Hedgehog Signaling and Development of the Dentition. J. Dev. Biol. 2017, 5, 6. [Google Scholar] [CrossRef] [PubMed]
  44. Schock, E.N.; Brugmann, S.A. Neural crest cells utilize primary cilia to regulate ventral forebrain morphogenesis via Hedgehog-dependent regulation of oriented cell division. Dev. Biol. 2017, 431, 168–178. [Google Scholar] [CrossRef] [PubMed]
  45. St-Jacques, B.; Dassule, H.R.; Karavanova, I.; Botchkarev, V.A.; Li, J.; Danielian, P.S.; McMahon, J.A.; Lewis, P.M.; Paus, R.; McMahon, A.P. Sonic hedgehog signaling is essential for hair development. Curr. Biol. 1998, 8, 1058–1068. [Google Scholar] [CrossRef]
  46. Apelqvist, A.; Ahlgren, U.; Edlund, H. Sonic hedgehog directs specialised mesoderm differentiation in the intestine and pancreas. Curr. Biol. 1997, 7, 801–804. [Google Scholar] [CrossRef]
  47. Van den Brink, G.R.; Hardwick, J.C.; Nielsen, C.; Xu, C.; ten Kate, F.J.; Glickman, J.; van Deventer, S.J.; Roberts, D.J.; Peppelenbosch, M.P. Sonic hedgehog expression correlates with fundic gland differentiation in the adult gastrointestinal tract. Gut 2002, 51, 628–633. [Google Scholar] [CrossRef] [PubMed]
  48. Feng, R.; Aihara, E.; Kenny, S.; Yang, L.; Li, J.; Varro, A.; Montrose, M.H.; Shroyer, N.F.; Wang, T.C.; Shivdasani, R.A.; et al. Indian Hedgehog mediates gastrin-induced proliferation in stomach of adult mice. Gastroenterology 2014, 147, 655–666 e659. [Google Scholar] [CrossRef] [PubMed]
  49. Van den Brink, G.R.; Bleuming, S.A.; Hardwick, J.C.; Schepman, B.L.; Offerhaus, G.J.; Keller, J.J.; Nielsen, C.; Gaffield, W.; van Deventer, S.J.; Roberts, D.J.; et al. Indian Hedgehog is an antagonist of Wnt signaling in colonic epithelial cell differentiation. Nat. Genet. 2004, 36, 277–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. 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] [PubMed]
  51. 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]
  52. 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]
  53. Wang, S.; Yang, K.; Chen, S.; Wang, J.; Du, G.; Fan, S.; Wei, L. Indian hedgehog contributes to human cartilage endplate degeneration. Eur. Spine J. 2015, 24, 1720–1728. [Google Scholar] [CrossRef] [PubMed]
  54. Shah, D.K.; Zuniga-Pflucker, J.C. An overview of the intrathymic intricacies of T cell development. J. Immunol. 2014, 192, 4017–4023. [Google Scholar] [CrossRef] [PubMed]
  55. Solanki, A.; Yanez, D.C.; Ross, S.; Lau, C.I.; Papaioannou, E.; Li, J.; Saldana, J.I.; Crompton, T. Gli3 in fetal thymic epithelial cells promotes thymocyte positive selection and differentiation by repression of Shh. Development 2018, 145, dev146910. [Google Scholar] [CrossRef] [PubMed]
  56. Outram, S.V.; Hager-Theodorides, A.L.; Shah, D.K.; Rowbotham, N.J.; Drakopoulou, E.; Ross, S.E.; Lanske, B.; Dessens, J.T.; Crompton, T. Indian hedgehog (Ihh) both promotes and restricts thymocyte differentiation. Blood 2009, 113, 2217–2228. [Google Scholar] [CrossRef] [PubMed]
  57. Solanki, A.; Lau, C.I.; Saldana, J.I.; Ross, S.; Crompton, T. The transcription factor Gli3 promotes B cell development in fetal liver through repression of Shh. J. Exp. Med. 2017, 214, 2041–2058. [Google Scholar] [CrossRef] [PubMed]
  58. 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]
  59. Lau, C.I.; Outram, S.V.; Saldana, J.I.; Furmanski, A.L.; Dessens, J.T.; Crompton, T. Regulation of murine normal and stress-induced erythropoiesis by Desert Hedgehog. Blood 2012, 119, 4741–4751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Marigo, V.; Davey, R.A.; Zuo, Y.; Cunningham, J.M.; Tabin, C.J. Biochemical evidence that patched is the Hedgehog receptor. Nature 1996, 384, 176–179. [Google Scholar] [CrossRef] [PubMed]
  61. Stone, D.M.; Hynes, M.; Armanini, M.; Swanson, T.A.; Gu, Q.; Johnson, R.L.; Scott, M.P.; Pennica, D.; Goddard, A.; Phillips, H.; et al. The tumour-suppressor gene patched encodes a candidate receptor for Sonic hedgehog. Nature 1996, 384, 129–134. [Google Scholar] [CrossRef] [PubMed]
  62. Briscoe, J.; Chen, Y.; Jessell, T.M.; Struhl, G. A hedgehog-insensitive form of patched provides evidence for direct long-range morphogen activity of sonic hedgehog in the neural tube. Mol. Cell. 2001, 7, 1279–1291. [Google Scholar] [CrossRef]
  63. Ingham, P.W. How cholesterol modulates the signal. Curr. Biol. 2000, 10, R180–R183. [Google Scholar] [CrossRef]
  64. Carpenter, D.; Stone, D.M.; Brush, J.; Ryan, A.; Armanini, M.; Frantz, G.; Rosenthal, A.; de Sauvage, F.J. Characterization of two patched receptors for the vertebrate hedgehog protein family. Proc. Natl. Acad. Sci. USA 1998, 95, 13630–13634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Gailani, M.R.; Stahle-Backdahl, M.; Leffell, D.J.; Glynn, M.; Zaphiropoulos, P.G.; Pressman, C.; Unden, A.B.; Dean, M.; Brash, D.E.; Bale, A.E.; et al. The role of the human homologue of Drosophila patched in sporadic basal cell carcinomas. Nat. Genet. 1996, 14, 78–81. [Google Scholar] [CrossRef] [PubMed]
  66. Johnson, R.L.; Rothman, A.L.; Xie, J.; Goodrich, L.V.; Bare, J.W.; Bonifas, J.M.; Quinn, A.G.; Myers, R.M.; Cox, D.R.; Epstein, E.H., Jr.; et al. Human homolog of patched, a candidate gene for the basal cell nevus syndrome. Science 1996, 272, 1668–1671. [Google Scholar] [CrossRef] [PubMed]
  67. Smyth, I.; Narang, M.A.; Evans, T.; Heimann, C.; Nakamura, Y.; Chenevix-Trench, G.; Pietsch, T.; Wicking, C.; Wainwright, B.J. Isolation and characterization of human patched 2 (PTCH2), a putative tumour suppressor gene inbasal cell carcinoma and medulloblastoma on chromosome 1p32. Hum. Mol. Genet. 1999, 8, 291–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Alcedo, J.; Ayzenzon, M.; Von Ohlen, T.; Noll, M.; Hooper, J.E. The Drosophila smoothened gene encodes a seven-pass membrane protein, a putative receptor for the hedgehog signal. Cell 1996, 86, 221–232. [Google Scholar] [CrossRef]
  69. Rana, R.; Carroll, C.E.; Lee, H.J.; Bao, J.; Marada, S.; Grace, C.R.; Guibao, C.D.; Ogden, S.K.; Zheng, J.J. Structural insights into the role of the Smoothened cysteine-rich domain in Hedgehog signalling. Nat. Commun. 2013, 4, 2965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Murone, M.; Rosenthal, A.; de Sauvage, F.J. Hedgehog signal transduction: From flies to vertebrates. Exp. Cell Res. 1999, 253, 25–33. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, Y.; Struhl, G. In vivo evidence that Patched and Smoothened constitute distinct binding and transducing components of a Hedgehog receptor complex. Development 1998, 125, 4943–4948. [Google Scholar] [PubMed]
  72. Taipale, J.; Cooper, M.K.; Maiti, T.; Beachy, P.A. Patched acts catalytically to suppress the activity of Smoothened. Nature 2002, 418, 892–897. [Google Scholar] [CrossRef] [PubMed]
  73. Jia, J.; Tong, C.; Wang, B.; Luo, L.; Jiang, J. Hedgehog signalling activity of Smoothened requires phosphorylation by protein kinase A and casein kinase I. Nature 2004, 432, 1045–1050. [Google Scholar] [CrossRef] [PubMed]
  74. 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] [PubMed]
  75. Yoon, J.W.; Liu, C.Z.; Yang, J.T.; Swart, R.; Iannaccone, P.; Walterhouse, D. GLI activates transcription through a herpes simplex viral protein 16-like activation domain. J. Biol. Chem. 1998, 273, 3496–3501. [Google Scholar] [CrossRef] [PubMed]
  76. Sasaki, H.; Hui, C.; Nakafuku, M.; Kondoh, H. A binding site for Gli proteins is essential for HNF-3beta floor plate enhancer activity in transgenics and can respond to Shh in vitro. Development 1997, 124, 1313–1322. [Google Scholar] [PubMed]
  77. Kinzler, K.W.; Vogelstein, B. The GLI gene encodes a nuclear protein which binds specific sequences in the human genome. Mol. Cell. Biol. 1990, 10, 634–642. [Google Scholar] [CrossRef] [PubMed]
  78. Abbasi, A.A.; Goode, D.K.; Amir, S.; Grzeschik, K.H. Evolution and functional diversification of the GLI family of transcription factors in vertebrates. Evol. Bioinform. Online 2009, 5, 5–13. [Google Scholar] [CrossRef] [PubMed]
  79. Hynes, M.; Stone, D.M.; Dowd, M.; Pitts-Meek, S.; Goddard, A.; Gurney, A.; Rosenthal, A. Control of cell pattern in the neural tube by the zinc finger transcription factor and oncogene Gli-1. Neuron 1997, 19, 15–26. [Google Scholar] [CrossRef]
  80. Santoni, M.; Burattini, L.; Nabissi, M.; Morelli, M.B.; Berardi, R.; Santoni, G.; Cascinu, S. Essential role of Gli proteins in glioblastoma multiforme. Curr. Protein Pept. Sci. 2013, 14, 133–140. [Google Scholar] [CrossRef] [PubMed]
  81. Persson, M.; Stamataki, D.; te Welscher, P.; Andersson, E.; Bose, J.; Ruther, U.; Ericson, J.; Briscoe, J. Dorsal-ventral patterning of the spinal cord requires Gli3 transcriptional repressor activity. Genes Dev. 2002, 16, 2865–2878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Aza-Blanc, P.; Lin, H.Y.; Ruiz i Altaba, A.; Kornberg, T.B. Expression of the vertebrate Gli proteins in Drosophila reveals a distribution of activator and repressor activities. Development 2000, 127, 4293–4301. [Google Scholar] [PubMed]
  83. Dai, P.; Akimaru, H.; Tanaka, Y.; Maekawa, T.; Nakafuku, M.; Ishii, S. Sonic Hedgehog-induced activation of the Gli1 promoter is mediated by GLI3. J. Biol. Chem. 1999, 274, 8143–8152. [Google Scholar] [CrossRef] [PubMed]
  84. Sasaki, H.; Nishizaki, Y.; Hui, C.; Nakafuku, M.; Kondoh, H. Regulation of Gli2 and Gli3 activities by an amino-terminal repression domain: Implication of Gli2 and Gli3 as primary mediators of Shh signaling. Development 1999, 126, 3915–3924. [Google Scholar] [PubMed]
  85. Denef, N.; Neubuser, 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]
  86. Hui, C.C.; Angers, S. Gli proteins in development and disease. Annu. Rev. Cell Dev. Biol. 2011, 27, 513–537. [Google Scholar] [CrossRef] [PubMed]
  87. 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] [PubMed]
  88. Aberger, F.; Ruiz, I.A.A. Context-dependent signal integration by the GLI code: The oncogenic load, pathways, modifiers and implications for cancer therapy. Semin. Cell Dev. Biol 2014, 33, 93–104. [Google Scholar] [CrossRef] [PubMed]
  89. Mastronardi, F.G.; Dimitroulakos, J.; Kamel-Reid, S.; Manoukian, A.S. Co-localization of patched and activated sonic hedgehog to lysosomes in neurons. Neuroreport 2000, 11, 581–585. [Google Scholar] [CrossRef] [PubMed]
  90. Rohatgi, R.; Milenkovic, L.; Scott, M.P. Patched1 regulates hedgehog signaling at the primary cilium. Science 2007, 317, 372–376. [Google Scholar] [CrossRef] [PubMed]
  91. Humke, E.W.; Dorn, K.V.; Milenkovic, L.; Scott, M.P.; Rohatgi, R. The output of Hedgehog signaling is controlled by the dynamic association between Suppressor of Fused and the Gli proteins. Genes Dev. 2010, 24, 670–682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Liem, K.F., Jr.; He, M.; Ocbina, P.J.; Anderson, K.V. Mouse Kif7/Costal2 is a cilia-associated protein that regulates Sonic hedgehog signaling. Proc. Natl. Acad. Sci. USA 2009, 106, 13377–13382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Cheung, H.O.; Zhang, X.; Ribeiro, A.; Mo, R.; Makino, S.; Puviindran, V.; Law, K.K.; Briscoe, J.; Hui, C.C. The kinesin protein Kif7 is a critical regulator of Gli transcription factors in mammalian hedgehog signaling. Sci. Signal. 2009, 2, ra29. [Google Scholar] [CrossRef] [PubMed]
  94. Endoh-Yamagami, S.; Evangelista, M.; Wilson, D.; Wen, X.; Theunissen, J.W.; Phamluong, K.; Davis, M.; Scales, S.J.; Solloway, M.J.; de Sauvage, F.J.; et al. The mammalian Cos2 homolog Kif7 plays an essential role in modulating Hh signal transduction during development. Curr. Biol. 2009, 19, 1320–1326. [Google Scholar] [CrossRef] [PubMed]
  95. Cohen, M.; Kicheva, A.; Ribeiro, A.; Blassberg, R.; Page, K.M.; Barnes, C.P.; Briscoe, J. Ptch1 and Gli regulate Shh signalling dynamics via multiple mechanisms. Nat. Commun. 2015, 6, 6709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Brennan, D.; Chen, X.; Cheng, L.; Mahoney, M.; Riobo, N.A. Noncanonical Hedgehog signaling. Vitam. Horm. 2012, 88, 55–72. [Google Scholar] [CrossRef] [PubMed]
  97. Regan, J.L.; Schumacher, D.; Staudte, S.; Steffen, A.; Haybaeck, J.; Keilholz, U.; Schweiger, C.; Golob-Schwarzl, N.; Mumberg, D.; Henderson, D.; et al. Non-Canonical Hedgehog Signaling Is a Positive Regulator of the WNT Pathway and Is Required for the Survival of Colon Cancer Stem Cells. Cell Rep. 2017, 21, 2813–2828. [Google Scholar] [CrossRef] [PubMed]
  98. Nolan-Stevaux, O.; Lau, J.; Truitt, M.L.; Chu, G.C.; Hebrok, M.; Fernández-Zapico, M.E.; Hanahan, D. GLI1 is regulated through Smoothened-independent mechanisms in neoplastic pancreatic ducts and mediates PDAC cell survival and transformation. Genes Dev. 2009, 23, 24–36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Petrova, E.; Matevossian, A.; Resh, M.D. Hedgehog acyltransferase as a target in pancreatic ductal adenocarcinoma. Oncogene 2015, 34, 263–268. [Google Scholar] [CrossRef] [PubMed]
  100. Chinchilla, P.; Xiao, L.; Kazanietz, M.G.; Riobo, N.A. Hedgehog proteins activate pro-angiogenic responses in endothelial cells through non-canonical signaling pathways. Cell Cycle 2010, 9, 570–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Rajurkar, M.; De Jesus-Monge, W.E.; Driscoll, D.R.; Appleman, V.A.; Huang, H.; Cotton, J.L.; Klimstra, D.S.; Zhu, L.J.; Simin, K.; Xu, L.; et al. The activity of Gli transcription factors is essential for Kras-induced pancreatic tumorigenesis. Proc. Natl. Acad. Sci. USA 2012, 109, E1038–E1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Jiang, X.; Yang, P.; Ma, L. Kinase activity-independent regulation of cyclin pathway by GRK2 is essential for zebrafish early development. Proc. Natl. Acad. Sci. USA 2009, 106, 10183–10188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Polizio, A.H.; Chinchilla, P.; Chen, X.; Kim, S.; Manning, D.R.; Riobo, N.A. Heterotrimeric Gi proteins link Hedgehog signaling to activation of Rho small GTPases to promote fibroblast migration. J. Biol. Chem. 2011, 286, 19589–19596. [Google Scholar] [CrossRef] [PubMed]
  104. 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]
  105. Bijlsma, M.F.; Borensztajn, K.S.; Roelink, H.; Peppelenbosch, M.P.; Spek, C.A. Sonic hedgehog induces transcription-independent cytoskeletal rearrangement and migration regulated by arachidonate metabolites. Cell Signal. 2007, 19, 2596–2604. [Google Scholar] [CrossRef] [PubMed]
  106. Bijlsma, M.F.; Peppelenbosch, M.P.; Spek, C.A.; Roelink, H. Leukotriene synthesis is required for hedgehog-dependent neurite projection in neuralized embryoid bodies but not for motor neuron differentiation. Stem Cells 2008, 26, 1138–1145. [Google Scholar] [CrossRef] [PubMed]
  107. Yam, P.T.; Langlois, S.D.; Morin, S.; Charron, F. Sonic hedgehog guides axons through a noncanonical, Src-family-kinase-dependent signaling pathway. Neuron 2009, 62, 349–362. [Google Scholar] [CrossRef] [PubMed]
  108. Hahn, H.; Wicking, C.; Zaphiropoulous, P.G.; Gailani, M.R.; Shanley, S.; Chidambaram, A.; Vorechovsky, I.; Holmberg, E.; Unden, A.B.; Gillies, S.; et al. Mutations of the human homolog of Drosophila patched in the nevoid basal cell carcinoma syndrome. Cell 1996, 85, 841–851. [Google Scholar] [CrossRef]
  109. Lo Muzio, L. Nevoid basal cell carcinoma syndrome (Gorlin syndrome). Orphanet J. Rare Dis. 2008, 3, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. 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] [PubMed]
  111. Goodrich, L.V.; Milenkovic, L.; Higgins, K.M.; Scott, M.P. Altered neural cell fates and medulloblastoma in mouse patched mutants. Science 1997, 277, 1109–1113. [Google Scholar] [CrossRef] [PubMed]
  112. Raffel, C.; Jenkins, R.B.; Frederick, L.; Hebrink, D.; Alderete, B.; Fults, D.W.; James, C.D. Sporadic medulloblastomas contain PTCH mutations. Cancer Res. 1997, 57, 842–845. [Google Scholar] [PubMed]
  113. Tostar, U.; Malm, C.J.; Meis-Kindblom, J.M.; Kindblom, L.G.; Toftgard, R.; Unden, A.B. Deregulation of the hedgehog signalling pathway: A possible role for the PTCH and SUFU genes in human rhabdomyoma and rhabdomyosarcoma development. J. Pathol. 2006, 208, 17–25. [Google Scholar] [CrossRef] [PubMed]
  114. Reifenberger, J.; Wolter, M.; Weber, R.G.; Megahed, M.; Ruzicka, T.; Lichter, P.; Reifenberger, G. Missense mutations in SMOH in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res. 1998, 58, 1798–1803. [Google Scholar] [PubMed]
  115. 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] [PubMed]
  116. Ellison, D.W.; Dalton, J.; Kocak, M.; Nicholson, S.L.; Fraga, C.; Neale, G.; Kenney, A.M.; Brat, D.J.; Perry, A.; Yong, W.H.; et al. Medulloblastoma: Clinicopathological correlates of SHH, WNT, and non-SHH/WNT molecular subgroups. Acta Neuropathol. 2011, 121, 381–396. [Google Scholar] [CrossRef] [PubMed]
  117. 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]
  118. Reifenberger, J.; Wolter, M.; Knobbe, C.B.; Kohler, B.; Schonicke, A.; Scharwachter, 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] [PubMed]
  119. Kool, M.; Jones, D.T.; Jager, 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] [PubMed]
  120. 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] [PubMed] [Green Version]
  121. Maesawa, C.; Tamura, G.; Iwaya, T.; Ogasawara, S.; Ishida, K.; Sato, N.; Nishizuka, S.; Suzuki, Y.; Ikeda, K.; Aoki, K.; et al. Mutations in the human homologue of the Drosophila patched gene in esophageal squamous cell carcinoma. Genes Chromosomes Cancer 1998, 21, 276–279. [Google Scholar] [CrossRef]
  122. Vorechovsky, I.; Unden, A.B.; Sandstedt, B.; Toftgard, R.; Stahle-Backdahl, M. Trichoepitheliomas contain somatic mutations in the overexpressed PTCH gene: Support for a gatekeeper mechanism in skin tumorigenesis. Cancer Res. 1997, 57, 4677–4681. [Google Scholar] [PubMed]
  123. Adolphe, C.; Hetherington, R.; Ellis, T.; Wainwright, B. Patched1 functions as a gatekeeper by promoting cell cycle progression. Cancer Res. 2006, 66, 2081–2088. [Google Scholar] [CrossRef] [PubMed]
  124. Aszterbaum, M.; Epstein, J.; Oro, A.; Douglas, V.; LeBoit, P.E.; Scott, M.P.; Epstein, E.H., Jr. Ultraviolet and ionizing radiation enhance the growth of BCCs and trichoblastomas in patched heterozygous knockout mice. Nat. Med. 1999, 5, 1285–1291. [Google Scholar] [CrossRef] [PubMed]
  125. Svard, J.; Heby-Henricson, K.; Persson-Lek, M.; Rozell, B.; Lauth, M.; Bergstrom, A.; Ericson, J.; Toftgard, R.; Teglund, S. Genetic elimination of Suppressor of fused reveals an essential repressor function in the mammalian Hedgehog signaling pathway. Dev. Cell. 2006, 10, 187–197. [Google Scholar] [CrossRef] [PubMed]
  126. Pepicelli, C.V.; Lewis, P.M.; McMahon, A.P. Sonic hedgehog regulates branching morphogenesis in the mammalian lung. Curr. Biol. 1998, 8, 1083–1086. [Google Scholar] [CrossRef]
  127. 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] [PubMed]
  128. Varnat, F.; Duquet, A.; Malerba, M.; Zbinden, M.; Mas, C.; Gervaz, P.; Ruiz i Altaba, A. Human colon cancer epithelial cells harbour active HEDGEHOG-GLI signalling that is essential for tumour growth, recurrence, metastasis and stem cell survival and expansion. EMBO Mol. Med. 2009, 1, 338–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Datta, S.; Datta, M.W. Sonic Hedgehog signaling in advanced prostate cancer. Cell. Mol. Life Sci. 2006, 63, 435–448. [Google Scholar] [CrossRef] [PubMed]
  130. Huang, S.; He, J.; Zhang, X.; Bian, Y.; Yang, L.; Xie, G.; Zhang, K.; Tang, W.; Stelter, A.A.; Wang, Q.; et al. Activation of the hedgehog pathway in human hepatocellular carcinomas. Carcinogenesis 2006, 27, 1334–1340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Kubo, M.; Nakamura, M.; Tasaki, A.; Yamanaka, N.; Nakashima, H.; Nomura, M.; Kuroki, S.; Katano, M. Hedgehog signaling pathway is a new therapeutic target for patients with breast cancer. Cancer Res. 2004, 64, 6071–6074. [Google Scholar] [CrossRef] [PubMed]
  132. Szkandera, J.; Kiesslich, T.; Haybaeck, J.; Gerger, A.; Pichler, M. Hedgehog signaling pathway in ovarian cancer. Int. J. Mol. Sci. 2013, 14, 1179–1196. [Google Scholar] [CrossRef] [PubMed]
  133. Dahmane, N.; Sanchez, P.; Gitton, Y.; Palma, V.; Sun, T.; Beyna, M.; Weiner, H.; Ruiz i Altaba, A. The Sonic Hedgehog-Gli pathway regulates dorsal brain growth and tumorigenesis. Development 2001, 128, 5201–5212. [Google Scholar] [PubMed]
  134. O’Reilly, K.E.; de Miera, E.V.; Segura, M.F.; Friedman, E.; Poliseno, L.; Han, S.W.; Zhong, J.; Zavadil, J.; Pavlick, A.; Hernando, E.; et al. Hedgehog pathway blockade inhibits melanoma cell growth in vitro and in vivo. Pharmaceuticals (Basel) 2013, 6, 1429–1450. [Google Scholar] [CrossRef] [PubMed]
  135. Thayer, S.P.; di Magliano, M.P.; Heiser, P.W.; Nielsen, C.M.; Roberts, D.J.; Lauwers, G.Y.; Qi, Y.P.; Gysin, S.; Fernandez-del Castillo, C.; Yajnik, V.; et al. Hedgehog is an early and late mediator of pancreatic cancer tumorigenesis. Nature 2003, 425, 851–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Mukherjee, S.; Frolova, N.; Sadlonova, A.; Novak, Z.; Steg, A.; Page, G.P.; Welch, D.R.; Lobo-Ruppert, S.M.; Ruppert, J.M.; Johnson, M.R.; et al. Hedgehog signaling and response to cyclopamine differ in epithelial and stromal cells in benign breast and breast cancer. Cancer Biol. Ther. 2006, 5, 674–683. [Google Scholar] [CrossRef] [PubMed]
  137. Fan, L.; Pepicelli, C.V.; Dibble, C.C.; Catbagan, W.; Zarycki, J.L.; Laciak, R.; Gipp, J.; Shaw, A.; Lamm, M.L.; Munoz, A.; et al. Hedgehog signaling promotes prostate xenograft tumor growth. Endocrinology 2004, 145, 3961–3970. [Google Scholar] [CrossRef] [PubMed]
  138. Tian, H.; Callahan, C.A.; DuPree, K.J.; Darbonne, W.C.; Ahn, C.P.; Scales, S.J.; de Sauvage, F.J. Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc. Natl. Acad. Sci. USA 2009, 106, 4254–4259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Ma, X.; Sheng, T.; Zhang, Y.; Zhang, X.; He, J.; Huang, S.; Chen, K.; Sultz, J.; Adegboyega, P.A.; Zhang, H.; et al. Hedgehog signaling is activated in subsets of esophageal cancers. Int. J. Cancer 2006, 118, 139–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. 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]
  141. Blotta, S.; Jakubikova, J.; Calimeri, T.; Roccaro, A.M.; Amodio, N.; Azab, A.K.; Foresta, U.; Mitsiades, C.S.; Rossi, M.; Todoerti, K.; et al. Canonical and noncanonical Hedgehog pathway in the pathogenesis of multiple myeloma. Blood 2012, 120, 5002–5013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Becher, O.J.; Hambardzumyan, D.; Fomchenko, E.I.; Momota, H.; Mainwaring, L.; Bleau, A.M.; Katz, A.M.; Edgar, M.; Kenney, A.M.; Cordon-Cardo, C.; et al. Gli activity correlates with tumor grade in platelet-derived growth factor-induced gliomas. Cancer Res. 2008, 68, 2241–2249. [Google Scholar] [CrossRef] [PubMed]
  143. Ma, Y.; Zhang, P.; Wang, F.; Yang, J.; Yang, Z.; Qin, H. The relationship between early embryo development and tumourigenesis. J. Cell. Mol. Med. 2010, 14, 2697–2701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Liu, S.; Dontu, G.; Mantle, I.D.; Patel, S.; Ahn, N.S.; Jackson, K.W.; Suri, P.; Wicha, M.S. Hedgehog signaling and Bmi-1 regulate self-renewal of normal and malignant human mammary stem cells. Cancer Res. 2006, 66, 6063–6071. [Google Scholar] [CrossRef] [PubMed]
  145. Zhao, C.; Chen, A.; Jamieson, C.H.; Fereshteh, M.; Abrahamsson, A.; Blum, J.; Kwon, H.Y.; Kim, J.; Chute, J.P.; Rizzieri, D.; et al. Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 2009, 458, 776–779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Santini, R.; Vinci, M.C.; Pandolfi, S.; Penachioni, J.Y.; Montagnani, V.; Olivito, B.; Gattai, R.; Pimpinelli, N.; Gerlini, G.; Borgognoni, L.; et al. Hedgehog-GLI signaling drives self-renewal and tumorigenicity of human melanoma-initiating cells. Stem Cells 2012, 30, 1808–1818. [Google Scholar] [CrossRef] [PubMed]
  147. Song, L.; Chen, X.; Gao, S.; Zhang, C.; Qu, C.; Wang, P.; Liu, L. Ski modulate the characteristics of pancreatic cancer stem cells via regulating sonic hedgehog signaling pathway. Tumour Biol. 2016. [Google Scholar] [CrossRef]
  148. Quint, K.; Tonigold, M.; Di Fazio, P.; Montalbano, R.; Lingelbach, S.; Ruckert, F.; Alinger, B.; Ocker, M.; Neureiter, D. Pancreatic cancer cells surviving gemcitabine treatment express markers of stem cell differentiation and epithelial-mesenchymal transition. Int. J. Oncol. 2012, 41, 2093–2102. [Google Scholar] [CrossRef] [PubMed]
  149. Sims-Mourtada, J.; Izzo, J.G.; Ajani, J.; Chao, K.S. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene 2007, 26, 5674–5679. [Google Scholar] [CrossRef] [PubMed]
  150. 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]
  151. P, D.I.F.; Montalbano, R.; Quint, K.; Alinger, B.; Kemmerling, R.; Kiesslich, T.; Ocker, M.; Neureiter, D. The pan-deacetylase inhibitor panobinostat modulates the expression of epithelial-mesenchymal transition markers in hepatocellular carcinoma models. Oncol. Lett. 2013, 5, 127–134. [Google Scholar] [CrossRef]
  152. Chen, W.; Tang, T.; Eastham-Anderson, J.; Dunlap, D.; Alicke, B.; Nannini, M.; Gould, S.; Yauch, R.; Modrusan, Z.; DuPree, K.J.; et al. Canonical hedgehog signaling augments tumor angiogenesis by induction of VEGF-A in stromal perivascular cells. Proc. Natl. Acad. Sci. USA 2011, 108, 9589–9594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Di Mauro, C.; Rosa, R.; D’Amato, V.; Ciciola, P.; Servetto, A.; Marciano, R.; Orsini, R.C.; Formisano, L.; De Falco, S.; Cicatiello, V.; et al. Hedgehog signalling pathway orchestrates angiogenesis in triple-negative breast cancers. Br. J. Cancer 2017, 116, 1425–1435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. 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] [PubMed]
  155. Sharpe, H.J.; Pau, G.; Dijkgraaf, G.J.; Basset-Seguin, N.; Modrusan, Z.; Januario, T.; Tsui, V.; Durham, A.B.; Dlugosz, A.A.; Haverty, P.M.; et al. Genomic analysis of smoothened inhibitor resistance in basal cell carcinoma. Cancer Cell. 2015, 27, 327–341. [Google Scholar] [CrossRef] [PubMed]
  156. Atwood, S.X.; Sarin, K.Y.; Whitson, R.J.; Li, J.R.; Kim, G.; Rezaee, M.; Ally, M.S.; Kim, J.; Yao, C.; Chang, A.L.; et al. Smoothened variants explain the majority of drug resistance in basal cell carcinoma. Cancer Cell. 2015, 27, 342–353. [Google Scholar] [CrossRef] [PubMed]
  157. Robinson, G.W.; Orr, B.A.; Wu, G.; Gururangan, S.; Lin, T.; Qaddoumi, I.; Packer, R.J.; Goldman, S.; Prados, M.D.; Desjardins, A.; et al. Vismodegib Exerts Targeted Efficacy Against Recurrent Sonic Hedgehog-Subgroup Medulloblastoma: Results From Phase II Pediatric Brain Tumor Consortium Studies PBTC-025B and PBTC-032. J. Clin. Oncol. 2015, 33, 2646–2654. [Google Scholar] [CrossRef] [PubMed]
  158. Rudin, C.M.; Hann, C.L.; Laterra, J.; Yauch, R.L.; Callahan, C.A.; Fu, L.; Holcomb, T.; Stinson, J.; Gould, S.E.; Coleman, B.; et al. Treatment of medulloblastoma with hedgehog pathway inhibitor GDC-0449. N. Engl. J. Med. 2009, 361, 1173–1178. [Google Scholar] [CrossRef] [PubMed]
  159. Danial, C.; Sarin, K.Y.; Oro, A.E.; Chang, A.L. An Investigator-Initiated Open-Label Trial of Sonidegib in Advanced Basal Cell Carcinoma Patients Resistant to Vismodegib. Clin. Cancer Res. 2016, 22, 1325–1329. [Google Scholar] [CrossRef] [PubMed]
  160. Ishii, T.; Shimizu, Y.; Nakashima, K.; Kondo, S.; Ogawa, K.; Sasaki, S.; Matsui, H. Inhibition mechanism exploration of investigational drug TAK-441 as inhibitor against Vismodegib-resistant Smoothened mutant. Eur. J. Pharmacol. 2014, 723, 305–313. [Google Scholar] [CrossRef] [PubMed]
  161. Lee, M.J.; Hatton, B.A.; Villavicencio, E.H.; Khanna, P.C.; Friedman, S.D.; Ditzler, S.; Pullar, B.; Robison, K.; White, K.F.; Tunkey, C.; et al. Hedgehog pathway inhibitor saridegib (IPI-926) increases lifespan in a mouse medulloblastoma model. Proc. Natl. Acad. Sci. USA 2012, 109, 7859–7864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Peukert, S.; He, F.; Dai, M.; Zhang, R.; Sun, Y.; Miller-Moslin, K.; McEwan, M.; Lagu, B.; Wang, K.; Yusuff, N.; et al. Discovery of NVP-LEQ506, a second-generation inhibitor of smoothened. ChemMedChem 2013, 8, 1261–1265. [Google Scholar] [CrossRef] [PubMed]
  163. Li, Q.R.; Zhao, H.; Zhang, X.S.; Lang, H.; Yu, K. Novel-smoothened inhibitors for therapeutic targeting of naive and drug-resistant hedgehog pathway-driven cancers. Acta Pharmacol. Sin. 2018. [Google Scholar] [CrossRef]
  164. Colmont, C.S.; Benketah, A.; Reed, S.H.; Hawk, N.V.; Telford, W.G.; Ohyama, M.; Udey, M.C.; Yee, C.L.; Vogel, J.C.; Patel, G.K. CD200-expressing human basal cell carcinoma cells initiate tumor growth. Proc. Natl. Acad. Sci. USA 2013, 110, 1434–1439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Palma, V.; Ruiz i Altaba, A. Hedgehog-GLI signaling regulates the behavior of cells with stem cell properties in the developing neocortex. Development 2004, 131, 337–345. [Google Scholar] [CrossRef] [PubMed]
  166. Fu, J.; Rodova, M.; Roy, S.K.; Sharma, J.; Singh, K.P.; Srivastava, R.K.; Shankar, S. GANT-61 inhibits pancreatic cancer stem cell growth in vitro and in NOD/SCID/IL2R gamma null mice xenograft. Cancer Lett. 2013, 330, 22–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Gu, D.; Liu, H.; Su, G.H.; Zhang, X.; Chin-Sinex, H.; Hanenberg, H.; Mendonca, M.S.; Shannon, H.E.; Chiorean, E.G.; Xie, J. Combining hedgehog signaling inhibition with focal irradiation on reduction of pancreatic cancer metastasis. Mol. Cancer Ther. 2013, 12, 1038–1048. [Google Scholar] [CrossRef] [PubMed]
  168. Huang, F.T.; Zhuan-Sun, Y.X.; Zhuang, Y.Y.; Wei, S.L.; Tang, J.; Chen, W.B.; Zhang, S.N. Inhibition of hedgehog signaling depresses self-renewal of pancreatic cancer stem cells and reverses chemoresistance. Int. J. Oncol. 2012, 41, 1707–1714. [Google Scholar] [CrossRef] [PubMed]
  169. Li, S.-H.; Fu, J.; Watkins, D.N.; Srivastava, R.K.; Shankar, S. Sulforaphane regulates self-renewal of pancreatic cancer stem cells through the modulation of Sonic hedgehog–GLI pathway. Mol. Cell. Biochem. 2013, 373, 217–227. [Google Scholar] [CrossRef] [PubMed]
  170. Ma, Y.; Yu, W.; Shrivastava, A.; Alemi, F.; Lankachandra, K.; Srivastava, R.K.; Shankar, S. Sanguinarine inhibits pancreatic cancer stem cell characteristics by inducing oxidative stress and suppressing sonic hedgehog-Gli-Nanog pathway. Carcinogenesis 2017, 38, 1047–1056. [Google Scholar] [CrossRef] [PubMed]
  171. Song, L.; Chen, X.; Wang, P.; Gao, S.; Qu, C.; Liu, L. Effects of baicalein on pancreatic cancer stem cells via modulation of sonic Hedgehog pathway. Acta Biochim. Biophys. Sin. 2018, 50, 586–596. [Google Scholar] [CrossRef] [PubMed]
  172. Yao, J.; An, Y.; Wie, J.S.; Ji, Z.L.; Lu, Z.P.; Wu, J.L.; Jiang, K.R.; Chen, P.; Xu, Z.K.; Miao, Y. Cyclopamine reverts acquired chemoresistance and down-regulates cancer stem cell markers in pancreatic cancer cell lines. Swiss Med. Wkly. 2011, 141, w13208. [Google Scholar] [CrossRef] [PubMed]
  173. Po, A.; Ferretti, E.; Miele, E.; De Smaele, E.; Paganelli, A.; Canettieri, G.; Coni, S.; Di Marcotullio, L.; Biffoni, M.; Massimi, L.; et al. Hedgehog controls neural stem cells through p53-independent regulation of Nanog. EMBO J. 2010, 29, 2646–2658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Read, T.A.; Fogarty, M.P.; Markant, S.L.; McLendon, R.E.; Wei, Z.; Ellison, D.W.; Febbo, P.G.; Wechsler-Reya, R.J. Identification of CD15 as a marker for tumor-propagating cells in a mouse model of medulloblastoma. Cancer Cell 2009, 15, 135–147. [Google Scholar] [CrossRef] [PubMed]
  175. Vanner, R.J.; Remke, M.; Gallo, M.; Selvadurai, H.J.; Coutinho, F.; Lee, L.; Kushida, M.; Head, R.; Morrissy, S.; Zhu, X.; et al. Quiescent sox2(+) cells drive hierarchical growth and relapse in sonic hedgehog subgroup medulloblastoma. Cancer Cell 2014, 26, 33–47. [Google Scholar] [CrossRef] [PubMed]
  176. Dierks, C.; Beigi, R.; Guo, G.R.; Zirlik, K.; Stegert, M.R.; Manley, P.; Trussell, C.; Schmitt-Graeff, A.; Landwerlin, K.; Veelken, H.; et al. Expansion of Bcr-Abl-positive leukemic stem cells is dependent on Hedgehog pathway activation. Cancer Cell 2008, 14, 238–249. [Google Scholar] [CrossRef] [PubMed]
  177. Irvine, D.A.; Zhang, B.; Allan, E.K.; Holyoake, T.L.; Dorsch, M.; Manley, P.W.; Bhatia, R.; Copland, M. Combination of the hedgehog pathway inhibitor LDE225 and nilotinib eliminates chronic myeloid leukemia stem and progenitor cells. Blood 2009, 114, 1428. [Google Scholar]
  178. Queiroz, K.C.; Ruela-de-Sousa, R.R.; Fuhler, G.M.; Aberson, H.L.; Ferreira, C.V.; Peppelenbosch, M.P.; Spek, C.A. Hedgehog signaling maintains chemoresistance in myeloid leukemic cells. Oncogene 2010, 29, 6314–6322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Sadarangani, A.; Pineda, G.; Lennon, K.M.; Chun, H.J.; Shih, A.; Schairer, A.E.; Court, A.C.; Goff, D.J.; Prashad, S.L.; Geron, I.; et al. GLI2 inhibition abrogates human leukemia stem cell dormancy. J. Transl. Med. 2015, 13, 98. [Google Scholar] [CrossRef] [PubMed]
  180. Kakiuchi, S.; Minami, Y.; Miyata, Y.; Mizutani, Y.; Goto, H.; Kawamoto, S.; Yakushijin, K.; Kurata, K.; Matsuoka, H.; Minami, H. NANOG expression as a responsive biomarker during treatment with Hedgehog signal inhibitor in acute myeloid leukemia. Int. J. Mol. Sci. 2017, 18, 486. [Google Scholar] [CrossRef] [PubMed]
  181. Kobune, M.; Takimoto, R.; Murase, K.; Iyama, S.; Sato, T.; Kikuchi, S.; Kawano, Y.; Miyanishi, K.; Sato, Y.; Niitsu, Y.; et al. Drug resistance is dramatically restored by hedgehog inhibitors in CD34+ leukemic cells. Cancer Sci. 2009, 100, 948–955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Minami, Y.; Hayakawa, F.; Kiyoi, H.; Sadarangani, A.; Jamieson, C.H.; Naoe, T. Treatment with hedgehog inhibitor, PF-04449913, attenuates leukemia-initiation potential in acute myeloid leukemia cells. Blood 2013, 122, 1649. [Google Scholar]
  183. Ji, Z.; Mei, F.C.; Johnson, B.H.; Thompson, E.B.; Cheng, X. Protein kinase A, not Epac, suppresses hedgehog activity and regulates glucocorticoid sensitivity in acute lymphoblastic leukemia cells. J. Biol. Chem. 2007, 282, 37370–37377. [Google Scholar] [CrossRef] [PubMed]
  184. Lang, F.; Badura, S.; Ruthardt, M.; Rieger, M.A.; Ottmann, O.G. Modulation of Leukemic Stem Cell Self-Renewal and Cell Fate Decisions by Inhibition of Hedgehog Signalling in Human Acute Lymphoblastic Leukemia (ALL). Blood 2012, 120, 2578. [Google Scholar]
  185. Lin, T.L.; Wang, Q.H.; Brown, P.; Peacock, C.; Merchant, A.A.; Brennan, S.; Jones, E.; McGovern, K.; Watkins, D.N.; Sakamoto, K.M. Self-renewal of acute lymphocytic leukemia cells is limited by the Hedgehog pathway inhibitors cyclopamine and IPI-926. PLoS ONE 2010, 5, e15262. [Google Scholar] [CrossRef] [PubMed]
  186. Grachtchouk, M.; Mo, R.; Yu, S.; Zhang, X.; Sasaki, H.; Hui, C.C.; Dlugosz, A.A. Basal cell carcinomas in mice overexpressing Gli2 in skin. Nat. Genet. 2000, 24, 216–217. [Google Scholar] [CrossRef] [PubMed]
  187. Otsuka, A.; Dreier, J.; Cheng, P.F.; Nageli, M.; Lehmann, H.; Felderer, L.; Frew, I.J.; Matsushita, S.; Levesque, M.P.; Dummer, R. Hedgehog pathway inhibitors promote adaptive immune responses in basal cell carcinoma. Clin. Cancer Res. 2015, 21, 1289–1297. [Google Scholar] [CrossRef] [PubMed]
  188. El Andaloussi, A.; Graves, S.; Meng, F.; Mandal, M.; Mashayekhi, M.; Aifantis, I. Hedgehog signaling controls thymocyte progenitor homeostasis and differentiation in the thymus. Nat. Immunol. 2006, 7, 418–426. [Google Scholar] [CrossRef] [PubMed]
  189. Furmanski, A.L.; Barbarulo, A.; Solanki, A.; Lau, C.I.; Sahni, H.; Saldana, J.I.; D’Acquisto, F.; Crompton, T. The transcriptional activator Gli2 modulates T-cell receptor signalling through attenuation of AP-1 and NFkappaB activity. J. Cell. Sci. 2015, 128, 2085–2095. [Google Scholar] [CrossRef] [PubMed]
  190. Rowbotham, N.J.; Hager-Theodorides, A.L.; Cebecauer, M.; Shah, D.K.; Drakopoulou, E.; Dyson, J.; Outram, S.V.; Crompton, T. Activation of the Hedgehog signaling pathway in T-lineage cells inhibits TCR repertoire selection in the thymus and peripheral T-cell activation. Blood 2007, 109, 3757–3766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Li, Z.; Jiang, J.; Wang, Z.; Zhang, J.; Xiao, M.; Wang, C.; Lu, Y.; Qin, Z. Endogenous interleukin-4 promotes tumor development by increasing tumor cell resistance to apoptosis. Cancer Res. 2008, 68, 8687–8694. [Google Scholar] [CrossRef] [PubMed]
  192. Ziegler, A.; Heidenreich, R.; Braumuller, H.; Wolburg, H.; Weidemann, S.; Mocikat, R.; Rocken, M. EpCAM, a human tumor-associated antigen promotes Th2 development and tumor immune evasion. Blood 2009, 113, 3494–3502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Furmanski, A.L.; Saldana, J.I.; Ono, M.; Sahni, H.; Paschalidis, N.; D’Acquisto, F.; Crompton, T. Tissue-derived hedgehog proteins modulate Th differentiation and disease. J. Immunol. 2013, 190, 2641–2649. [Google Scholar] [CrossRef] [PubMed]
  194. Rowbotham, N.J.; Furmanski, A.L.; Hager-Theodorides, A.L.; Ross, S.E.; Drakopoulou, E.; Koufaris, C.; Outram, S.V.; Crompton, T. Repression of hedgehog signal transduction in T-lineage cells increases TCR-induced activation and proliferation. Cell Cycle 2008, 7, 904–908. [Google Scholar] [CrossRef] [PubMed]
  195. Tabs, S.; Avci, O. Induction of the differentiation and apoptosis of tumor cells in vivo with efficiency and selectivity. Eur. J. Dermatol. 2004, 14, 96–102. [Google Scholar] [PubMed]
  196. Athar, M.; Li, C.; Tang, X.; Chi, S.; Zhang, X.; Kim, A.L.; Tyring, S.K.; Kopelovich, L.; Hebert, J.; Epstein, E.H., Jr.; et al. Inhibition of smoothened signaling prevents ultraviolet B-induced basal cell carcinomas through regulation of Fas expression and apoptosis. Cancer Res. 2004, 64, 7545–7552. [Google Scholar] [CrossRef] [PubMed]
  197. Von Hoff, D.D.; LoRusso, P.M.; Rudin, C.M.; Reddy, J.C.; Yauch, R.L.; Tibes, R.; Weiss, G.J.; Borad, M.J.; Hann, C.L.; Brahmer, J.R.; et al. Inhibition of the hedgehog pathway in advanced basal-cell carcinoma. N. Engl. J. Med. 2009, 361, 1164–1172. [Google Scholar] [CrossRef] [PubMed]
  198. Yauch, R.L.; Gould, S.E.; Scales, S.J.; Tang, T.; Tian, H.; Ahn, C.P.; Marshall, D.; Fu, L.; Januario, T.; Kallop, D. A paracrine requirement for hedgehog signalling in cancer. Nature 2008, 455, 406–410. [Google Scholar] [CrossRef] [PubMed]
  199. Jimeno, A.; Weiss, G.J.; Miller, W.H., Jr.; Gettinger, S.; Eigl, B.J.; Chang, A.L.; Dunbar, J.; Devens, S.; Faia, K.; Skliris, G.; et al. Phase I study of the Hedgehog pathway inhibitor IPI-926 in adult patients with solid tumors. Clin. Cancer Res. 2013, 19, 2766–2774. [Google Scholar] [CrossRef] [PubMed]
  200. Atwood, S.X.; Chang, A.L.; Oro, A.E. Hedgehog pathway inhibition and the race against tumor evolution. J. Cell. Biol. 2012, 199, 193–197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Morgan, H.; Olivero, C.; Patel, G.K. Identification of Human Cutaneous Basal Cell Carcinoma Cancer Stem Cells. Methods Mol. Biol. 2018. [Google Scholar] [CrossRef]
  202. Li, T.; Liao, X.; Lochhead, P.; Morikawa, T.; Yamauchi, M.; Nishihara, R.; Inamura, K.; Kim, S.A.; Mima, K.; Sukawa, Y.; et al. SMO expression in colorectal cancer: Associations with clinical, pathological, and molecular features. Ann. Surg. Oncol. 2014, 21, 4164–4173. [Google Scholar] [CrossRef] [PubMed]
  203. Yoshikawa, K.; Shimada, M.; Miyamoto, H.; Higashijima, J.; Miyatani, T.; Nishioka, M.; Kurita, N.; Iwata, T.; Uehara, H. Sonic hedgehog relates to colorectal carcinogenesis. J. Gastroenterol. 2009, 44, 1113–1117. [Google Scholar] [CrossRef] [PubMed]
  204. Zhang, X.; Zhang, S.S.; Wei, G.J.; Deng, Z.M.; Hu, Y. Dysregulation of hedgehog signaling pathway related components in the evolution of colonic carcinogenesis. Int. J. Clin. Exp. Med. 2015, 8, 21379–21385. [Google Scholar] [PubMed]
  205. Bian, Y.H.; Huang, S.H.; Yang, L.; Ma, X.L.; Xie, J.W.; Zhang, H.W. Sonic hedgehog-Gli1 pathway in colorectal adenocarcinomas. World J. Gastroenterol. 2007, 13, 1659–1665. [Google Scholar] [CrossRef] [PubMed]
  206. Oku, Y.; Shimoji, T.; Takifuji, K.; Hotta, T.; Yokoyama, S.; Matsuda, K.; Higashiguchi, T.; Tominaga, T.; Nasu, T.; Tamura, K.; et al. Identification of the molecular mechanisms for dedifferentiation at the invasion front of colorectal cancer by a gene expression analysis. Clin. Cancer Res. 2008, 14, 7215–7222. [Google Scholar] [CrossRef] [PubMed]
  207. Mazumdar, T.; DeVecchio, J.; Agyeman, A.; Shi, T.; Houghton, J.A. The GLI genes as the molecular switch in disrupting Hedgehog signaling in colon cancer. Oncotarget 2011, 2, 638–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. Mazumdar, T.; Devecchio, J.; Agyeman, A.; Shi, T.; Houghton, J.A. Blocking Hedgehog survival signaling at the level of the GLI genes induces DNA damage and extensive cell death in human colon carcinoma cells. Cancer Res. 2011, 71, 5904–5914. [Google Scholar] [CrossRef] [PubMed]
  209. Wu, J.Y.; Xu, X.F.; Xu, L.; Niu, P.Q.; Wang, F.; Hu, G.Y.; Wang, X.P.; Guo, C.Y. Cyclopamine blocked the growth of colorectal cancer SW116 cells by modulating some target genes of Gli1 in vitro. Hepatogastroenterology 2011, 58, 1511–1518. [Google Scholar] [CrossRef] [PubMed]
  210. Qualtrough, D.; Rees, P.; Speight, B.; Williams, A.C.; Paraskeva, C. The Hedgehog Inhibitor Cyclopamine Reduces beta-Catenin-Tcf Transcriptional Activity, Induces E-Cadherin Expression, and Reduces Invasion in Colorectal Cancer Cells. Cancers (Basel) 2015, 7, 1885–1899. [Google Scholar] [CrossRef] [PubMed]
  211. Chen, Q.; Zhang, H.; Wu, M.; Wang, Q.; Luo, L.; Ma, H.; Zhang, X.; He, S. Discovery of a potent hedgehog pathway inhibitor capable of activating caspase8-dependent apoptosis. J. Pharmacol. Sci. 2018, 137, 256–264. [Google Scholar] [CrossRef] [PubMed]
  212. Song, L.; Li, Z.Y.; Liu, W.P.; Zhao, M.R. Crosstalk between Wnt/beta-catenin and Hedgehog/Gli signaling pathways in colon cancer and implications for therapy. Cancer Biol. Ther. 2015, 16, 1–7. [Google Scholar] [CrossRef] [PubMed]
  213. Cai, X.; Yu, K.; Zhang, L.; Li, Y.; Li, Q.; Yang, Z.; Shen, T.; Duan, L.; Xiong, W.; Wang, W. Synergistic inhibition of colon carcinoma cell growth by Hedgehog-Gli1 inhibitor arsenic trioxide and phosphoinositide 3-kinase inhibitor LY294002. OncoTargets Ther. 2015, 8, 877–883. [Google Scholar] [CrossRef] [PubMed]
  214. Batsaikhan, B.E.; Yoshikawa, K.; Kurita, N.; Iwata, T.; Takasu, C.; Kashihara, H.; Shimada, M. Cyclopamine decreased the expression of Sonic Hedgehog and its downstream genes in colon cancer stem cells. Anticancer Res. 2014, 34, 6339–6344. [Google Scholar] [PubMed]
  215. Didiasova, M.; Schaefer, L.; Wygrecka, M. Targeting GLI Transcription Factors in Cancer. Molecules 2018, 23, 1003. [Google Scholar] [CrossRef] [PubMed]
  216. Memmi, E.M.; Sanarico, A.G.; Giacobbe, A.; Peschiaroli, A.; Frezza, V.; Cicalese, A.; Pisati, F.; Tosoni, D.; Zhou, H.; Tonon, G.; et al. p63 Sustains self-renewal of mammary cancer stem cells through regulation of Sonic Hedgehog signaling. Proc. Natl. Acad. Sci. USA 2015, 112, 3499–3504. [Google Scholar] [CrossRef] [PubMed]
  217. Takebe, N.; Hunsberger, S.; Yang, S.X. Expression of Gli1 in the hedgehog signaling pathway and breast cancer recurrence. Chin. J. Cancer Res. 2012, 24, 257–258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Tao, Y.; Mao, J.; Zhang, Q.; Li, L. Overexpression of Hedgehog signaling molecules and its involvement in triple-negative breast cancer. Oncol. Lett. 2011, 2, 995–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  219. Harris, L.G.; Pannell, L.K.; Singh, S.; Samant, R.S.; Shevde, L.A. Increased vascularity and spontaneous metastasis of breast cancer by hedgehog signaling mediated upregulation of cyr61. Oncogene 2012, 31, 3370–3380. [Google Scholar] [CrossRef] [PubMed]
  220. Kurebayashi, J.; Kanomata, N.; Koike, Y.; Ohta, Y.; Saitoh, W.; Kishino, E. Comprehensive immunohistochemical analyses on expression levels of hedgehog signaling molecules in breast cancers. Breast Cancer 2018, 25, 759–767. [Google Scholar] [CrossRef] [PubMed]
  221. Ruiz-Borrego, M.; Jimenez, B.; Antolin, S.; Garcia-Saenz, J.A.; Corral, J.; Jerez, Y.; Trigo, J.; Urruticoechea, A.; Colom, H.; Gonzalo, N.; et al. A phase Ib study of sonidegib (LDE225), an oral small molecule inhibitor of smoothened or Hedgehog pathway, in combination with docetaxel in triple negative advanced breast cancer patients: GEICAM/2012-12 (EDALINE) study. Investig. New Drugs 2018. [Google Scholar] [CrossRef]
  222. Wolf, I.; Bose, S.; Desmond, J.C.; Lin, B.T.; Williamson, E.A.; Karlan, B.Y.; Koeffler, H.P. Unmasking of epigenetically silenced genes reveals DNA promoter methylation and reduced expression of PTCH in breast cancer. Breast Cancer Res. Treat. 2007, 105, 139–155. [Google Scholar] [CrossRef] [PubMed]
  223. Polydorou, C.; Mpekris, F.; Papageorgis, P.; Voutouri, C.; Stylianopoulos, T. Pirfenidone normalizes the tumor microenvironment to improve chemotherapy. Oncotarget 2017, 8, 24506–24517. [Google Scholar] [CrossRef] [PubMed]
  224. Salazar, L.G.; Lu, H.; Reichow, J.L.; Childs, J.S.; Coveler, A.L.; Higgins, D.M.; Waisman, J.; Allison, K.H.; Dang, Y.; Disis, M.L. Topical Imiquimod Plus Nab-paclitaxel for Breast Cancer Cutaneous Metastases: A Phase 2 Clinical Trial. JAMA Oncol. 2017, 3, 969–973. [Google Scholar] [CrossRef] [PubMed]
  225. Lauth, M.; Bergstrom, A.; Shimokawa, T.; Toftgard, R. Inhibition of GLI-mediated transcription and tumor cell growth by small-molecule antagonists. Proc. Natl. Acad. Sci. USA 2007, 104, 8455–8460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Kasper, M.; Jaks, V.; Fiaschi, M.; Toftgard, R. Hedgehog signalling in breast cancer. Carcinogenesis 2009, 30, 903–911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Colavito, S.A.; Zou, M.R.; Yan, Q.; Nguyen, D.X.; Stern, D.F. Significance of glioma-associated oncogene homolog 1 (GLI1)expression in claudin-low breast cancer and crosstalk with the nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) pathway. Breast Cancer Res. 2014, 16, 444. [Google Scholar] [CrossRef] [PubMed]
  228. Koury, J.; Zhong, L.; Hao, J. Targeting Signaling Pathways in Cancer Stem Cells for Cancer Treatment. Stem Cells Int. 2017, 2017. [Google Scholar] [CrossRef] [PubMed]
  229. Ercan, G.; Karlitepe, A.; Ozpolat, B. Pancreatic cancer stem cells and therapeutic approaches. Anticancer Res. 2017, 37, 2761–2775. [Google Scholar] [PubMed]
  230. Lau, J.; Kawahira, H.; Hebrok, M. Hedgehog signaling in pancreas development and disease. Cell. Mol. Life Sci. 2006, 63, 642–652. [Google Scholar] [CrossRef] [PubMed]
  231. Martin, S.; Sato, N.; Dhara, S.; Chang, R.; Hustinx, S.R.; Abe, T.; Maitra, A.; Goggins, M. Aberrant methylation of the Human Hedgehog interacting protein (HHIP) gene in pancreatic neoplasms. Cancer Biol. Ther. 2005, 4, 728–733. [Google Scholar] [CrossRef] [PubMed]
  232. Feldmann, G.; Dhara, S.; Fendrich, V.; Bedja, D.; Beaty, R.; Mullendore, M.; Karikari, C.; Alvarez, H.; Iacobuzio-Donahue, C.; Jimeno, A. Blockade of hedgehog signaling inhibits pancreatic cancer invasion and metastases: A new paradigm for combination therapy in solid cancers. Cancer Res. 2007, 67, 2187–2196. [Google Scholar] [CrossRef] [PubMed]
  233. Feldmann, G.; Habbe, N.; Dhara, S.; Bisht, S.; Alvarez, H.; Fendrich, V.; Beaty, R.; Mullendore, M.; Karikari, C.; Oullette, M.M. Hedgehog inhibition prolongs survival in a genetically engineered mouse model of pancreatic cancer. Gut 2008, 57, 1420–1430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Ko, A.H.; LoConte, N.; Tempero, M.A.; Walker, E.J.; Kelley, R.K.; Lewis, S.; Chang, W.-C.; Kantoff, E.; Vannier, M.W.; Catenacci, D.V. A phase I study of FOLFIRINOX plus IPI-926, a hedgehog pathway inhibitor, for advanced pancreatic adenocarcinoma. Pancreas 2016, 45, 370–375. [Google Scholar] [CrossRef] [PubMed]
  235. Catenacci, D.V.; Junttila, M.R.; Karrison, T.; Bahary, N.; Horiba, M.N.; Nattam, S.R.; Marsh, R.; Wallace, J.; Kozloff, M.; Rajdev, L. Randomized phase Ib/II study of gemcitabine plus placebo or vismodegib, a hedgehog pathway inhibitor, in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2015, 33, 4284–4292. [Google Scholar] [CrossRef] [PubMed]
  236. Kim, E.J.; Sahai, V.; Abel, E.V.; Griffith, K.A.; Greenson, J.K.; Takebe, N.; Khan, G.N.; Blau, J.L.; Balis, U.G.; Craig, R. Pilot clinical trial of hedgehog pathway inhibitor GDC-0449 (vismodegib) in combination with gemcitabine in patients with metastatic pancreatic adenocarcinoma. Clin. Cancer Res. 2014, 20, 5937–5945. [Google Scholar] [CrossRef] [PubMed]
  237. Pietsch, T.; Waha, A.; Koch, A.; Kraus, J.; Albrecht, S.; Tonn, J.; Sorensen, N.; Berthold, F.; Henk, B.; Schmandt, N.; et al. Medulloblastomas of the desmoplastic variant carry mutations of the human homologue of Drosophila patched. Cancer Res. 1997, 57, 2085–2088. [Google Scholar] [PubMed]
  238. Berman, D.M.; Karhadkar, S.S.; Hallahan, A.R.; Pritchard, J.I.; Eberhart, C.G.; Watkins, D.N.; Chen, J.K.; Cooper, M.K.; Taipale, J.; Olson, J.M.; et al. Medulloblastoma growth inhibition by hedgehog pathway blockade. Science 2002, 297, 1559–1561. [Google Scholar] [CrossRef] [PubMed]
  239. Kim, J.; Tang, J.Y.; Gong, R.; Kim, J.; Lee, J.J.; Clemons, K.V.; Chong, C.R.; Chang, K.S.; Fereshteh, M.; Gardner, D.; et al. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell. 2010, 17, 388–399. [Google Scholar] [CrossRef] [PubMed]
  240. 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] [PubMed]
  241. Hegde, G.V.; Peterson, K.J.; Emanuel, K.; Mittal, A.K.; Joshi, A.D.; Dickinson, J.D.; Kollessery, G.J.; Bociek, R.G.; Bierman, P.; Vose, J.M. Hedgehog-induced survival of B-cell chronic lymphocytic leukemia cells in a stromal cell microenvironment: A potential new therapeutic target. Mol. Cancer Res. 2008, 6, 1928–1936. [Google Scholar] [CrossRef] [PubMed]
  242. Long, B.; Zhu, H.; Zhu, C.; Liu, T.; Meng, W. Activation of the Hedgehog pathway in chronic myelogeneous leukemia patients. J. Exp. Clin. Cancer Res. 2011, 30, 8. [Google Scholar] [CrossRef] [PubMed]
  243. Phi, L.T.H.; Sari, I.N.; Yang, Y.G.; Lee, S.H.; Jun, N.; Kim, K.S.; Lee, Y.K.; Kwon, H.Y. Cancer Stem Cells (CSCs) in Drug Resistance and their Therapeutic Implications in Cancer Treatment. Stem Cells Int. 2018, 2018, 5416923. [Google Scholar] [CrossRef] [PubMed]
  244. Ochs, M.; Farrar, J.; Considine, M.; Ries, R.E.; Trevino, L.R.; Alonzo, T.A.; Guidry-Auvil, J.; Davidsen, T.M.; Gesuwan, P.; Muzny, D.M. Genome wide promoter methylation patterns predict AML subtype outcomes and identify novel pathways characterizing diagnostic and relapsed disease in children. Blood 2012, 120, 1287. [Google Scholar]
  245. Campbell, V.; Tholouli, E.; Quigley, M.T.; Keilty, J.; Kelly, P.; McGovern, K.; Read, M.; Kutok, J.L.; Byers, R. Evidence that activated hedgehog signaling predicts for poor clinical outcome in acute myeloid leukemia. Blood 2012, 120, 1441. [Google Scholar]
  246. Burns, M.A.; Liao, Z.W.; Yamagata, N.; Pouliot, G.P.; Stevenson, K.E.; Neuberg, D.S.; Thorner, A.R.; Ducar, M.; Silverman, E.A.; Hunger, S.P. Hedgehog pathway mutations drive oncogenic transformation in high-risk T-cell acute lymphoblastic leukemia. Leukemia 2018, 32, 2126–2137. [Google Scholar] [CrossRef] [PubMed]
  247. Dagklis, A.; Demeyer, S.; De Bie, J.; Radaelli, E.; Pauwels, D.; Degryse, S.; Gielen, O.; Vicente, C.; Vandepoel, R.; Geerdens, E.; et al. Hedgehog pathway activation in T-cell acute lymphoblastic leukemia predicts response to SMO and GLI1 inhibitors. Blood 2016, 128, 2642–2654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Dagklis, A.; Pauwels, D.; Lahortiga, I.; Geerdens, E.; Bittoun, E.; Cauwelier, B.; Tousseyn, T.; Uyttebroeck, A.; Maertens, J.; Verhoef, G.; et al. Hedgehog pathway mutations in T-cell acute lymphoblastic leukemia. Haematologica 2015, 100, e102–105. [Google Scholar] [CrossRef] [PubMed]
  249. Gao, J.; Graves, S.; Koch, U.; Liu, S.; Jankovic, V.; Buonamici, S.; El Andaloussi, A.; Nimer, S.D.; Kee, B.L.; Taichman, R.; et al. Hedgehog signaling is dispensable for adult hematopoietic stem cell function. Cell Stem Cell 2009, 4, 548–558. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the mammalian canonical Hh signaling pathway. (A) In the absence of Hh, PTCH1 localizes in the cilia and thus prevents the membrane localization and activation of SMO. SMO then internalizes in the membrane of intracellular endosomes and degrades. The full-length GLI (GLIFL) is modified by protein kinases protein kinase A (PKA), glycogen synthase kinase-3 (GSK3), casein kinase 1 (CK1), and the E3 ubiquitin ligase β-TrCP, which is then proteolytically cleaved into the transcriptional repressor form GLIR. The active form of GLI (GLIA) is suppressed by SUFU. After that, GLIR translocates to the nucleus and inhibits the expression of its target genes. (B) Binding of Hh to PTCH1 results in its internalization as well as the ciliary translocation and activation of SMO. GLIFL maintains its full length and bypasses the phosphorylation by PKA, GSK3, and CK1, which leads to the formation of activated GLI (GLIA). GLIA then translocates to the nucleus where it induces the expression of Hh target genes. Interestingly, Kif7 not only participates in the movement of GLIFL within the cilia but also inhibits Sufu which usually suppresses the formation of GLIA from GLIFL.
Figure 1. Schematic representation of the mammalian canonical Hh signaling pathway. (A) In the absence of Hh, PTCH1 localizes in the cilia and thus prevents the membrane localization and activation of SMO. SMO then internalizes in the membrane of intracellular endosomes and degrades. The full-length GLI (GLIFL) is modified by protein kinases protein kinase A (PKA), glycogen synthase kinase-3 (GSK3), casein kinase 1 (CK1), and the E3 ubiquitin ligase β-TrCP, which is then proteolytically cleaved into the transcriptional repressor form GLIR. The active form of GLI (GLIA) is suppressed by SUFU. After that, GLIR translocates to the nucleus and inhibits the expression of its target genes. (B) Binding of Hh to PTCH1 results in its internalization as well as the ciliary translocation and activation of SMO. GLIFL maintains its full length and bypasses the phosphorylation by PKA, GSK3, and CK1, which leads to the formation of activated GLI (GLIA). GLIA then translocates to the nucleus where it induces the expression of Hh target genes. Interestingly, Kif7 not only participates in the movement of GLIFL within the cilia but also inhibits Sufu which usually suppresses the formation of GLIA from GLIFL.
Cells 07 00208 g001aCells 07 00208 g001b
Figure 2. Schematic model of Hh activation and the therapeutic strategy for cancers. (A) In the presence of the Hh ligand, PTCH1 is internalized and SMO is activated, leading to the localization of GLI-1 in the nucleus. Gli-1 is activated and promotes the transcription of its target genes, which is important for the growth of cancer cells, promotion of epithelial–mesenchymal transition (EMT) mechanisms, drug resistance, and the maintenance of CSC population (upper). Conventional chemotherapy eliminates bulk cancer cells. However, residing CSCs escape from the therapy and trigger cancer relapse (bottom). (B) Inhibition by either Hh ligand, SMO, or GLI1 inhibitors led to the inactivation of Hh signaling. Gli-1 is suppressed by SUFU and fails to localize in the nucleus and activate Hh target genes (upper). Inhibition of Hh signaling together with conventional chemotherapeutics results in the death of cancer bulk cells and eradication of the cancer stem cell population which has escaped from conventional chemotherapeutics (bottom).
Figure 2. Schematic model of Hh activation and the therapeutic strategy for cancers. (A) In the presence of the Hh ligand, PTCH1 is internalized and SMO is activated, leading to the localization of GLI-1 in the nucleus. Gli-1 is activated and promotes the transcription of its target genes, which is important for the growth of cancer cells, promotion of epithelial–mesenchymal transition (EMT) mechanisms, drug resistance, and the maintenance of CSC population (upper). Conventional chemotherapy eliminates bulk cancer cells. However, residing CSCs escape from the therapy and trigger cancer relapse (bottom). (B) Inhibition by either Hh ligand, SMO, or GLI1 inhibitors led to the inactivation of Hh signaling. Gli-1 is suppressed by SUFU and fails to localize in the nucleus and activate Hh target genes (upper). Inhibition of Hh signaling together with conventional chemotherapeutics results in the death of cancer bulk cells and eradication of the cancer stem cell population which has escaped from conventional chemotherapeutics (bottom).
Cells 07 00208 g002
Table 1. Hedgehog signaling activation in cancers.
Table 1. Hedgehog signaling activation in cancers.
TypeCharacteristicCancer Type
Type ILigand independentBasal cell carcinoma
Medulloblastoma (MB)
Pediatric brain tumor & Rhabdomyosarcoma
Type IILigand-dependent autocrine/juxtacrine signalingColorectal
Prostate
Liver
Breast
Ovarian
Brain
Melanoma
Type IIILigand-dependent paracrine signalingPancreatic
Leukemia (reverse paracrine)
Table 2. Hedgehog signaling inhibition in cancer stem cells (CSCs).
Table 2. Hedgehog signaling inhibition in cancer stem cells (CSCs).
Tumor TypeCSC MarkerHh InhibitorsCombination TherapyReferences
Basal cell carcinoma (BCC)CD200+CD45Smo antagonistCD200+ neutralizing antibody[164]
Colon cancerCD133+PTCH1 inhibitor (RU-SKI 43), Cyclopamine [97,128]
Breast cancerLinCD44+CD24Cyclopamine [144,165]
Pancreatic cancerCD44+CD24+ESA+Sulforaphane, Baicalein, Sangunarine, GANT61Gemtacibine and cyclopamine, cyclopamine derivates (CyT)
+2gy irradiation
[166,167,168,169,170,171,172]
MedulloblastomaCD15+, Sox2+KAAD-cyclopamine [173,174,175]
Chronic myelogenous leukemia (CML)LinSca1+cKit+CD34+Sonidegib, cyclopamine, PF-04449913Sonidegib and nilotinib, cyclopamine and nilotinib, PF-04449913 and dasatinib[176,177,178,179]
Acute myelogenous leukemia (AML) PF-04449913, cyclopamine, PF-913Cyclopamine and Ara-C[180,181,182]
Acute lymphoblastic leukemia (ALL) Cyclopamine, IPI-926, KAAD-cyclopamine, SANT1 [183,184,185]
Table 3. Hedgehog signaling inhibitors in clinical trials. Data from www.clinicaltrials.gov.
Table 3. Hedgehog signaling inhibitors in clinical trials. Data from www.clinicaltrials.gov.
CompoundTargetConditionsPhaseNCT NumberCombination DrugLocationsStatus
GDC-0449 (Vismodegib/Erivedge)SMOBasal cell carcinomaEarly Phase 1NCT01631331-USCompleted
Phase 1NCT02639117-USCompleted
Phase 2NCT01543581--Completed
NCT00833417-USCompleted
NCT01201915-USCompleted
NCT03035188-GermanyRecruiting
NCT02667574-FranceActive
NCT01815840-USCompleted
NCT01367665-Australia, ArgentinaCompleted
NCT01898598-USTerminated
NCT02067104-USActive
NCT01700049-USActive
NCT01835626-USRecruiting
NCT01556009-USCompleted
NCT02956889-ItalyRecruiting
NCT00959647FOLFOX, FOLFIRI, BevacizumabUSCompleted
Phase 4NCT02436408-USRecruiting
Phase 1, 2NCT02690948Pembrolizumab USActive
-NCT01160250-USApproved
NCT02371967-SwedenActive
NCT02781389-GermanyActive
NCT02674009-GermanyActive
NCT02438644--Completed
Colon cancerPhase 2NCT00636610Bevacizumab, Modified FOLFOX, FOLFIRI-Completed
NCT00959647FOLFOX, FOLFIRI, bevacizumabUSCompleted
Breast cancerPhase 2NCT02694224Paclitaxel, Epirubicin, CyclophosphamideSpainRecruiting
Pancreatic cancerEarly Phase 1NCT01713218gemcitabineBelgiumUnknown
Phase 1NCT00878163erlotinib hydrochloride, gemcitabineUSActive
NCT01537107sirolimusUSSuspended
Phase 2NCT01088815Gemcitabine, nab-PaclitaxelUSUnknown
NCT01195415Gemcitabine HydrochlorideUSCompleted
Phase 1, 2NCT01064622gemcitabine hydrochlorideUSCompleted
Medullo blastomaPhase 1NCT00822458-USCompleted
Phase 2NCT00939484-USCompleted
NCT01239316-USCompleted
Phase 1, 2NCT01601184TemozolomideFranceRecruiting
LeukemiaPhase 2NCT01880437cytarabineUSTerminated
NCT01944943-FranceTerminated
NCT02073838Ribavirin, DecitabineUSCompleted
LDE225 (Erismodegib/Sonidegib/Odomzo)SMOBasal cell carcinomaPhase 1NCT01208831-Hong Kong, Japan, TaiwanCompleted
NCT00880308-US, SpainCompleted
Phase 2NCT01327053-USActive
NCT02303041BuparlisibUSCompleted
NCT01033019-Austria, AustraliaTerminated
NCT03534947ImiquimodAustraliaNot yet
NCT00961896-Austria, SwitzerlandCompleted
NCT01350115-Austria, Belgium, CanadaCompleted
Phase 2, 3NCT03070691-Belgium, CanadaWithdrawn
-NCT01529450-USCompleted
Breast cancerPhase 1NCT02027376DocetaxelSpainUnknown
Phase 2NCT01757327--Withdrawn
Pancreatic cancerEarly Phase 1NCT01694589-USWithdrawn
Phase 1NCT01487785gemcitabineUSCompleted
NCT01485744Fluorouracil, Leucovorin, Oxaliplatin, IrinotecanUSActive
Phase 1, 2NCT02358161nab paclitaxelNetherlandUnknown
-NCT01911416-USWithdrawn
Medullo blastomaPhase 1NCT01208831-Hong Kong, Japan, TaiwanCompleted
NCT00880308-Spain, USCompleted
Phase 2NCT01708174-USCompleted
Phase 1, 2NCT01125800-USCompleted
LeukemiaPhase 1NCT01456676nilotinibCanada, France, GermanyCompleted
NCT02129101Azacitidine, DecitabineUSActive
Phase 2NCT01826214-Australia, USCompleted
IPI-926 (Saridegib)SMOBasal cell carcinoma-NCT01609179-USCompleted
Pancreatic cancerPhase 1NCT013835385-fluorouracil, Leucovorin, Irinotecan, OxaliplatinUSCompleted
Phase 1, 2NCT01130142gemcitabineUSCompleted
PF-04449913 (Glasdegib)SMOLeukemiaPhase 1NCT01546038ARA-C, Decitabine, Daunorubicin, CytarabineUSActive
NCT02038777ARA-C, Daunorubicin, Cytarabine, AzacitidineJapanRecruiting
NCT02367456AzacitidineUSRecruiting
NCT00953758-USCompleted
Phase 2NCT01841333-USRecruiting
NCT01842646-USActive
NCT03390296Avelumab, azacitidine, utomilumabUSRecruiting
Phase 3NCT03416179-USRecruiting
BMS-833923SMOLeukemiaPhase 2NCT01357655DasatinibUSTerminated
Phase 1, 2NCT01218477DasatinibUSCompleted

Share and Cite

MDPI and ACS Style

Sari, I.N.; Phi, L.T.H.; Jun, N.; Wijaya, Y.T.; Lee, S.; Kwon, H.Y. Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells. Cells 2018, 7, 208. https://doi.org/10.3390/cells7110208

AMA Style

Sari IN, Phi LTH, Jun N, Wijaya YT, Lee S, Kwon HY. Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells. Cells. 2018; 7(11):208. https://doi.org/10.3390/cells7110208

Chicago/Turabian Style

Sari, Ita Novita, Lan Thi Hanh Phi, Nayoung Jun, Yoseph Toni Wijaya, Sanghyun Lee, and Hyog Young Kwon. 2018. "Hedgehog Signaling in Cancer: A Prospective Therapeutic Target for Eradicating Cancer Stem Cells" Cells 7, no. 11: 208. https://doi.org/10.3390/cells7110208

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